HAEMOPOIETIC PROGENITOR CELLS IN HUMAN PERIPHERAL

F.E. ZWAAN HAEMOPOIETIC PROGENITOR CELLS IN HUMAN PERIPHERAL BLOOD

PROEFSCHRIFT

TER VERKRIJGING VAN DE GRAAD VAN DOCTOR IN DE GENEESKUNDE AAN DE RIJKSUNIVERSITEIT TE LEIDEN, OP GEZAG VAN DE RECTOR MAGNIFICUS DR. A.A.H. KASSENAAR, HOOGLERAAR IN DE FACULTEIT DER GENEESKUNDE, VOLGENS BESLUIT VAN HET COLLEGE VAN DEKANEN TE VERDEDIGEN OP WOENSDAG 5 NOVEMBER 1980 TEKLOKKE 16.15 UUR

DOOR

FERDINAND EMILE ZWAAN i GEBOREN TE 's-GRAVENHAGE IN 1944

1980 DRUKKERIJ J.H. PASMANS B.V., 's-GRAVENHAGE Promotores: Prof. Dr. J.J. Veltkamp en Prof. Dr. Th.G. van Rijssel

Referenten: Prof. Dr. J.J. van Rood en Dr. G. van den Engh i Though this be madness, there is method in it.

W. Shakespeare The studies reported in this thesis were performed in the Laboratory for Experimental Haematology (Department of Haematology; Head: Prof. Dr. J.J. Veltkamp), Leiden University Medical Centre, with the assistance of Miss H.M. Goselink, Miss W.F.J. Veenhof, Mrs. M. Puliga-Wildschut and Mr. C. Martijn. The processing of diffusion chambers for histological examination was taken care of by Dr. J. te Velde, Miss J.K. Kleiverda, Mrs. C.F. Leenheers- Binnendijk and Miss R. Lobatto at the Department of Pathology (Heads: Prof. Dr. Th.G. van Rijssel and Prof. Dr. A. Schaberg). The photography of histologic preparations was performed by Mr. R.M.L. Heruer and Mr. K.J. van der Ham. Animals and blood cells were irradiated at the Department of Radio- therapy (Head: Prof. Dr. P. Thomas), according to the instructions of Dr. J.H. Mellink. Miss T. Goedkoop took care of the delivery of leukocyte suspensions obtained from blood donors at the Bloodbank, while Drs. L.J.M. Sabbe performed the "in vitro" stimulation tests and MLC-studies at the Department of Immunohaematology (Head: Prof. Dr. J.J. van Rood). Peripheral blood cells from patients with myeloproliferative disorders were obtained with the help of Miss A. van Nieuwennuyzen. The cytochemical stains were performed and interpreted by Miss L. Huibregtsen, Miss L. Colla and Drs. G.J. den Ottolander (chief Central Clinical Haematology Laboratory) at the department of Haematolo- morphology (Head: Prof. Dr. P. Lopes Cardozo). Statistical calculations were carried out by Dr. J. Hermans at the Department of Medical Statistics (Head: Prof. H. de Jonge). Figures were drawn by Mr. J.J. Magdelijns and Mr. J. van Leeuwen at the Audiovisual Service, while the three-dimensional constructions of haemopoietic colonies were drawn by Mr. J. Tinkelenberg at the Depart- ment of Anatomy (Head: Prof. Dr. J.M.F. Landsmeer). The manuscript was typed by Miss H.M. Plugge and read by Miss R.H.F. de Ru for correction of linguistic errors. CONTENTS

Chapter 1 GENERAL INTRODUCTION 1.1 Review of the literature 11 1.1.1 Haemopoietic progenitor cells in peripheral blood; animal studies 11 1.1.1.1 CFU-s: definition and several characteristics 11 1.1.1.2 CFU-c: definition and several characteristics 14 1.1.1.3 Dog studies: leucapheresis and transplantation of peripheral blood progenitor cells 15 1.1.1.4 Methods for mobilization of progenitor cells to the peripheral blood in animals 16 1.1.1.5 Cryopreservation and density gradient separation of peripheral blood progenitor cells before autologous transplantation 18 1.1.2 Haemopoietic progenitor cells in peripheral blood; human studies 21 1.1.2.1 CFU-c: definition and several characteristics 21 1.1.2.2 Leucapheresis in humans; quantitative data on CFU-c yields 23 1.1.3 Collection and purification of progenitor cells from peripheral blood — separation techniques 25 1.2 Conclusions and discussion 30 1.3 Purpose and design of the investigation 31 1.4 References 32

Chapter 2 PURIFICATION OF CFU-c FROM HUMAN PERIPHERAL BLOOD BY A TWO-STEP DENSITY SEPARATION TECH- NIQUE 2.1 Introduction 40 2.2 Material and methods 45 2.2.1 Isolation of leucocytes and mononuclear cells by centrifugation techniques; general principles 45 2.2.2 Isolation of leucocytes by methylcellulose sedimenta- tion 46 2.2.3 Preparation of monocyte-free lymphocyte suspensions 46 2.2.4 Identification of B- with direct immuno- fluorescence on surface immunoglobulins (SIg) 46 2.2.5 Identification of B-lymphocytes using goat anti- human Fab (GahuFab) coated sheep red blood cells (SRBC); B-lymphocyte rosette (= B-RFC). 47 2.2.6 Identification of mouse-erythrocyte (MRBC) rosette forming B-cells (= M-RFC) 47 2.2.7 Identification of IgG-Fc receptor bearing cells with EA-rosette formation, using human red blood cells (HuRBC) coated with anti D-IgG (= EA, -RFC) 48 2.2.8 Identification of IgG-Fc receptor bearing cells with EA-rosette formation, using SRBC coated with rabbit- anti-sheep IgG (= EA2 -RFC) 48 2.2.9 Identification of IgG-Fc receptor bearing cells with EA-rosette formation, using ox red blood cells (ORBC) coated with rabbit-anti-ox IgG (EA3-RFC) 49 2.2.10 Identification of T-cells with E-rosette formation, using SRBC (= E-RFC) 50 2.2.11 Separation of rosette forming cells from non-rosette forming cells 50 2.2.12 Technique for simultaneous rosette formation 51 2.2.13 Elimination of B-lymphocytes and monocytes by nylon-wool filtration 51 2.2.14 Preparation of and cell processing with continuous Ficoll-Isopaque density gradients 52 2.2.15 Cell staining and counting 53 2.2.16 CFU-c culture technique 54 2.2.17 Statistical methods 55 2.3 Results 55 2.3.1 Linear density distribution of monocytes, T-cells, B-cells and CFU-c 55 2.3.2 Linear density distribution of T-cells, B-cells, and CFU-c after initial monocyte elimination 57 2.3.3 Comparison of two different densities (ds. 1.072 g/ml; ds. 1.077 g/ml) during the second separation step 59 2.3.4 Results of a two-step density gradient separation technique; elimination of monocytes and T-cells and subsequent CFU-c purification 62 2.3.5 Results of a three-step separation technique, including density gradients and nylon-wool filtration; removal of monocytes, T-cells, B-cells and subsequent CFU-c purification 64 2.3.6 The effect of simultaneous E-RFC, EA,-RFC, and M-RFC removal during the second separation step on CFU-c purification 69 2.3.7 Modification of a B-cell rosette (B-RFC) technique, using goat anti-human Fab (GahuFab) coated SRBC (= B-RFC), as indicator 72 2.3.8 Testing of the B-RFC-technique in a three-step sepa- ration experiment, in combination with monocyte, E-RFC and EAy -RFC-elimination 73 2.3.9 Testing of the B-RFC -technique in a two-step sepa- ration experiment; effect of B-RFC removal on monocytes 77 2.3.10 The effect of a simultaneous "triple-rosette" elimina- tion (E-+EA1- + B-RFC) during the second separation step on CFU-c purification 77 2.3.11 Another approach for monocyte removal and sub- sequent testing in a two- and one-step separation technique; effect on CFU-c purification 78 2.3.12 The effect of a two-step density gradient technique using monocyte removal during the first step and si- multaneous E- and B-RFC-elimination during the second step 83 2.3.13 The effect of a two-step density gradient technique using monocyte removal during the first step and si- multaneous E-, B-, and EA2-RFC-elimination during the second step 85 2.4 Conclusions and discussion 87 2.5 References 94

Chapter 3 PURIFIED CFU-c SUSPENSIONS OBTAINED FROM HUMAN PERIPHERAL BLOOD; SEVERAL "IN VITRO" CHARACTERISTICS 3.1 Introduction 100 3.2 Material and methods 101 8

3.2.1 Cytochemical staining techniques of cytocentrifuge smears 101 3.2.2 Lymphocyte transformation tests and mixed lympho- cyte culture 101 3.2.3 Determination of the turnover state of CFU-c with the3 H-thymidine suicide technique 102 3.2.4 Determination of CFU-c radiosensitivity (Do-value) 103 3.2.5 Cell volume analysis 104 3.2.6 Statistical methods 105 3.3 Results 105 3.3.1 Cytological aspects of "CFU-c purified" null cell suspensions 105 3.3.2 Null cells: their capacity to respond to PHA, and MLC-reactivity 105 3.3.3 Determination of the turnover rate of CFU-c with the 3 H-thymidine suicide technique 111 3.3.4 Determination of CFU-c radiosensitivity (Do-value) 112 3.3.5 Volume profile of MNC-subpopulations 120 3.4 Conclusions and discussion 123 3.5 References 130

Chapter 4 QUANTIFICATION AND QUALIFICATION OF COLONY FORMATION BY HUMAN PERIPHERAL BLOOD HAEMO- POIETIC PROGENITOR CELLS IN PLASMA CLOT DIF- FUSION CHAMBERS 4.1 Introduction 135 4.2 Material and methods 137 4.2.1 Mice 137 4.2.2 Irradiation of mice 137 4.2.3 Phenylhydrazine pretreatment of mice 137 4.2.4 Diffusion chamber technique 138 4.2.5 Cell staining and counting 139 4.2.6 Plasma clot diffusion chamber technique 139 4.2.7 Methyl-methacrylate embedding technique and "histo- logical" processing of plasma clot diffusion chambers 139 4.2.8 Human peripheral blood mononuclear cell suspen- sions used in diffusion chamber cultures 140 4.2.9 Statistical methods 142 4.3 Results 143 4.3.1 Effect of irradiation and/or phenylhydrazine pretreat- ment on the survival ana peripheral blood counts of mice 143 4.3.2 Proliferation and differentiation of human peripheral mononuclear cells in conventional diffusion chambers. Cytological aspects and growth profile 146 4.3.3 Effect of pre-culturing of haemopoietic progenitor cells in diffusion chambers on CFU-c forming capacity in vitro 147 4.3.4 The proliferation and differentiation of haemopoietic progenitor cells in plasma clot diffusion chambers; "histological" aspects 149 4.3.5 Quantitative aspects of cell aggregate (CA) formation in plasma clot diffusion chambers 154 4.3.6 Three-dimensional construction of whole colonies in plasma clot diffusion chambers; emphasis on the structure of mixed colonies 158 4.3.7 Cell aggregate (CA) formation in plasma clot diffusion chambers by human haemopoietic progenitor cells in irradiated and phenyihydrazine pretreated mice 162 4.4 Conclusions and discussion 163 4.5 References 171

Chapter 5 HAEMOPOIETIC PROGENITOR CELLS IN HUMAN PERI- PHERAL BLOOD IN MYELOPROLIFERATIVE DIS- ORDERS 5.1 Introduction 175 5.2 Material and methods 176 5.3 Results 177 5.4 Conclusions and discussion 184 5.5 References 187

SUMMARY 192

SAMENVATTING 197

ABBREVIATIONS AND DEFINITIONS 202

CURRICULUM VITAE 206 11

Chapter 1

GENERAL INTRODUCTION

1.1 REVIEW OF THE LITERATURE

1.1.1 Haemopoietic progenitor cells in peripheral blood; animal studies 1.1.1.1 CFU-s: definition and several characteristics Most of our current knowledge about haemopoietic progenitor cells is derived from animal studies. In 1961 Till and McCulloch (135)* described the colony assay, which demonstrated the existence of a cell able to form erythroid, granulocytic, megakaryocytic, or mixed colonies in the spleen of lethally irradiated mice (colony-forming unit spleen = CFU-s). A linear relationship was demonstrated between the number of bone marrow cells injected and the number of spleen colonies counted after 8-10 days (Fig. 1). The dependence of colony formation on cell proliferation was shown by constructing radiation survival curves for suspensions irradiated both in vivo and in vitro (73); these were found to have the form characteris- tic of the radiation survival curve for mammalian cells. Becker et al. (47) showed clonality using chromosome markers; sub- lethal irradiation induced chromosomal lesions at random and the probability that two cells should bear the same defect after irradiation is very small. Sublethally irradiated mice were used as bone marrow donors. In each splenic colony where a chromosomal abnormality was found, it was unique and present in 100% of the cells in metaphasis. The big size of the splenic colonies on day 10 after the graft is an evident manifestation of the proliferative potential of this precursor cell (4, 135). The splenic colonies principally consist of differentiated blood cells, the presence of which illustrates the differentiation ability of the CFU-s (40, 74, 149). Furthermore, if cells from a splenic colony are re- transplanted into another irradiated recipient, new colonies will be ob- served (75, 127). This demonstrates the proliferative, differential, and * Numbers in brackets refer to the references listed at the end of each chapter. 12

800-1000 rad x-rays «/) 5x104 bone marrow cells H

8-10 days

spleen with nodules

Fig. 1: Technique for production of haemopoietic clonal nodules (CFU-s) on the spleen of irradiated mice by injection of syngeneic bone marrow cells. self renewing capacity of stem cells in splenic colonies. However, lymphoid splenic colonies were never observed (82). In spite of this, there is cyto- logical and chromosomal evidence that CFU-s and antibody-producing lymph node cells (137) or T-lymphocytes (93) may belong to the same clone. The existence of a common ancestor for CFU-s and lymphoid pro- genitors more primitive than either is also plausible (1). These pluripotent stem cells (HSC) are mainly in a quiescent state, as demonstrated by Fliedner et al. (35). Thus, in normal adult mice the CFU-s turnover rate is slow as shown by a 3H-thymidine "suicide" value of less than 10% (5). However, during recovery from irradiation (62) and cytotoxic drug damage (142) the 3H-thymidine suicide value of CFU-s can rise to 50%. It is currently assumed that in the normal steady state the majority of CFU-s are in a non-proliferative (Go) state, from which they can be triggered either into proliferation cycle, leading to cell division, or into differentiation. In mice the highest number of CFU-s is observed in the bone marrow (135), but a significant number of CFU-s are also found in the spleen, liver (51), and among peritoneal exudate cells (23). CFU-s mobility and migratory capacity has also been well documented with the endogenous spleen colony assay. In this setting, CFU-s from a bone marrow area shielded with lead migrate into the spleen of irradiated animals (48, 52, 114). Eventually, using chromosome markers, direct demonstration of stem cell migration from non-irradiated to irradiated sites was given by Maloney and Patt (69). 13

From early experiments by Brecher and Cronkite (11) on radiopro- tection in parabiotic animals it has become clear that HSC circulate in the blood stream. More direct evidence was provided by transplantation experiments. Goodman et al. (44) demonstrated, that, if peripheral leucocytes from mice were transplanted into lethally irradiated recipients, they prolifer- ated and gave rise to , and erythroblasts of donor origin. These observations were later extended to show that such CFU-s exists in the peripheral blood of rodents in a concentration of approximately one-hundredth of that in bone marrow (10, 68, 80, 138). Similarly, haemopoietic progenitor cells are circulating in the peripheral blood of dogs (14, 17,20,25,26). Gidali et al. (42) studied some properties of CFU-s circulating in the peripheral blood of mice, which could be compared with already known data from bone marrow CFU-s. Peripheral blood CFU-s showed a lower radio-sensitivity (Do = 140 Rad) than bone marrow CFU-s (Do = 100 Rad), a higher seeding effi- ciency (f-factor: 20.9%, f-factor for bone marrow CFU-s: 12.9%), and a higher sensitivity to 3H-thymidine suicide in vitro (killing effect 32% versus 2.3% for bone marrow CFU-s). The doubling time was the same for peripheral blood and bone marrow CFU-s (21 h), but the logarithmic growth phase of the peripheral blood CFU-s population started approxi- mately 36 h later than for bone marrow derived CFU-s. All these findings support the idea that CFU-s in the peripheral blood represent a subpopulation of the bone marrow CFU-s (121). Heterogen- eity of bone marrow CFU-s is demonstrated by different self renewal capacities and turnover rates found in CFU-s fractions of different den- sities after gradient separation (50, 65, 147). The morphological identification of these stem cells, however, proved to be difficult (86). Experimental work on mice and monkeys suggest that these cells may be small mononuclear cells (MNC) (30). Support in favour of this view comes from the observation of Fliedner, Haas et al. (36). In their experiments continuous 3 H-thymidine labelling in rats was started shortly after conception and discontinued (only several weeks) after birth. It was observed that the label was rapidly diluted in all cells except epithelial, reticular and small MNC that resembled lymphocytes. A haemopoietic challenge given at that time led to a decrease in the labelling of the lymphocytes but not of the epithelial or reticular cells. It was shown that these marrow "small mononuclear-lymphocyte-like" cells can be concentrated by centrifugation on a discontinuous albumin 14

gradient and that a correlation exists between cytokinetically resting small MNC and the regenerative process in lethally X-irradiated recipients (47). It was postulated that such cells entered the peripheral blood via lymphatic vessels, but attempts to demonstrate HSC in the thoracic ducts of dogs have been unsuccessful (128).

1.1.1.2 CFU-c: definition and several characteristics The study of haematopoiesis progressed tremendously with the finding that murine haemopoietic cells can form granulopoietic and monocytic colonies (colony forming unit culture = CFU-c) in cultures of semi- solid medium supplemented with suitable sources of colony stimulating factor (CSF)( 10, 102,103). Further research has indicated that haemopoietic colonies in vitro are clones derived from single colony forming cells (78, 96, 102) and that there is a linear relationship between the number of cells cultured and the number of developing colonies (78). During the past decade, culture techniques have been extensively de- veloped, so that various haemopoietic cell lines can now be grown in semi-solid media (55,81). The cell which forms a colony lies intermediate in differentiation between the multipotential haemopoietic stem cell and the earliest morphologically recognizable cells. When bone marrow cells are grown in agar in the presence of CSF, either pure granolocytic or pure monocy te-macrophage colonies or mixtures of these cells in colonies are developed. Although restricted to differentiate in the -macrophage lineage, the CFU-c can replicate its own population. This was demon- strated by transferring single cells from agar colonies to new petri dishes, where eventually mixed colonies developed from single cells (79). However, CFU-s and CFU-c are separate classes of progenitor cells. This was demonstrated by the fact that bone marrow CFU-s and CFU-c can be physically separated (50, 148), and that neutropenia causes a significant increase in the number of CFU-c without affecting the num- ber of CFU-s (113). Furthermore, most of the CFU-s in normal mouse bone marrow are not in cell cycle, while 30-50% of CFU-c are in active cell cycle as shown by thymidine suicide in vivo (62) and in vitro (54). Besides that, CFU-s express a membrane antigen which is not present on CFU-c (140). In spite of these differences, there is good evidence as shown by trans- plantation and by cytogenetic studies, that the committed CFU-c is 15

derived from the pluripotent CFU-s (150).* The mouse CFU-c is a highly heterogeneous cell population showing striking differences in their responsiveness to CSF and their capacity to generate differentiated colonies (141, 146). This degree of heterogeneity most likely reflects a discontinuous spectrum of differentiation. The CFU-c appear to have little or no capacity for self renewal but evidence on this point is in- complete (81).

1.1.1.3 Dog studies: leucapheresis and transplantation of peripheral blood progenitor cells Data on the number of peripheral blood leucocytes necessary to recon- stitute the bone marrow oflethally irradiated animals were mainly derived from studies in dogs. Debelak-Fehir et al. (26) investigated the CFU-c numbers in peripheral blood buffy coat and in Ficoll-Isopaque isolated MNC from 12 normal dogs. They found 7 ± 2.3 and 13 ± 3.8 CFU-c respectively per 1 x 10s cells**. In comparison, bone marrow yields 39 ± 5.0 CFU-c per 1 x 10s cells. It was shown that the number of cells infused was critical: 10-20 x 109 autologous blood leucocytes were sufficient to reconstitute dogs weighing 7-15 kg, while there were no survivors among animals which received less than 9 x 109 cells (17). Later Storb et al. (128) observed repopulation of bone marrow after infusion of 21-74 x 109 stored autologous peripheral leucocytes. This corresponded approximately to a dose of 0.5-2.5 x 109 MNC/kg b.w. of the recipient dogs. These values are in agreement with those obtained by Ross et al. (116); in dogs subjected to leucapheresis on the NCI-IBM cell separator an average of 25 x 109 leucocytes could be collected during one 5-hour procedure and 33% of these cells were MNC. This number of fresh or cryopreserved cells was adequate to restore the aplastic marrow in beagles used as recipients after whole body irradiation (1,200 Rad). A surprising observation was the absence of a significant difference in the repopulating ability of equal numbers of MNC from autologous or allogeneic source. Further transfusion experiments in dogs have confirmed that HSC capable of repopulating the haemopoietic and/or lymphopoietic system, are contained in the peripheral blood (16, 25, 38, 53, 90). * Recently, a culture technique was described, in which mixed erythro-, leuco-, and megakaryo- cytic cells in single colonies can be detected in vitro (Exp. Hemat. 7, suppl. 6: 3, 1979). ** CFU-c values in this chapter expressed as mean ± s.d., unless stated otherwise. 16

In dogs numerical changes in CFU-c during leucapheresis have been monitored as a possible indicator of changes in HSC numbers in the peri- pheral blood. Rosset al. (116) showed that the CFU-c yield during a single procedure reached a peak in the second hour of the leucapheresis and then declined over the next three hours. If, however, leucapheresis was repeated two days later, the total CFU-c yield was actually increased. Further increases were observed during the third and fourth consecutive procedures. During this time the MNC-fraction as a whole remained con- stant and it seems probable that MNC and CFU-c numbers are regulated by different mechanisms.

1.1.1.4 Methods for mobilization of progenitor cells to the peripheral blood in animals Efforts to increase the number of circulating CFU-c have centred on the administration of lymphocyte mobilizing agents such as polyvinyl sul- phuric acid (PVSA) and dextran sulphate (DS) (Table 1.1).

Table l.L The effect of several agents on haemopoietic stem cell and progenitor cell mobilization into the peripheral blood

Agent Test animal CFU-s CFU-c

increment maximal increment maximal (x) effect (x) effect (time) (time)

Dextran sulphate Dog 7-10 3 hours Polyvinyl-sulphuric acid Dog 2 5 hours Pyran-copolymer Dog 5 day 2 Pyran-copolymer Mouse 20 day 5 4 day 5

The intravenous administration of PVSA increases the MNC number in dogs to 4-5 times the unstimulated value after 5 hours; CFU-c numbers increased by almost 100% during this procedure (116). The heparinoid polyamon DS may be still more effective: within 3 hours after admini- stration peripheral blood CFU-c numbers increased by a factor 7-10, while MNC increased only by a factor 2(117). For DS of 10,000 m.w., mobilization increases with higher doses up to 15 mg/kg b.w.; a higher dose (20 mg/kg b.w.) does not seem to enhance the effect any further. The molecular weight of DS preparations apparently does not effect CFU-c mobilization (117, 118). Repeated administration of DS (in a dose of 15 mg/kg b.w.) has occasionally been accompanied by bleeding 17

complications. It has also been observed to increase the degree of extra- medullary haemopoiesis in the spleen, especially megakaryopoiesis. A moderate increase in extramedullary haemopoiesis is also observed after leucapheresis procedures. Recently Zander et al. (151) studied the effect of pyran co-polymer, an immuno-adjuvant, for its effect on circulating stem cells in mice and dogs. Mice were treated with 25 mg/kg b.w. intravenously and CFU-s and CFU-c in the peripheral blood and bone marrow were followed for 11 days. No immediate increase was noted during the first 24 hours. Peri- pheral blood CFU-s increased from 16±4to314±91 on day 5, whereas the MNC count only doubled. Peripheral blood CFU-c increased from 45 ± 12.7 to 163 ± 64 on day 5. Bone marrow CFU-s showed a gradual increase from 10 ± 5 to 40 ±11 (CFU-s per 104 cells), with a maximum on day 7. Pyran (25 mg/kg b.w.) was also given to 6 healthy dogs. Maximal CFU-c levels were found, with a more than 5-fold increase on day 2. From the above-mentioned studies the suggestion is derived that DS and to a lesser extent PVSA may selectively release CFU-c from the mar- row storage pools within several hours, and that pyran seems to affect the mobilization and production of haemopoietic progenitor cells, which shows its maximal effect after several days. In mice substances such as endotoxin and certain proteases can transiently mobilize CFU-s (105, 143), but the number of CFU-s in the circulation can increase in a variety of circumstances, namely during recovery from whole body irradiation (104), from cytostatic agents, and following antigenic stimulation (2, 87). Formation of increased numbers of endogenous spleen colonies also results from bilateral adrenalectomy (57) and, inversely, injection of ACTH (6) or thymectomy (100) inhibits CFU-c migration from shielded areas of the bone marrow. Randomly collected data and systematical serial studies show that the concentration of CFU-c is in the range of 10 to 1,000 per ml blood as has been demonstrated for the mouse (80), and dog (14, 20, 26, 60). The considerable differences between the blood CFU-c levels of the various species and strains and even individuals may be realistic or depend on variations between the assay methods. However, a good correlation between blood and bone marrow CFU-c number has been obtained during haemopoietic recovery of dogs after chemotherapy (20). Further- more, the CFU-c levels in the blood of dogs after whole body irradiation and autologous leucocyte transfusion obviously reflected the recovery of the haemopoietic tissue within the first two weeks (26). 18

1.1.1.5 Cryopreservation and density gradient separation of peripheral blood progenitor cells before autologous transplantation A number of studies involving peripheral blood and bone marrow in a variety of animal species suggest that conventional methods for freezing and cryopreserving produce little or no loss of subsequent functional ability of progenitor cells (45, 66, 106, 129), as assayed by these meth- ods. Nothdurft et al. (92) studied a group of 8 lethally irradiated dogs that were transfused autologously with cryopreserved MNC, derived from the peripheral blood by leucapheresis, according to the method described by Buckner et al. (15). The number of MNC transfused per kg b.w. ranged from 0.32 to 1.63 x 109. The CFU-c content infused varied between 0.02 and 1.38 x 10s, which corresponded with 0,2 to 1,5 x 104 CFU-c/ kg b.w. In all anin. a haematological recovery was observed using conven- tional cytomorphological methods of bone marrow evaluation. In cultures of bone marrow, obtained from irradiated dogs, CFU-c were not detectable or present at only very low numbers on day 3 after transplan- tation. Thereafter a rapid rise of the CFU-c numbers, related to the number of MNC infused, was observed in 6 out of 8 dogs studied, approximating or reaching the pre-irradiation level between day 15 and 20 after grafting. The reappearance of CFU-c in the peripheral blood varied consider- ably in time from dog to dog. But in 2 dogs that had received the lowest CFU-c numbers, the regeneration of the bone marrow CFU-c content was markedly delayed. In general, the time course of the bone marrow repopulation by CFU-c for single dogs corresponded with the regeneration pattern of the blood CFU-c numbers. The total number of CFU-c in the bone marrow returned to normal levels around day +15 to +20, if the number of MNC transfused was higher than 1 x 109 per kg b.w. On the other hand, the time course of the curves for the blood CFU-c levels was of the same kind as for the granulocyte values, which reached normal levels mainly around day +30 and thereafter. For the lymphocytes the normal range was not reached until day +100, whereas approximated the pre-irradiation values on about day +50 and thereafter. That the recovery of the haemopoietic system after white infusion had to be attributed to the transfused pro- genitor cells, became apparent from cytogenetic observations in dogs, by the use of allogeneic leucocytes from donors of the opposite sex (34,37). 19

Sarpel et al. (120) studied semi-continuous flow centrifugation (SCC) in canines as a method to obtain adequate numbers of cells for auto- logous marrow repopulation following supralethal cyclophosphamide administration (100 mg/kg). Four cycles using a 225 ml bowl and 30 ml/min. flow rate were the conditions for procurement. In 30 dogs collections averaged 8.4 ± 0.4 x 109 cells. MNC comprised 75 ± 2% of the cell population and CFU-c totalled 1.9 ± 0.09 x 10s per collection. 86% of MNC were T-lymphocytes in the 30-90 second fraction, compared with 49% in the 150-210 second fraction. CFU-c were equally distributed throughout the collection period. Following freezing and rapid thawing 92 ± 12% of CFU-c were recovered. Using a dose of 0.5 x 109 MNC/kg b.w. for reinfusion 4 out of 4 dogs died, with only one dog showing temporary evidence of early repopula- tion, whereas a dose of 1 x 109/kg b.w. was followed by a marrow repopulation and clinical recovery in 4 out of 4 dogs. In an effort to establish the best timing for cell administration, serial serum samples of dogs having received cyclophosphamide were tested for inhibition of CFU-c. Sera from 6 dogs obtained 1 hoi"" following cyclo- phosphamide administration totally suppressed the CFU-c activity of normal dog peripheral blood cells. The 24 hour post cyclophosphamide sera enhanced CFU-c activity above control values. Recently, Storb et al. (132) demonstrated the presence of HSC in the blood of baboons, in which haemopoietic grafts could be achieved by means of a cross-circulation technique. Sustained long term bone marrow repopulation indicated that HSC were present among the infused peri- pheral blood leucocytes (37, 128) or in the blood of non-irradiated cross- circulated partners (33). Circulating HSC could therefore be used instead of marrow or as a supplement to marrow for allogeneic haemopoietic transplantation (132). The time until onset and the rate of haemopoietic recovery can be shortened and increased, respectively, when large numbers of mononuclear cells are transfused (38, 53, 92). However, graft versus host (GVH)reaction was observed in many dogs after allogeneic, DLA matched leucocyte transfusions (38, 130), indistinguishable in its appear- ance from that seen after bone marrow transplantation (131). Some authors (134) doubt the feasibility of reducing the incidence, morbidity, and mortality of GVH-reaction by obtaining a purified "blood stem cell" population by removing the cells that initiate, promote, or enhance GVH-reaction from leucocyte suspensions. However, the observations by Dicke and Van Bekkum in mice, monkeys and men indicate that GVH-reaction was reduced or eliminated when albumin gradient separated bone marrow cells were transfused (29). 20

Korbling et al. (58) demonstrated that this albumin gradient separation technique is also feasible for peripheral progenitor cells. They presented data which indicate that a 4-step procedure, consisting of leucocyte mobilization by means of DS, leucapheresis by means of continuous flow centrifugation (CFC), red cell removal by a Ficoll-Isopaque gradient and finally an albumin gradient cell separation, results in a "purified" popu- lation of CFU-c without immunocompetent cells. In that study dog blood leucocytes were collected from an arterio-venous shunt by means of leucapheresis using the IBM-blood cell separator. In order to increase the yield of CFU-c, DS (15 mg/kg b.w.) was given intravenously 30 minutes before the onset of centrifugation (117). The number of MNC collected during a 3-4 h leucapheresis was between 6.5 and 14 x 109. After elimination of red cells by a Ficoll-Isopaque gradient separation according to B^yum (8). between 0.1 and 1.3 x 109 cells were loaded onto a discontinuous albumin gradient that is essentially the same as described by Dicke et al. (27). This gradient was centrifuged at 1,000 g for 30 minutes at 10°C, and cells from fraction 1 to 6 were stored at -196°C in 10% dimethylsulphoxide (DMSO) and used later for in vivo transplantation. It was found that the highest CFU-c accumulation (82%) was in fraction 2 (between 17% and 25% albumin), which correlates with a density between 1.052 and 1.073 g/ml. The cells of fractions 1, 2, 3, and 4 were thawed rapidly and suspended in 10 ml medium with 20% foetal calf serum and injected immediately into 4 recipient dogs respectively. The donor dog was histocompatible with the recipient dogs, using the criteria of DLA and MLC, as well as blood group identity, but was of the opposite sex to allow cytogenetic detection of chimerism. The survival time of dog I was too short (+11 days) to allow evaluation of possible GVH-reactions. Dog II, receiving fraction 2 cells (10 ml, con- taining 42 x 106 MNC, among which were 3.23 x 106 CFU-c), showed a rapid and permanent marrow recovery and displayed no signs of GVH- reaction up to 14 months after grafting. Dogs III and IV died on day +21 and +25 after grafting, with signs of GVH-reaction and aplasia of the bone marrow. The dog receiving fraction 4 cells showed little haemo- poietic recovery, but a marked lymph node hyperplasia consisting of plasma cells. It is of interest to note that a positive correlation between haemo- poietic engraftment and the number of MNC transfused does not seem to exist to that extent for the MNC cell population as it does for the quantity of CFU-c it contains. For instance, in the above mentioned 21

experiment, dog II received a total of 42 x 106 MNC, while dog IV re- ceived 899 x 106 MNC. If one compares the number of CFU-c trans- fused, there were 3.2 x 106 transfused into dog II, and 0.12 x 106 into dog IV. All dogs showed a rapid bone marrow regeneration, but only dog II survived and did so without GVH-reaction. From these observations it appears essential to specify not only the number of MNC transfused, but also the number of CFU-c contained in the cell suspension.

1.1.2 Haemopoietic progenitor cells in peripheral blood; human studies

1.1.2.1 CFU-c: definition and several characteristics The observation that HSC can be detected in the peripheral blood in a variety of animal species, suggests that such cells may also circulate in man. In order to prove this, a HSC-assay suitable for use in man has yet to be developed. Until then, the demonstration that human peripheral blood leucocytes include a population of cells committed to granulocyte/ monocyte differentiation (CFU-c) may be taken as indirect evidence that HSC are also present.* The observation that the Philadelphia chromosome, characteristic of chronic myelogeneous leukaemia, can be identified in granulocytes, monocytes, erythroid, and megakaryocytic cells, might count as some evidence that HSC exist in man (63, 108). The capacity of transplanted bone marrow to repopulate depleted myeloid tissue in man, does not unequivocally prove the existence of a HSC in human marrow, for the same result could be achieved by a series of unipotential (committed) progenitor cells, i.e. if all the committed precursors of the various specific cell lines had self regenerating capacity. Indirect evidence that a HSC circulates in the peripheral blood of man was derived from the observation that peripheral blood leucocytes I transfused from one identical twin to another appeared to be responsible for early bone marrow recovery after chemotherapy in a study reported by McCredie et al. (70). This observation is supported by circumstantial evidence derived from other clinical experiences (12, 122). The development of methods for growing murine bone marrow cells

•Recently, for human bone marrow and peripheral blood too, a "mixed" colony assay was de- scribed, which probably detects human HSC (Blood, 53: 1023,1979). 22

in semi-solid media rapidly led to methods for growing human bone mar- row CFU-c in vitro (101). The number of CFU-c assayed in normal human marrow largely depends on the details of the culture system used and the criteria for defining colonies. With these restrictions most workers have reported CFU-c number in normal marrow between 25 and 50 per 10s nucleated cells. Granulocytic and erythrocytic progenitor cells have been cultured in vitro from human peripheral blood (19, 22,43, 71, 94, 110, 112). The concentration of CFU-c in the peripheral blood varies between 0.5 and 20 per 106 nucleated cells, which represents 1-10% of that found in bone marrow. If one defines a colony as more than or equal to 20 cells per colony, Tebbi et al. (133) found 60 ± 23 CFU-c per 1 x 106 MNC in 15 experiments. The same results were found by Rubin et al. (119): in 26 individuals a mean of 65.9 ±31.5 CFU-c per 1x10* plated cells were found, and 79.8 ± 30.4 and 55.9 ± 28.3 were obtained from males and females respectively. Debelak-Fehir et al. (26) investigated the CFU-c capacity of fresh and cryopreserved isolated MNC from 15 normal individuals, defining a colony as more than or equal to 50 cells; a mean (± s.e.m.) of 26 ± 1.69 CFU-c was found per 1 x 106 MNC, and 14.0 ± 1.54 for nucleated buffy coat cells. For erythroid precursor cells Clarke et al. (22) found a mean number of 15.6 ± 2.1 BFU-e per 1 x 10s MNC in normal human peripheral blood after 13 days of culture. From the above data it can be concluded that the ratio of CFU-c in blood and bone marrow is approximately 1:25 to 1:50, which is reason- able close to the ratio observed in mice. It is, however, possible that peripheral blood CFU-c values are an underestimate. The presence of granulocytes in a culture system inhibits the proliferation of CFU-c. Experiments in which MNC are cultured after removal of granulocytes may thus show proportionately greater increase in CFU-c number; with a linear relationship between the number of plated cells and counted colonies (119). I Tebbi et al. (133) studied some properties of human peripheral CFU-c. Cell populations in peripheral blood responsible for granulopoie- sis in culture resemble those in marrow: in both cases granulopoietic progenitors coexist with cells that produce colony stimulating activity (CSA). Thus, addition of CSA to unseparated peripheral blood cells has little effect on colony formation, and there is a linear relationship be- tween the number of nucleated cells plated and the number of observed 23

colonies in ihe presence or absence of added CSA. Depletion of leuco- cyte suspensions of monocytes reduced colony formation; addition of either CSA or adherent cells restored colony formation capacity. This cell-cell interaction underlying granulopoiesis in cultures of peripheral blood cells is similar to that previously described for human marrow cells (50). Velocity sedimentation showed a peak at 4 mm/h for CFU-c; this in contrast to the value of marrow CFU-c which is between 5 and 6 mm (76). 3 H-thymidine was used to determine the percentage of peripheral CFU-c in DNA synthetic phase (S-phase) of the cell cycle. Their data showed that few, if any, granulopoietic progenitors incorporated suffi- cient radio-isotope to destroy their colony-forming capacity. In contrast, the marrow specimens showed greater reductions (35%), indicating a significant number of cells in S-phase (46). Finally, a corre- lation was observed between the CFU-c capacity of marrow cells and of peripheral leucocytes selected on the basis of their density in Ficoll- Isopaque gradients. Studies in mice showed that bone marrow CFU-s and CFU-c concen- trations increase in parallel after administration of vinblastine (18) and cyclophosphamide (9), so the same could be true in man. In patients receiving adriamycin and cyclophosphamide for treatment of a variety of solid tumors Richman et al. (110) found that CFU-c numbers reached their peak 21 days after chemotherapy at which time values up to 20 times the starting number were found. The authors calculated that the increase in circulating peripheral blood CFU-c after chemotherapy made leucapheresis a suitable method for collecting adequate numbers of precursor cells, because peripheral blood CFU-c could be maintained with cyropreservation for months with a median CFU-c recovery of 99.5% (26, 110). These cells could be used as auto- logous support for patients undergoing intensive chemotherapy. In their example comparable numbers of CFU-c could be collected in one 90 min. bone marrow harvest and in one 17 liter leucapheresis.

1.1.2.2 Leucapheresis in humans; quantitative data on CFU-c yields Both and granulocyte preparations obtained by pheresis con- tain large numbers of MNC, consisting of immunocompetent as well as haemopoietic progenitor cells (19, 71, 110, 116). 24

The ability to procure high concentrations of progenitor cells may facilitate studies of autologous reconstitution for the support of severely myelosuppressed patients. Specific pheresis techniques, designed to harvest MNC efficiently, must provide large numbers of these cells with a high degree of purity in relatively small volumes. Recently Weiner et al. (144) investigated the use of SCC for collecting and concentrating large numbers of MNC and quantitated the yield, purity and function of immunocovnpetent cells and haemopoietic precursor cells from human peripheral blood. The yield of MNC obtained from 15 donors undergoing pheresis was 6.4 ± 7 x 109 (mean ± s.e.m.) in a 30 ml volume. The yields were based on the assumption that 600 ml of blood entered the centrifugation bowl in order to generate the buffy coat. Each buffy coat can yield up to 1 x 109 MNC with a blood loss of 30 ml and requires only 20 minutes. The distribution of various cell types in the buffy coat was investigated. The buffy coats generated by pheresis were collected in 5 ml aliquots (at a flow rate of 30ml/min.), beginning with clear plasma. The maximum concentration of platelets preceded the maximum concentration of leu- cocytes. Platelets, however, remained present in high concentrations throughout the leucocyte collection. Further centrifugation (150 g, 15 min., 15°C) eliminated 80% of the platelets without loss of MNC or CFU-c. Sequential 5 ml fractions were analysed with respect to E-rosette and CFU-c content. The CFU-c concentration (colony defined as more than or equal to 20 cells) per 2 x 105 FicolHsopaque separated MNC was around 35 in all fractions. Monocytes and lymphocytes were dis- tributed similarly throughout the buffy coat and selective pheresis of either element was not possible under the conditions used in their experiments. In addition, no separation of progenitor cells from immuno- competent cells was observed. With CFC, a mean (± s.e.m.) of 1.5 ± 0.14 x 1010 leucocytes could be collected in a 4-hour centrifugation program (89). Comparison of the leucocyte yields in the first, second, third and fourth hour of leucaphe- resis indicated a progressive increase with 3.8 times more leucocytes I collected in the last hour of leucapheresis as compared to the first hour. Although the mechanism of this improvement in yield with time is not known, these data indicate that moderate prolongation of the leucapheresis-duration can result in an increase in efficiency, which is comparable to that produced by the use of hydroxy ethyl starch (HES), etiocholanolone or corticosteroids (67, 72, 85, 91). Korbling et al. (59) studied 9 healthy volunteers who were each 25

subjected to CFC. Twenty-six 4-hour leucaphereses yielded between 6.3 and 18.7x 109 MNC (mean 12.4 x 109) and between 1.5 and 15.8 x 10s CFU-c (mean 8.3 x 10s). The blood volume processed during one run was 12 L, representing 2.3 times the donor's blood volume. The collected MNC, among which the CFU-c, were cryopreserved using 10% DMSO as a cryoprotective agent. The cells were stored for at least 2 weeks in liquid nitrogen, then thawed quickly in a 56°C water bath and washed to remove practically all DMSO. The cell yields after thawing were 88.8% for the MNC and 89.9% for the CFU-c. If several CFC were performed in a rapid sequence (up to 3 in one week), an increase in CFU-c yield per leucapheresis was observed. The increment varied between 36.8 and 232.8%. In canine experiments, the lowest observed value for MNC and CFU-c respectively to obtain successful permanent autologous engraftment was 0.06 x 109 MNC/kg b.w. and 0.04 x 10s CFU-c/kg b.w. If these values should also apply to a 70 kg adult human being, these data can be interpreted to mean that the yields of one leucapheresis is sufficient to obtain a stem cell engraftment in a patient with an aplastic bone marrow. The precise dose of peripheral blood progenitor celJs required for trans- plantation has not yet been clearly established. Epstein et al., studying dogs, determined that a mean of 1.5 x 104 CFU-c/kg b.w. are necessary for a successful autologous engraftment, which would correspond to a total of 1.0 x 106 CFU-c for a 70 kg human adult. Considering the discomfort endured by donors of haemo- poietic tissue, the use of such an approach offers evident advantages. Moreover, a vast number of cells can be collected with ease. With the introduction of even more aggressive forms of cytotoxic therapy, there is a potential for an enormous expansion of the supporting role of haemo- poietic progenitor cells, particularly in the form of autografts (136) (Table 1 .II).

1.1.3 Collection and purification of progenitor cells from peripheral blood Methods for separation of various cell populations are based upon differ- ences in physical or membrane properties of cells. The first methods allowing preparative physical fractionation of discrete types of cells from complex mixtures were based on fractionation in various kinds of gra- dients. These include different buoyant density separation systems (126). When a cell is centrifuged in a density gradient it will sink or rise until it reaches the region corresponding to its buoyant density. Thus, the fractionation in these systems is based on differences in the densities of Table 1.II. Quantitative information on CFU-c yields obtained with leucapheresis in dogs and man

A. Dogs: - Semicontinuous flow centrifugation (SCO (Ref. 1): results from 4 runs (2 hours each); 225 ml bowl, filling rate: 60-80 ml/min. Collection: 30 ml/min during 210 sec. Results are com- Stem cell source pared to routine multiple bone marrow as- pirations (in dogs). Bone marrow Peripheral blood

- Total yield of MNC (xlO9) 4.7(±0.8) (N = 14) 8.4(±0.4) (N = 50) - Total yield of CFU-c (xlO5) 1.7(±0.5) (N = ll) 1.9(±0.3) (N = 50) - Final volume (ml) 135(±5) (N = 17) 98(±5.8) (N = 30) - Autologous engraftment observed with: MNC l(±O.6)xlO-*/kgb.w. l(±O.l)xlO9/kgb.w. CFU-c 1.3(±0.3)xl04/kgb.w. 1.6(±0.2)xl0«/kgb.w. to B. Man: - Continuous flow centrifugation (CFC) (Ref. 2) BUT: 4 hour run in AMINCO-blood cell separa- mean MNC yield: 12x10' per run Supplemental increase of CFU-c tor; SO ml/min flow rate. Results obtained yield during 3 consecutive phere- from 35 normal donors. Calculations based mean CFU-c yield: 8.7xl0s per run sis-proceduies:mean yield of 3 runs on CFU-c frequency in peripheral blood: (N = 7 donors) 6 50-80/lxlO MNC * 250 CFU-c/ml blood. CFU-c: MNC = 1:14,000 = 23xl0s CFU-c i for a 70 kg adult, this represents: 3.2x10* CFU-c/kg b.w. I Conclusion: engraftment with peripheral haemopoietic stem cells in man is feasible

Ref. 1: R.B. Epstein et al. (1978): Exp. Hemat., Vol. 7, suppl. 5,125-134 Ref. 2: T.M. Fliedner et al. (1978): Exp. Hemat., Vol. 7, suppl. 5, 398-408 27 the cells. These methods have been applied for fractionation of white blood cells of different types (124, 145) and separation of bone marrow stem cells from immunocompetcnt cells present In the marrow (27). Several other cell separation procedures have been described such as free flow electrophoresis and zonal centrifugation, techniques that have been thoroughly reviewed by Shortman (126) and Cutts (24). Miller and Philips (83) developed a method to fractionate popu- lations of living cells based on differences in size by sedimentation at unit gravity. Sedimentation rate is a function of both cell size and den- sity but is determined primarily by differences in cell size. Nevertheless, density does influence the sedimentation velocity. Consequently, cells with different sedimentation velocities can have the same volume and cells with the same sedimentation velocity can differ in size (126). The unit gravity separation procedure has been applied to human blood cells by Brubaker and Evans (13). They obtained good separation of the various elements in 2.5 h, the order of increasing sedimentation rate being platelets, erythrocytes, lymphocytes, monocytes and granulo- cytes. The separation largely followed the order of cell size, but the high density of erythrocytes caused them to approach the lymphocyte peak, and the high density of granulocytes increased their sedimentation relative to monocytes. Peterson and Evans (99) initially demonstrated that different mor- phological classes of cells from bone marrow showed different sedi- mentation characteristics. Worton et al. (147) demonstrated the separa- tion of cells forming CFU-s and CFU-c by unit gravity separation. Separation of cells on the basis of buoyant density differences, how- ever, has been one of the most extensively applied physical separation procedures. Three general approaches have been used: 1. neutral density separation, where cells are separated simply by their floating or sinking in medium with a particular density; 2. discontinuous gradient separation, where a series of discrete layers of medium of different density are used and the cells concentrate at the various interphases; 3. true isopycnic gradient separation, where the density of the medium continuously changes and the cells band at their buoyant density. Since the density of a cell depends on its water content, and since cells act as osmometers, the actual buoyant density will vary with the osmolarity of the medium. Buoyant density values are not meaningful unless they are precisely controlled, and a true density distribution of a cell population will only be obtained in a density gradient with con- stant, physiological osmolarity throughout. 28

The first really effective analytical and preparative isopycnic-separa- tion procedure was developed by Leif and Vinograd (64). Shortman (125) has adapted many of their concepts in a system originally designed to give optimal separation of lymphoid cells. Discontinuous gradient procedures are commonly used because of their simplicity. Adequate separation is achieved between those cells that differ sufficiently in density (27). When the aim is to isolate only cells of one particular density from a complex population, this can be accomplished with neutral density separation. It is particularly effective when a prior continuous density gradient analysis has indicated the most effective density to isolate the cells under investigation. Numerous workers have applied continuous density gradients for the separation of human white blood cells (97, 98, 152). The relative order of increasing cell density is monocytes, lymphocytes and granulo- cytes. Monocytes band close to the lymphocyte peak, so separation is only partial, but there is a good separation between mononuclear cells and the polymorphonuclear cells. It is of interest that human blood lymphocytes form essentially a single density peak, in contrast to the multiple peaks found with lymphoid cells obtained from mice and rats (123). Density separation has been used to purify and characterize haemo- poietic stem cells in mice and men. Turner et al. (139) described a continuous albumin gradient technique by which they separated mouse bone marrow cells. A discontinuous albumin gradient was employed by Raid et al. (107) and by Harris and Joseph (49). The application of density separation procedures in the field of human bone marrow has been pioneered by Dicke et al. (27, 28, 29, 30), who applied separation in discontinuous albumin gradients to eliminate lym- phocytes that cause acute secondary disease from the marrow stem cells. Using albumin density gradients Dicke et al. (30) and Moore et al. (88) have achieved up to a 100-fold purification of bone marrow CFU-c from primates. Messner et al. (77) have fractionated human bone marrow by velocity sedimentation at 1 g and have studied functional aspects of the resulting purified populations. Although the cells obtained by these techniques are homogeneous in cell size and/or density, they are heterogeneous in function, with CFU-c comprising only a small percen- tage of the total cells. Not any physical separation procedure is likely to accomplish a complete purification or unique characterization of any particular cell, 29

so several procedures have to be combined (84). The fragility of living cells demands the number of steps and time involved to be kept to a minimum. However, other methods are available, reflecting biological specificity of different cells; they will increase in importance. For example, T- lymphocytes with a specific affinity to sheep erythrocytes can be easily isolated after interaction with red cells to form large "rosettes" (56), which then have an enhanced sedimentation velocity (32). During the last few years considerable advance has been made in the field of specific surface markers to allow selective depletion or enrichment of a given subpopulation of cells (7, 31, 115, 145). The use of surface membrane marker properties to obtain a unique subpopulation of mononuclear cells offers an alternative approach to purify CFU-c and study their charac- teristics. Recent studies have identified the precursor cells for both mature B- and T-lymphocytes in a population of peripheral MNC that lack intrinsic surface immunoglobulin and sheep erythrocyte receptors (21, 41). One might predict that the progenitor cells for the granulocytic series, and maybe also for the megakaryocytic and erythroid series, are also present in this so-called "null cell" population. Richman and Chess (109) recently provided evidence, that the number of CFU-c is increased in a preparation of null lymphocytes, using Ficoll- Isopaque density gradient centrifugation followed by Sephadex-G-200 anti-Fab column chromatography (= depletion of B-cells) and E-rosette sedimentation (= depletion of T-cells). 6hl et al. (95) used a laborious 5-step purification technique with subsequent elimination of monocytes, B- and Fc-receptor bearing lymphocytes and finally T-cells. They ob- tained a CFU-c enriched fraction, which equals the mean CFU-c content in unfractionated bone marrow. Also because of the multistep approach, a considerable loss of cells occurred, and ultimately 4% of the original MNC-population was recovered. A laborious multi-step procedure was also used by Barr et al. (3), using rosette sedimentation techniques combined with velocity sedimentation. Richman et al. (Ill), using immuno-absorbent column chromatography and rosette depletion techniques (E- and EAC-rosette sedimentation), confirmed that CFU-c are not present in either purified T- or B-lympho- cyte populations, but are highly concentrated in the null cell population. Besides that, they also demonstrated that CFU-c do not bear receptors for complement. 30

1.2 CONCLUSIONS

A) From animal studies it can be concluded that: 1) haemopoietic progenitor cells are circulating in the peripheral blood in a variety of animal species; 2) in dogs, consistent haemopoietic engraftment can be achieved using MNC from the peripheral blood; 3) the minimal "dose" of peripheral blood progenitor cells required for transplantation has not yet been clearly established, but varies between 0,32 and l>63x 109 MNC/kg b.w., corresponding with approximately 0,2-1,5 x 104 CFU-c/kg b.w. in dogs; 4) using equal numbers of peripheral blood CFU-c, there seems to be no difference in haemopoietic recovery patterns if the transplan- tation is allogeneic or autologous in origin; 5) the speed of haemopoietic engraftment is directly correlated with the number of CFU-c infused; 6) the number of peripheral blood CFU-c collected with different leucapheresis-techniques can be increased by chemotherapy, se- quantial pheresis and lymphocyte mobilizing agents such as DS, pyran, and PVSA; 7) cryopreservation of peripheral progenitor cells for prolonged periods is feasible, with only a limited loss in number and/or function; 8) separation between progenitor cells and immunocompetent cells, although incomplete, is observed after leucapheresis; 9) in dogs, it seems possible to prevent GVH-reaction by using pro- genitor cells obtained from fraction 2, after discontinuous albumin gradient separation of peripheral MNC. B) From human studies it can be concluded that: 1) CFU-c are circulating in the peripheral blood in a concentration of approximately 60 CFU-c/1 x 106 MNC, if a colony is defined as equal to or more than 20 cells; 2) CFU-c are present in the null cell population in the peripheral blood; 3) cryopreservation is possible; 4) no functional separation between haemopoietic progenitor cells and immunocompetent cells is achieved with leucapheresis; 5)usingCFC, between 6.3 and 18.7 x 109 MNC (mean 12.4 x 10?) can be collected, corresponding with 1.5-15.8 x 10s CFU-c (mean 8.3 x 10s) during a single 4-hour pheresis. 31

6) sequential pheresis will increase the CFU-c yield with a value be- tween 36.8 and 232.8%; 7) if the cell dose required for haemopoietic engraftment in dogs (1,6 x 104 CFU-c/kg b.w.) is applied to man, then approximate 1,0 x 106 CFU-c in total are required for a 70 kg adult. This could be achieved with about 1-3 CFC procedures (Table 1 .II).

1.3 PURPOSE AND DESIGN OF THE INVESTIGATION

The purpose of the investigation to be reported here is to purify haemo- poietic progenitor cells from human peripheral blood using density gra- dient centrifugation in order to isolate a progenitor cell fraction without immunocompetent cells. This technique could then be used in the clinical situation where limited numbers of progenitor cells are required, for instance in pediatric transplantation, to obtain a haemopoietic repopulation without the risk of acute secondary disease. In situations where it appears impossible to obtain bone marrow stem cells, an alternative could be to collect these cells from the peripheral blood. Otherwise, this technique could be used in the situation where insufficient numbers of bone marrow cells are harvested, so that this deficit could be compensated with peripheral pro- genitor cells. By combining different rosette techniques it was feasible to achieve a progenitor cell purification in a limited number of density gradient centrifugation steps. The final suspension contained a very small number of immunocompetent cells. To justify clinical use in the future, more characteristics of this progenitor cell population were investigated with in vitro techniques, e.g. CFU-c capacity, in vitro response to mitogens and MLC-tests, number of CFU-c in S-phase, radiosensitivity (Do-value) and volume distribution. Furthermore, "purified" progenitor cells were cultured using various diffusion chamber techniques, to obtain more information about the proliferation and differentiation capacity of human peripheral progenitor cells. Especially "histological" investigation of methyl-methacrylate embed- ded plasma clot diffusion chambers affords a clear insight into the differen- tiation capacity of the various haemopoietic progenitor cells, circulating in human peripheral blood. These techniques could also contribute to the understanding of the pathophysiology of several haematological dis- 32

orders. Therefore we investigated qualitative and quantitative aspects of peripheral progenitor cell activity in several patients with myeloprolifer- ative disorders. The material and methods used in this thesis are described in chapters II, IIIand IV. Chapter II deals with the purification technique of peripheral blood CFU-c by means of density gradient centrifugation and a combined depletion of various rosettes. In chapter HI the results of several "in vitro" characteristics of purified CFU-c suspensions are presented. In chapter IV the results are described of the plasma clot diffusion chamber culture technique. Emphasis is laid on the "histological" analysis of colony formation in this "in vivo" culture system. Chapter V finally deals with results obtained with the different techniques described in chapters II and IV by analysing peripheral pro- genitor cell activity in myeloproliferative disorders.

1.4 REFERENCES

1. Abramson S, Miller RG and Phillips RA: The identification in adult bo.;e marrow of pluri- potent and restricted stem cells of the myeloid and lymphoid system. J. Exp. Med., 145: 1567,1977. 2. Barnes DWH and Loutit JF: Haemopoietic stem cells in the peripheral blood. Lancet, II, 1138,1967. 3. Barr RD, Whang-Peng J and Perry S: Hemopoietic stem cells in human peripheral blood. Science, 190: 284,1975. 4. Becker AJ, McCulloch EA and Till JE: Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature, 197: 452, 1963. 5. Becker AJ, McCulloch EA, Siminovitch L and Till JE: The effect of differing demands for blood cell production on DNA synthesis by hemopoietic colony forming cells of mice. Blood, 26: 296,1965. 6. Bezin GI, Khaitov RM, Moroz BB, Petrov RV and Romashko OO: The factors controlling stem cell circulation. II. ACTH-induced inhibition of migration of hemopoietic stem cells. Blood, 46: 79,1975. 7. Bianco C, Patric R and Nussenzweig V: A population of lymphocytes bearing a membrane receptor for antigen-antibody complement complexes. I. Separation and characterization. J. Exp. Med., 132:702,1970. 8. B^yum A: Separation of leukocytes from blood and bone marrow. Scand. J. Clin. Lab. Invest., 21, suppl. 97,1968. 9. B^yum A, Carsten A and Laerum O: Haematopoiesis measured by spleen colony and diffusion chamber techniques in mice treated with one or two injections of cyclophos- phamide. Brit. J. Haemat., 26: 605,1974. 10. Bradley TR and Metcalf D: The growth of mouse bone marrow cells in vitro. Aust. J. Exp. Biol. Med. Sci., 44: 287,1966. 33

11. Brecher G and Cronkite EP: Postradiation parabiosis and survival in rats. Proc. Soc. Exp. Biol. Med., 77:292, 1951. 12. Bronson WR, McGinniss MH and Morse EE: Hematopoietic graft detected by a change in ABO group. Blood, 23: 239,1964. 13. Brubaker LH and Evans WH: Separation of granulocytes, monocytes, lymphocytes, ery- throcytes, and platelets from human blood and relative tagging with diisopropylfluoro- phosphate (DFP). J. Lab. Clin. Med., 73: 1036,1969. 14. Bruch C, Herbst EW, Calvo W, Hugct R, Flad HD, and Fliedner TM: Freezing of blood leukocytes for transplantation into lethally irradiated dogs. In RS Weiner, RK Oldham & L Schwarzenberg (eds), La cryopreservation des cellules normales et neoplastiques, 51. Institut National de la Same', Paris, 1973. 15. Suckner E, Eisel R, and Perry S: Blood cell separation in the dog by continuous flow centrifugation. Blood, 31: 653,1968. 16. Calvo W, Fliedner TM, Herbst EW, Hiigl E and Bruch C: Regeneration of blood forming organs after autologous leukocyte transfusion in lethally irradiated dogs. II. Distribution and ccllularity of the marrow in irradiated and transfused animals. Blood, 47: 593, 1976. 17. Cavins JA, Schecr SC, Thomas ED and Ferrebee JW: The recovery of lethally irradiated dogs given infusions of autologous leukocytes preserved at -80°C. Blood, 23: 38, 1964. 18. Chen M and Schooly J: Recovery of proliferative capacity of agar colony forming cells following ionizing radiation and vinblastine. J. Cell. Physiol., 75: 89, 1969. 19. Chervenick PA and Boggs DR: In vitro growth of granulocytic mononuclear cell colonies from blood of normal individuals. Blood, 37: 131, 1971. 20. Chervenick PA: Interrelationship of colony stimulating factor (CSF) and colony forming cells (CFC) in blood and bone marrow during marrow regeneration. (Abstract) J. Clin. Invest., 52: 18A, 1973. 21. Chess L, Levine H, MacDermott RP and Schlossman SF: lmmunologic functions of iso- lated human lymphocyte subpopulations. IV. Further characterization of the surface Ig negative, E-rosette negative (null-cell) subset. J. Immunol., 115: 1483, 1975. 22. Clarke BJ and Housman D: Characterization of an erythroid precursor cell of high pro- liferative capacity in normal human peripheral blood. Proc. Natl. Acad. Sci. U.S.A, 74, No. 3: 1105,1977. 23. Cole LJ: Hemopoietic restoration in lethally X-irradiated mice injected with peritoneal cells. Amer. J. Physiol., 204: 265,1963. 24. Cutts JH: Cell separation: Methods in Hematology: New York & London: Academic press, 1970. 25. Debelak-Fehir KM and Epstein RB: Restoration of hemopoiesis in dogs by infusion of cryopreserved autologous peripheral white cells following busulfan-cyclophosphamide treatment. Transplantation, 20: 63,1975. 26. Debelak-Fehir KM, Catchatourian R and Epstein RB: Hemopoietic colony forming units in fresh and cryopreserved peripheral blood cells of canines and man. Exp. Hemat., 3: 109,1975. 27. Dicke KA, Van Hooft JIM, and Van Bekkum DW: The selective elimination of immuno- logically competent cells from bone marrow and lymphatic cell mixtures. II. Mouse spleen cell fractionation on a discontinuous albumin gradient. Transplantation, 6: 562, 1968. 28. Dicke K A: Bone marrow transplantation after separation by discontinuous albumin density gradient centrifugation. Thesis. Rijswijk, The Netherlands: Radiobiological Institute, TNO, 1970. 29. Dicke KA and Van Bekkum DW: AUogeneic bone-marrow transplantation after elimina- tion of immunocompetent cells by means of density gradient centrifugation. Transpl. Proa, 3: 666,1971. 30. Dicke KA, Van Noord MJ, Schaefer UW and Van Bekkum DW: Attempts at morphological identification of the haemopoietic stem cell in primates and rodents. In Haemopoietic stem cells. CIBA Foundation symposium 13. North-Holland, 48,1973. 34

31. Dickler HB and Kunkel HG: Interaction of aggregated gamma globulin with human B-lymphocytes. J. Exp. Med., 136: 191,1972. 32. Edwards GE, Miller RG and Philips RA: Differentiation of rosette-forming cells from myeloid stem cells. J. Immunol., 105: 719,1970. 33. Epstein RB, Graham TC, Buckner CD, Bryant J and Thomas ED: AUogeneic marrow en- graftment by cross circulation in lethally irradiated dogs. Blood, 28: 692,1966. 34. Flad HD, Goldmann SF, Huget RP et al.: Die Bedeutung der Histocompatibilitatsbestim- mung fur die Transfusion allogener Blutstammzellen beim Hund. Forschungsergebnisse der Transfusionsmedizin und Immunhaematologie, Berlin Medicus Verlag, 2: 834, 197S. 35. Fliedner TM, Thomas ED, Meyer LM and Cronkite EP: The fate of transfused 3 H-thymi- dine labelled bone-marrow cells in irradiated recipients. Ann. N.Y. Acad. Sci., 114: 510, 1964. 36. Fliedner TM, Haas RJ, Stehle H and Adams AC: Complete labelling of all cell nuclei in newborn rats with 3H-thymidine. Lab. Invest., 18: 249,1968. 37. Fliedner ThI: Pathophysiologische Grundlagen der Transfusion hamopoetischer Stamm- zellen und Profe'eme ihrer Gewinnung. Forschungsergebnisse der Transfusionsmedizin und Immunhaematologie, Berlin Medicus Verlag, 2: 781,1975. 38. Fliedner, TM, Flad HD, Bruch C, et al.: Treatment of aplastic anemia by blood stem cell transfusion: a canino model. Haematologica, 61: 141,1976. 39. Ford CE, Hamerton JL, Barnes DWH and Loutit JF: Cytological identification of radia- tion-chimaeras. Nature, 177: 452,1956. 40. Fowler JH, Wu AM, Till JE, McCulloch EA and Siminovitch L: The cellular composition of hemopoietic spleen colonies. J. Cell. Physiol., 69: 65,1967. 41. Gatien JG, Schneeberger EE and Merler E: Analysis of human subpopulations using discontinuous gradients of albumin: precursor lymphocytes in human thymus. Eur. J. Immunol.,5: 312,1975. 42. Gidali J, Fe'her J and Antal S: Some properties of the circulating hemopoietic stem cells. Blood, 43: 573,1974. 43. Golde DW and Cline MJ: Identification of the colony-stimulating cell in human peripheral blood. J. Clin. Invest., 51: 2981,1972. 44. Goodman JW and Hodgson GS: Evidence for stem cells in the peripheral blood of mice. Blood, 19: 702,1962. 45. Gray JL and Robinson WA: In vitro colony formation by human bone marrow cells after freezing. J. Lab. Clin. Med., 81:317,1973. 46. Greenberg P, Bax I, Mara B and Schrier S: Alterations of granulopoiesis following chemo- therapy. Blood, 44: 375,1974. 47. Haas RJ, Flad HD, Fliedner TM and Fache I: Correlation between cytokinetically resting lymphocytes and bone-marrow restoration: Experiments using a discontinuous albumin gradient. Blood, 42: 209,1973. 48. Hanks GE: In vivo migration of colony-forming units from shielded bone marrow in the irradiated mouse. Nature, 203: 1393,1964. 49. Harris PF and Joseph S: Fractionation of bone marrow suspensions and their use to protect against lethal irradiation. J. of Physiol. 182: 7,1966. 50. Haskill JS, McNeill TA and Moore MAS: Density distribution analysis of in vivo and in vitro colony forming cells in bone marrow. J. Cell. Physiol., 75: 2,1970. 51. Hays EF, Hays DM and Golde DW: Hemopoietic stem cells in mouse liver. Exp. Hemat, 6: 18,1978. 52. Hellman S: Circulating stem cells: Variation with duration of partial body X-irradiation. Nature, 205: 100,1965. 5 3. Herbst EW, Fliedner TM, Calvo W, Schnappauf HP and Meyer H: Untersuchungen iiber die Gewinnung hamopoetischer Stammzellen aus dem peripheren Blut von Hunden und iiber ihre Fahigkeit, die Blutzellbildung zu regenerieren. Blut, 30: 265,1975. 35

54. Iscove NN, Senn JS, Till JE and McCulloch EA: The proliferation states of mouse gra- nulopoietic progenitor cells. Proc. Soc. Exp. Biol. Med., 134: 33,1970. 55. Iscove NN, Senn JS and Till JE: Colony formation by normal and leukemic human mar- row cells in culture. Effect of conditioned medium from human leukocytes. Blood, 37: 1,1971. 56. Jondal M, Holm G and Wigzell H: Surface markers on human B- and T-lymphocytes. I. A large population of lymphocytes forming non-immune rosettes with sheep red blood cells. J. Exp. Med., 136: 207,1972. 57. Khaitov RM, Petrov RV, Moroz BB and Bczin Gl: The factors controlling stem cell recir- culation. I. Migration of hemopoietic stem cells in adrenalectomized mice. Blood, 46: 73,1975. 58. Kdrbling M, Fliedner TM, Calvo W, Nothdurft W and Ross WM: In-vitro and in-vivo properties of canine blood mononuclear leukocytes separated by discontinuous albumin density gradient centrifugation. Biomedicine, 26: 275,1977. 59. Kdrbling M, Fliedner TM, Pflieger H, Vassileva D and Heimpel H: Yield and efficiency of collecting and cryopreserving human hemopoietic blood stem cells (HBSC) by means of continuous flow centrifugation (CFC). Abstract, Suppl. No. 3: 95, Exp. Hemat., 6,1978. 60. Kovacs P, Bruch C and Fliedner TM: Colony formation by canine haemopoietic cells "in vitro". Inhibition by polymorphonuclear leukocytes. Acta Haematol., 56: 107, 1976. 61. Kurnick JE and Robinson WA: Colony growth in human peripheral white blood cells in vitro. Blood, 37: 136,1971. 62. Lajtha LG, Pozzi LV, Schofield R and Fox M: Kinetic properties of haematopoietic stem cell. Cell. Tissue Kinet., 2: 39,1969. 63. Lawler SD: The cytogenetics of chronic granulocytic leukaemia. Clin. Haemat., 6: 55, 1977. 64. Leif RC and Vinograd J: The distribution of buoyant density of human erythrocytes in bovine albumin solutions. Proc. Nat. Acad. Sci. (Wash.), 51: 520,1964. 65. Lord BI: Growth kinetics of CFU of different densities. Annual report of the Paterson laboratories (Christie Hospital and Holt Radium Institute, Manchester) 102,1970. 66. Lowenthal RM, Park DS, Goldman JM, Th'ng KH, Hill RS and Whyte G: The cryopre- servation of leukaemia cells: morphological and functional changes. Brit. J. Haemat., 34: 105,1976. 67. MacPherson JL, Nusbacher J and Bennett JM: The acquisition of granulocytes by leuko- pheresis in normal and corticosteroid-stimulated donors. Transfusion, 16: 221, 1976. 68. Malinin TI, Perry VP, Kerby CC, Dolan MF: Peripheral leukocyte infusion into lethally irradiated guinea pigs. Blood, 25: 693,1965. 69. Maloney MA and Patt HM: Migration of cells from shielded to irradiated marrow. Blood, 39: 804,1972. 70. McCredie KB, Freireich EJ, Hersh EM, Curtis JE and Kaizer H: Early bone-marrow re- covery after chemotherapy following the transfusion of peripheral blood leukocytes in identical twins. Proc. Amer. Ass. Cancer Res., 11:50,1970. 71. McCredie KB, Hersh EM and Freireich EJ: Cells capable of colony formation in the peri- pheral blood of man. Science, 171: 293,1971. 72. McCredie KB, Freireich EJ, Hesch JP and Vallejos C: Increased granulocyte collection with the blood cell separator and the addition of etiocholanolone and hydroxyethyl starch. Transfusion, 14: 357,1974. 73. McCulloch EA and Till JE: The sensitivity of cells from normal bone-marrow to gamma radiation in vitro and in vivo. Radiat. Res., 16: 822,1962. 74. McCulloch EA: Les clones de cellules hematopoietiques "in vivo". Rev. Franc. Etudes Clin. Biol., 8: 15,1963. 75. McCulloch EA and Till JE: Proliferation of hemopoietic colony forming cells transplanted into irradiated mice. Radiat. Res., 22: 383,1964. 36

76. Messner HA, Till JE and McCulloch EA: Interacting cell populations affecting granulo- poietic colony formation by normal and leukcmic marrow cells. Blood, 42: 701, 1973. 77. Messner HA, Till JE and McCulloch EA: Specificity of interacting population affecting granulopoiesis in culture. Blood, 44: 671,1974. 78. Metcalf D, Bradley TR and Robinson W: Analysis of colonies developing in vitro from mouse bone-marrow cells stimulated by kidney feeder layers or leukemie serum. J. Cell. Physiol., 69: 93,1967. 79. Metcalf D: Transformation of granulocytes to macrophages in bone-marrow colonies in vitro. J. Cell. Physiol., 77: 277,1971. 80. Metcalf D: Depressed response of the granulocyte-macrophage system to bacterial anti- gens following pre-immunization. , 26: 1115, 1974. 81. Metcalf D: Ilemopoictic colonies. Springer Verlag, Berlin, Heidelberg, New York, 1977. 82. Micklem HS: Effect of phytohemagglutinin-H (PIIA) on the spleen colony-forming capa- city of mouse lymph node and blood cells. Transplantation, 4: 732, 1966. 83. Miller RG and Phillips RA: Separation of cells by velocity sedimentation. J. Cell. Physiol., 73: 191,1969. 84. Miller RG, Gorczynski RM, Laflcur L, MacDonald HR and Philips RA: Cell separation analysis of B-and T-lymphocytc differentiation. Transpl. Rev., 25: 59,1975. 85. Mishler JM, Hadlock DC, Fortuny IE, et al.: Increased efficiency of leukocyte collection by the addition of hydroxyethyl starch to the continuous flow centrifuge. Blood, 44: 571,1974. 86. Moffat DJ, Rosse C and Yoffey JM: Identity of the haemopoietic stem cell. Lancet II, 547, 1967. 87. Monette FC, Morse BS, Howard D, Niskanen E and Stohlman F: Hemopoietic stem cell proliferation and migration following bordetella pertussis vaccine. Cell. Tissue Kinet., 5: 121,1972. 88. Moore MAS, Williams N and Metcalf D: Purification and characterization of in vitro colony forming cells in monkey hematopoietic tissue. J. Cell. Physiol., 79: 283, 1972. 89. Naparstek E, Halpern J and Hershko C: Time-related increase in leukocyte yields by continuous flow centrifugation (CFC) leukopheresis. Scand. J. Haem., 19: 79, 1977. 90. Nelson B, Calvo W, Fliedner TM, et al.: The repopulation of lymph-nodes of dogs after 1200 R whole-body X-irradiation and intravenous administration of mononuclear blood leukocytes. Amer. J. Pathol., 84: 259, 1976. 91. Netzel B, Brehm G, Gross-Wilde H, et al.: Enrichment of large quantities of granulocytic progenitor cells from human peripheral blood. Exp. Hemat., 2: 275, 1974. 92. Nothdurft W, Bruch C, Fliedner TM and Riiber E: Studies on the regeneration of the CFU-c population in blood and bone-marrow of lethally irradiated dogs after autologous transfusion of cryopreserved mononuclear blood cells. Scand. J. Haem., 19: 470, 1977. 93. Nowell PC, Hirsch BE, Fox DH and Wilson DB: Evidence for the existence of multipoten- tial lymphohematopoietic stem cells in the adult rat. J. Cell. Physiol., 75: 151, 1970. 94. Ogawa M, MacEachern MD, Wilson JM, et al.: Erythropoietic precursors in human umbi- lical cord blood. Blood,48: 980,1976. 95. Ohl S, Schaefer UW, Boecker WR, Kalden JR and Peter Ml: Human peripheral blood "null-lyrnphocytes", a highly enriched pool for granulocytic stem cells (CFU-c). Z. Immun. Forsch., 152:423,1977. 96. Paran M and Sachs L: The continuous requirement for inducer for the development of macrophage and granulocyte colonies. J. Cell. Physiol., 72: 247, 1968. 97. Pertoft H: Separation of blood cells using colloidal silicapolysaccharidc gradients. Exp. Cell. Res., 46: 621,1967. 98. Pertoft H, Back O, Lindahl-Kiesling K: Separation of various blood cells in colloidal silica- polyvinyl-pyrrolidone gradients. Exp. Cell. Res., 50: 355, 1968. 99. Peterson EA and Evans WH: Separation of bone marrow cells by sedimentation at unit gravity. Nature, 214: 824,1967. 37

100. Petrov RV, Kaitov RM, Aleinikova NV and Gulak LV: Factors controlling stem cell recii- culation. III. Effect of the thymus on the migration and differentiation of hemopoietic stem cells. Blood, 49: 865,1977. 101. Pike B and Robinson WA: Bone-marrow colony growth in agar gel. J. Cell. Physiol., 76: 77,1970. 102. Pluznik DH and Sachs L: The cloning of normal "mast" cells in tissue culture. J. Cell. Comp. Physiol., 66: 319,1965. 103. Pluznik DH and Sachs L: The induction of clones of normal "mast" cells by a substance in conditioned medium. Exp. Cell. Res., 43: 553,1966. 104. Porteous DD and Lajtha LG: On stem cell recovery after irradiation. Brit. J. Haemal., 12: 177,1966. 105. Quesenberry PJ, Morley A, Ryan M, Howard D and Stohlman F, Jr.: The effect of endo- toxin on murine stem cells. J. Cell. Physiol., 82: 239,1973. 106. Ragab AH, Gilkerson E, and Choi SC: The cry ©preservation of colony-forming cells from bone marrow of children with acute lymphoblastic leukaemia. Cancer Res., 34: 942,1974. 107. Raidt DJ, MisheU RI and Dutton RW: Cellular events in the immune response. Analysis and in vitro response of mouse spleen cell populations separated by differential flotation in albumin gradients. J. Exp. Med., 128: 681,1968. 108. Rastrick JH: A method for the positive identification of erythropoietic cells in chromo- some preparations of bone marrow. Brit. J. Haemat., 16: 185, 1969. 109. Richman CM and Chess L: Concentration of human peripheral blood stem cells in the null cell fraction. Blood, 36: 1011,1975. 110. Richman CM, Weiner RS and Yankee RA: Increase in circulating stem cells following chemotherapy in man. Blood, 47: 1301,1976. 111. Richman CM, Chess L and Yankee RA: Purification and characterization of granulocytic progenitor cells (CFU-c) from human peripheral blood using immunologic surface markers. Blood, 51: 1,1978. 112. Rickard KA, Rencricca NJ, Shadduck RK, et al.: Myeloid stem cell kinetics during ery- thropoietic stress. Brit. J. Haemat., 20: 537,1971. 113. Rickard KA, Morley A, Howard D and Stohlman F. Jr: The in vitro colony forming cell and the response to neutropenia. Blood, 37: 6,1971. 114. Robinson CV, Commeford SL and Bateman JL: Migration of hemopoietic stem ceils from the tail of the mouse. Radiat. Res., 25: 234,1965. 115. Ross GD, Rabellino EM, Polley MJ and Grey HM: Combined studies of complement receptor and surface immunoglobulin-bearing cells and sheep erythrocyte rosette-forming cells in normal and leukemic human lymphocytes. J. Clin. Invest., 52: 377,1973. 116. Ross WM, Bruck C, Fliedner TM, Herbst E, et al.: Factors influencing the yield of blood CFU-c during continuous flow cell separation in dogs. In leukocytes: separation, collec- tion and transfusion. Eds. Goldman JM, Lowenthal RM, Academic press, London, 261, 1975. 117. Ross WM, Fliedner TM, Korbling M and Nothdurft WH: Mobilization of haemopoietic stem cells by dextran sulphate into peripheral blood (dogs) (Abstract) Exp. Hemat., 4, Suppl. 52, 1976. 118. Ross WM, Calvo S, Fliedner TM, Korbling M and Nothdurft WH: Hemopoietic blood stem cell mobilization by dextran sulphate. Abstract. Suppl. No. 2, 13, Exp. Hemat. Vol. 5,1977. 119. Rubin SH and Cowan DH: Assay of granulocytic progenitor cells in human peripheral blood. Exp. Hemat., 1: 127,1973. 120. Sarpel S, Harvath L, and Epstein R: Peripheral blood stem cell separation, preservation and marrow repopulating potential in dogs. Abstract. Suppl. No. 3, 22, Exp. Hemat., 6,1978. 38

121. Schofield R: A comparative study of the repopulating potential of grafts frim various haemopoietic sources: CFU repopulation. Cell. Tissue Kinet., 3: 119,1970. 122. Schwarzenberg L, Mathe G, Amid JL, et al.: Study of factors determining the usefulness and complications of leukocyte transfusions. Amer. J. Med., 43: 206,1967. 123. Shortman K: The separation of different cell classes from Iymphoid organs. II. The purifi- cation and analysis of lymphocyte populations by equilibrium density gradient centrifu- gation. Aust. J. Exp. Biol. Med. Sci., 46: 375,1968. 124. Shortman K and Szenberg A: The size and density distribution of flow blood lymphocytes initiating a graft-versus-host reaction. Aust. J. Exp. Biol. Med. Sci., 47: 1,1969. 125. Shortman K: In "The separation of haemopoietic cell suspensions", ed. Van Bekkum DW and Dicke KA, Rijswijk, The Netherlands, Radiobiological Institute TNO, 1970. 126. Shortman K: Physical procedures for the separation of animal cells. Ann. Rev. Biophys. Bio-engineer, 1:93,1972. 127. Siminovitch L, McCulloch EA and Till JE: The distribution of colony forming cells among spleen colonies. J. Cell. Comp. Physiol., 62: 327,1963. 128. Storb R, Epstein RB and Thomas ED: Marrow repopulating ability of peripheral blood cells compared to thoracic duct cells. Blood, 32: 662,1968. 129. Storb R, Epstein RB, Le Blond RF, Rudolph RH and Thomas ED: Transplantation of allo- geneic canine bone marrow stored at -80 C in dimethyl sulfoxide. Blood, 33: 918,1969. 130. Storb R, Epstein RB and Thomas ED: Hemopoietic grafts after irradiation and buffy coat cell transfusions. Paris Editions du Centre National de la Recherche Scientifique, 61,1970. 131. Storb R, Rudolph RH and Kolb HJ: Marrow grafts between DL-A matched canine litter- mates. Transplantation, 15: 92,1973. 132. Storb R, Graham TC, Epstein RB, Sale GE and Thomas ED: Demonstration of hemopoie- tic stem cells in the peripheral blood of baboons by cross circulation. Blood, 50, 537, 1977. 133. Tebbi K, Rubin S, Cowan DH and McCulloch EA: A comparison of granulopoiesis in cul- ture from blood and marrow cells of non-leukemic individuals and patients with, acute leukemia. Blood, 48: 235,1976. 134. Thomas ED, Storb R, and Clift RA: Bone-marrow transplantation (first of two parts). The New Engl. J. Med., 292: 832,1975. 135. Till JE and McCulloch EA: A direct measurement of the radiation sensitivity of normal mouse bone-marrow cells. Radiot. Res., \4: 213,1961. 136. Tobias JS and Tattersal MHN: Autologtus marrow support and intensive chemotherapy in cancer patients. Eur. J. Cancer, 12: 1,1976. 137. Trentin JJ, Wolf N, Cheng U, Fahlberg W, Weiss D and Bowhag R: Antibody production by mice repopulated with limited numbers of clones of lymphoid cells precursors. J. Immunol., 98: 1326,1968. 138. Trobaugh FE and Lewis JP: Repopulating potential of blood and marrow. J. Clin. Invest., 43: 1306,1964. 139. Turner RWA, Siminovitch L, McCulloch EA and Till JE: Density gradient centrifugation of haemopoietic colony forming cells. J. Cell. Physiol., 69: 73,1967. 140. Van den Engh GJ and Golub ES: Antigenic differences between hemopoietic stem cells and myeloid progenitors. J. Exp. Med., 139: 1621,1974. 141. Van den Engh GJ, Mulder D, Williams N, and Bol S: Physical characterization of a sub- population of granulocyte/monocyte progenitor cells (CFU-c). In Experimental Hema- tology Today (Baum JS, Ledney GD, eds.), New York, Heidelberg, Berlin, 157, 1977. 142. Vassort F, Winterhole' »i, Frindel E and Tubiana M: Kinetic parameters of bone marrow stem cells using F~, vivo suicide by tritiated thymidine or by hydroxyurea. Blood, 41: 789,1973. 143. Vos O, Buurman WA, and Ploemacher RE: Mobilization of haemopoietic stem cells (CFU) into the peripheral blood of the mouse; effects of endotoxin and other compounds. Cell. Tissue Kinet., 5: 467,1972. 39

144. Weiner RS, Richman CM and Yankee RA: Semicontinuous flow centrifugation for the pheresis of immunocompetent cells and stem cells. Blood, 49: 391,1977. 145. Wigzell H and Andersson B: Isolation of lymphoid cells with active surface receptor sites. Ann. Rev. Microbiol., 25: 291,1971. 146. Williams NT and Van den Engh GJ: Separation of subpopulating of in vitro colony forming cells from mouse bone marrow by equilibrium density centrifugation. J.Cell. Physiol. 86: 237,1975. 147. Worton RG, McCulloch EA and Till JE: Physical separation of hemopoietic stem cells dif- ferent in their capacity for self-renewal. J. Exp. Med., 130: 91,1969. 148. Worton RG, McCulloch EA and Till JE: Physical separation of hemopoietic stem cells from cells forming colonies in culture. J. Cell. Physiol., 74: 171,1969. 149. Wu AM, Till JE, Siminovitch L, and McCulloch EA: A cytological study of the capacity for differentiation of normal hemopoietic colony forming cells. J. Cell. Physiol., 69: 177,1967. 150. Wu AM, Siminovitch L, Till JE and McCulloch EA: Evidence for a relationship between mouse hemopoietic stem cells and cells forming colonies in culture. Proc. Nat. Acad. Sci. USA.59: 1209,1968. 151. Zander ER, Templeton J, Armour V, Verma DA, Spitzer G and Dicke KA: mobilization by pyran (NSC 46015) in dogs and mice. Abstract. Suppl. No. 3, 34, Exp. Hemat., 6,1978. 152. Zucker RM and Cassen B: The separation of normal human leukocytes by density and classification by size. Blood, 34: 591,1969. 40

Chapter 2

PURIFICATION OF CFU-C FROM HUMAN PERIPHERAL BLOOD BY A TWO-STEP DENSITY SEPARATION TECHNIQUE

2.1 INTRODUCTION

Since the proportion of CFU-c in human peripheral blood is small relative to the other cells (15, 49), purification is an important step in the study of these cells and their interaction with other mononuclear cell (MNC) subpopulations. The combined use of density separation techniques and membrane surface markers offers a logical approach to purifying CFU-c and studying the influence of various MNC on the proliferative capacity of CFU-c. Several investigators, using multi-step separation techniques, confirmed the prediction that the progenitor cells for the granulocytic series (CFU-c) are present in the null cell (non-T, non-B) population (3, 72, 82, 83). Nowadays Ficoll-Isopaque centrifugation is by far the most common technique for isolating MNC from human blood (10). In this technique buffy coat cells in medium are layered onto a mixture of Ficoll-Isopaque and centrifuged; the MNC concentrate at the interphase between the separating fluid and plasma; the erythrocytes and virtually all granulo- cytes sediment to the bottom of the tube. In most studies MNC yields of about 60% were obtained and since only a few MNC can be found in the pellet of the tube, the missing MNC are probably lost by adherence to the tube walls or discarded along with the diluted plasma, excess Ficoll-Isopaque, or washing fluids. Such cell losses can be partly prevented by centrifugation at higher speeds during the washing procedures, by addition of bovine serum albumin (BSA) to the washing fluid, and/or by using Ca++- and Mg++-free medium. Defibrination of the cell suspension should be avoided, because it causes depletion of MNC, in particular B-cells (28). Furthermore, B-cells tend to be more adherent than T-cells and can selectively stick to the tube walls during the washing procedures (36, 53). To judge the effi- ciency of the procedure, the total white cell count and the relative 41

contribution made by each of the leucocyte populations should be determined before centrifugation. It is now well recognized that although lymphocytes show little mor- phological heterogeneity, functional subpopulations do exist. One of the best examples of this functional heterogeneity was the demonstration of the existence of Thymus-dependent (T) and Bursa-dependent (B) lymphocytes (21, 28, 84, 92). In the last decade various subpopulations of lymphocytes have been identified by the presence or absence of particular cell surface structures. These surface markers serve the practical purpose of enabling one to visually recognize and physically separate living lymphocytes belonging to different subpopulations. For instance, cells bearing a particular surface receptor may attach to red blood cells (RBC) carrying the relevant receptor binding substance. This will then artificially modify their density and allow their separation from other cells by density centrifugation. In this way enrichment or depletion of cells of a particular type can be obtained. A short review of the different surface markers on various MNC is worthwile for a better understanding of the various cell separation techniques described in this chapter.

In the analysis of surface markers on lymphocytes it is essential to exclude or at least identify monocytes, since these cells can share markers (e.g.: SIg, IgG-Fc, and C3-receptors) with lymphocytes (96). it can be very difficult \o differentiate monocytes from lymphoid cells, both in light microscopy of fixed stained smears and in the cell suspensions commonly used for rosette and immunofluorescence testing. This can complicate the interpretation of these assays, unless vigorously controlled. The presence and intracellular localization of certain enzymes as demonstrated by cytochemical stains has been widely used as marker for different cell types. Very useful for monocyte identification is the a-naphtyl-butyrate-esterase stain technique (73). Monocytes are always strongly positive with a diffuse staining pattern of the cycoplasm. In lymphocytes the reaction shows 1-2 small dots (T-cells) or some scattered granules (stimulated B-cells). Acid-phosphatase staining is also heavily positive in monocytes, whereas T-cells again show paranuclear dots; unstimulated B-cells are negative. Another useful characteristic of monocytes is their ability to ingest small latex particles in the presence of serum or plasma (35). A drawback of this technique is that in the common version of the tests to detect 42 complement- and IgG-Fc receptors on lymphocytes, the monocytes within the rosettes cannot be easily identified. This problem can be surmounted by using tetramethylrhodamine- isothiocyanate (TRITC) labelled latex particles, which make the phago- cytic cells easily recognizable (35). Another approach is to remove the monocytes via iron-carbonyl treatment of the blood sample. Disadvan- tages of this method are significant reduction of the final lymphocyte yield, a selective loss of B-cells owing to their adherent properties (35) and, finally, incomplete removal of monocytes. Surface immunoglobulins (SIg). SIg have been convincingly shown to provide the receptor for antigens. Although immunoglobulins (Ig) are readily detected on B-lymphocytes (78), considerable debate has raged concerning their presence on T-cells (95). Ig, in particular IgG, has also been detected on non-B, non-T lymphoid cells (so-called "null cells"), if they had been precultured in vitro for several days (17). However, in man, the detection of SIg is by definition limited to cells of the B-lymphocyte line and has been demonstrated most commonly by direct immunofluorescence techniques (38, 57, 74, 80). It was also found that the Ig on many B-lymphocytes was loosely bound to the cell surfaces. These Ig (mainly of the IgG-class) could be eluted by incubation of the cells for a short period at 37°C in a medium devoid of human or heterologous Ig, followed by washing at 37°C, or by overnight culture. Once the IgG has been removed, it does not appear again on the same cells. Therefore, the most probable origin of the "heat-labile" IgG is the donor's plasma, from which it must have been adsorbed onto the cell surfaces. The 37°C incubation has no effect if the washing has been carried out at 4°C, which suggests a reassociation of eluted IgG with the cells at lower temperatures. Other techniques for demonstrating SIg have been described. Anti- sera can be labelled with peroxidase to detect SIg (1), but also cytoxicity tests (98) and autoradiography are used (101). Furthermore, with the use of sensitized red blood cells (RBC), SIg can be detected by rosette techniques (43). These "B-cell rosettes" can be used to enrich or deplete cell suspensions for B-cells. Another technique to remove B-lymphocytes is a complement-dependent cytolysis with rabbit anti-human light chain sera (35), resulting in T-cell purification. The ability to adhere to glass and nylon surfaces is not only a property of monocytes: also B-cells will adhere to such surfaces (14), while most T-cells do not adhere (32). Finally B-cells can be identified by the pres- ence of receptors for C3, Epstein-Barr virus (EBV) (52) and macaca speciosa RBC (76). 43

Affinity to sheep red blood cells (SRBC). Brain et al. (11), Co wibs et al. (24), and Lay et al. (60) observed a large proportion of peripheral MNC, which formed rosettes with SRBC, independent of the presence of antibodies against SRBC on the cells or in the serum. The recognition that such cells were T-lymphocytes followed quickly (105); rosette tech- niques for detecting these erythrocyte (E) receptor-bearing eel1" were reported (51) and subsequently widely used in the study of immunolo- gical disorders. Some technical alterations in the preparation of the SRBC, such as pretreatment with neuraminidase, may stabilize the rosettes formed and allow more distinct separation in density gradients (97). Another method to stabilize rosette formation with T-cells is pretreatment of the SRBC with 2 aminoethyl isothiouronium bromide hydrobromide (AET). This latter method, however, also gives a higher affinity of AET-pretreated SRBC for SIg-positive cells (35). Using different techniques and modifications of the E-rosette test, most laboratories have constantly reported T-cell percentages of 60-70% (6,56) in human peripheral blood. Affinity to mouse erythrocytes (MRBC). MRBC have been shown to form clusters around human B-lymphocytes (39, 40, 93). The cells which bind to MRBC are generally accepted to be a B-lymphocyte subpopula- tion (41). They do not appear to form rosettes around monocytes and granulocytes and their capability to recognize cells is independent of SIg, Fc- and C3-receptors on these cells (41). The usefulness of this technique lies mainly in the investigation of monoclonal B-lymphocytic expansions, such as chronic lymphocytic leukaemia (75). The number of MRBC rosette forming B-cells in normal individuals is reported to be between 1 and 10% (5, 41, 75, 93). Receptors for the Fc-fragment of igG (IgG-Fc). White blood cells of different origin have receptors which can bind to the Fc-portion of IgG- molecules, immune-complexes or aggregated IgG (25, 26). Monocytes can form rosettes with IgG coated RBC (48), but promonocytes, gra- I nulocytes, eosinophils, and mastcells are also capable of binding IgG via their Fc-receptor (42, 48). Certain lymphoid cells can also bind the Fc- fragment of IgG-molecules (4, 25, 26, 31, 94). Originally, all such cells were generally regarded as B-cells, but Fc- receptors for IgG have also been detected on T-cells and null cells (88). That Fc-receptors for IgG can be found on T-cells is in accordance with observations done in guinea-pigs and mice (9, 106). IgG-Fc receptors have been demonstrated with different techniques. 44

Dickler and Kunkel (25) described the use of heat aggregated human IgG conjugated to fluorochromes, which is probably the most sensitive system. Other workers have employed antigen-antibody complexes in which one member of the complex was conjugated to fluorescein (30, 102). Rosette formation has been effectively employed by Haegert et al. (43), who used ox-RBC coated with IgG antibody (A) to ox-RBC (E) as the indicator (so-called EA-rosette technique) (44). Bach et al. (2) described a rosette test which employed human (Rh) D+ RBC coated with human anti-Rh-serum. With these techniques 10-20% of the lym- phoid cells were reported to be positive (1, 31, 32, 54). However, the percentage of cells with Fc-IgG receptors will greatly depend on the assay used. Almost complete overlap between cells bearing SIg and those binding homologous aggregated IgG was observed. For a rosette technique the most reliable results were obtained with ox-RBC, because they do not have spontaneous affinity for T-cells like SRBC (69). Receptors for the Fc-fragment of IgM. Using ox-RBC coated with rabbit anti-human IgM, IgM-Fc receptors were found on 50% of the T-cells, which do not overlap the IgG-Fc receptors on the same cells. IgM-Fc receptors are also found on a fraction of B-cells and possibly on null cells too (70). Complement receptors. Cells bearing receptors for cleaved fragments of the activated third complement include SIg bearing B-lymphocytes (8), monocytes, granulocytes (86), most null cells (77, 86), eosinophils, basophils and erythrocytes (14). Complement receptors are commonly detected by rosetting assays utilizing erythrocytes (E) coated with antibody (A) and complement (C) (7, 102), hence the term EAC-rosette. It is important that the antibody is of the IgM-class, to avoid any "background" antibody binding to the IgG-Fc receptor. Various RBC have been used as indicator for the EAC-rosette tech- nique (19), which can also be used for the isolation of C3 -receptor bearing cells by the use of density gradients. Mendes et al. (66) introduced the use of zymosan particles, which forms zymosan-complement (ZC) complexes after mixing with serum. ZC provides an indicator distinct from RBC, which both avoids the problem of spontaneous rosetting (1, 12) and facilitates the demonstra- tion of double rosettes (61). Other investigators have used nucleated avian RBC (20), RBC of dif- 45 ferent size (91),orfluorochrome-labelled RBC (76) for the formation of mixed rosettes, with which C3-receptors and other membrane surface markers could simultaneously be detected. In summary, the domain of lymphocytes (monocytes excluded) is divided into three major parts: the T-, B- and null lymphocytes. It is generally accepted that SIg and affinity to SRBC are two markers for B-cells and T-cells respectively, both with reasonable sensitivity. The Fc-receptor for IgM (IgM-Fc) occurs mainly on T-lymphocytes, but also on B-cells. It is as yet unknown if they occur on null cells. The demonstration of IgG-Fc receptors on lymphocytes depends on the technique used. If aggregated IgG is used as the marker system, then IgG-Fc receptor occurs on a large fraction of B-cells, null cells and on some T-lymphocytes. Complement receptors are distributed on most B-cells and null cells, and maybe also in very small proportions on T-cells. In this chapter a rapid and reproducible method is described by which a null cell fraction, depleted of monocytes, T-cells and B-cells is ob- tained by combining density centrifugation with simultaneous sedimen- tation of various rosettes. This null cell fraction proved to be highly enriched for CFU-c.

2.2 MATERIALS AND METHODS

2.2.1 Isolation of leucocytes and mononuclear cells by centrifugation techniques; general principles 440 ml blood was collected from healthy volunteers in 60 ml ACD [2,77c (w/v) anhydrous disodium citrate and 2.3% (w/v) anhydrous glucose]. After centrifugation (1,200 g, 8 min., 20°C) plasma and cells were separ- ated and 50 ml of the upper layer of the packed cells was collected (= buffy coat). This buffy coat was diluted to 200 ml with RPMI-medium (Flow Labs., Irvine, Ayrshire, Scotland). All media were supplemented with 100 ng streptomycin and 100 units penicillin/ml. To obtain mono- nuclear cells (MNC) 25 ml buffy coat was layered onto a 10 ml Ficoll- Isopaque density gradient (ds. 1.077 g/ml) and centrifuged for 20 min. (1,000 g, 20°C). The Ficoll-Isopaque solutions were prepared and sterilized at the Department of Pharmacy, University Hospital Leiden, according to B^yum (10). All cell separations in this study were performed with a Sorvall RC-3 centrifuge. The MNC in the interphase were collected by means of a Pasteur pipette and washed (10 min., 450 g, 20°C) three 46

times with RPMI-medium using a Sorvall GLC-1 centrifuge. Contamin- ating red blood cells (RBC) in the pellet were eliminated by isotonic lysis with 155 mM NH4C1 -10 mM KHCO3 - 0.1 mM EDTA - buffer (pH 7.4) for 10 min. at 0°C, according to Roos et al. (85).

2.2.2 Isolation of leucocytes by methylcellulose sedimentation Buffy coat cells were collected from 440 ml ACD-blood and diluted to 200 ml with RPMI-medium and mixed with 10 ml methylcellulose 2% (Fluka AG, Buchs, Switzerland). The cell suspension was divided into several 18 ml polystyrene tubes (Cat no. 3026, Falcon Plastics, Oxnard, Cal., U.S.A.). These tubes were settled for 30 min: at 20°C in a 45 degree position. The cell-rich plasma was collected and washed twice with RPMI- medium (10 min., 450 g, 20°C).

2.2.3 Preparation of monocyte-free lymphocyte suspensions To remove phagocytic cells, 50 ml buffy coat cells were diluted to 100 ml in RPMI-medium and incubated with 500 mg sterile carbonyl-iron powder (grade SF, Aristofarma, Delft, The Netherlands) in Erlenmeyer glass flasks (45 min. at 37°C, with continuous agitation) in a volume of 25 ml per flask. Thereafter a sterile magnet was added and the cell sus- pension was shaken for another 5 min. The cells were collected and sub- sequently diluted with 100 ml RPMI-medium. 25 ml of this cell suspen- sion was layered onto 10 ml Ficoll-Isopaque (ds. 1.077 g/ml) and the lymphocytes in the interphase were collected after centrifugation (1,000 g, 20 min., 20°C). Another method to obtain monocyte-free leucocyte suspensions was as follows: after leucocyte collection by methylcellulose sedimentation, incubation with carbonyl-iron and 5 min. shaking with a sterile magnet added, the cell suspension was carefully poured off.

2.2.4 Identification of B-lymphocytes with direct immunofluorescence on surface immunoglobulins (SIg) 1 x 106 plasma-free MNC were pelleted in a small plastic tube and resus- pended in 100 ^1 of a 1:4 dilution of polyspecific fluorescein isothio- cyanate (FITC)-conjugated goat anti-human F(ab) preparation (Nordic Pharmaceuticals, Tilburg, The Netherlands). After incubation (30 min., 20°C) the cells were washed three times with phosphate buffered saline 47

(PBS). The final pellet was resuspended on a microscopic slide into one drop of glycerol, containing 10% PBS (pH 7.2), and then covered with a cover-glass and sealed with paraffin. The percentage of SIg-bearing cells was determined by counting 200 leucocytes with a Leitz orthoplan fluorescence microscope. MNC, which could be recognized as monocy tes using phase contrast optics, were sub- stracted from the total percentage of SIg-bearing cells.

2.2.5 Identification of B-lymphocytes using goat anti-human Fab (GahuFab) coated sheep red blood cells (SRBC); B-lymphocyte rosette (= B-RFC) SRBC were obtained from the National Institute for Public Health (R.I.V., Bilthoven, The Netherlands) and stored at 4°C in Alsevers solu- tion. Before use, the SRBC were washed twice with PBS and once with NaCl 0.9%. The coating of the SRBC with GahuFab was done according to a modification of the technique described by Giuliano et al. (34): 1) 0.1 ml SRBC containing 10 x 108 cells, to which was added 2) 0.1 ml CrCl3; 0.38 M; 1:100 diluted in NaCl 0.9%, and 3) 0.1 ml GahuFab (Nordic Pharmaceuticals, Tilburg, The Netherlands); 1:100 diluted in NaCl 0.9%. 1), 2) and 3) were prepared as fresh as possible and added to each other sequentially. After 15 min. of incubation at 20°C, the SRBC were cen- trifuged (10 min., 400 g) and after washing [once in NaCl 0.9%, and twice in Hank's balanced salt solution (HBSS, Flow Lab).] they were resus- pended in HBSS up to a cell density of 8 x 108 /ml. Rosettes were formed by incubating 0.1 ml of lymphocytes (2.5 x 106/ml) with 0.1 ml SRBC (8 x 107/ml). This mixture was then centri- fuged (5 min., 150 g, 4°C) and the pellet carefully resuspended. The percentage of B-RFC was determined by counting 200 cells in a haemo- cytometer after addition of 1 drop Brilliant cresyl blue 1% (1:1 diluted in PBS). Only MNC binding 3 or more GahuFab-coated SRBC were scored as rosettes.

2.2.6 Identification of mouse-erythrocyte (MRBC) rosette forming B-cells (= M-RFC) MRBC were obtained by cardiac puncture of blood from NMRI-mice, 2-3 months old, and collected in Alsevers solution. Before use the 48

MRBC were washed three times with PBS and resuspended in RPMI- medium, supplemented with 0.5% bovine serum albumin (BSA, Poviet Producten NV., Amsterdam, The Netherlands). Rosettes were formed by incubating lymphocytes and MRBC in a 1:50 ratio in RPMI-medium, containing 10% foetal calf serum (FCS) (Micro- biological Associates, Bethesda, Md., U.S.A.). The mixture was then centrifuged for 5 min. (200 g, 20°C) and further incubated for 1 hour at 0°C. Next the pellet was carefully resuspended and the percentage of M-RFC determined by counting 200 cells in a haemocytometer after addition of 1 drop Brilliant cresyl blue 1% (1:1 diluted in PBS). Only lymphocytes binding 3 or more MRBC were scored as rosettes.

2.2.7 Identification of IgG-Fc receptor-bearing cells with EA-rosette formation, using human red blood cells (HuRBC) coated with anti-D IgG (= EA, -RFC) Human O-Rh erythrocytes (HuRBC) were stored in Alsevers solution and washed three times with PBS before use. HuRBC were coated with anti-D IgG in the following way: - 0.2 ml anti-D IgG (CLB, Amsterdam, The Netherlands), - 10 x 108 HuRBC, in 1 ml RPMI-medium, - RPMI-medium added up to 4 ml (final dilution of anti-D IgG: 1:20*). The mixture was incubated for 30 min. at 37°C, then washed three times, and resuspended in 2 ml RPMI (5 x 108 HuRBC/ml). EAj-rosettes were made by mixing lymphocytes and coated HuRBC in a 1:25 ratio in RPMI + 10% FCS. The mixture was then centrifuged for 5 min. (150 g, 20°C) and further incubated for 1 hour at 20°C. Next the pellet was carefully resuspended and the percentage of EA,-RFC was determined by counting 200 cells in a haemocytometer after addition of 1 drop Brilliant cresyl blue 1% (1:1 diluted in PBS). Only MNC binding 3 or more HuRBC were scored as rosettes.

2.2.8 Identification of IgG-Fc receptor bearing cells with EA-rosette for- mation, using SRBC coated with rabbit anti-sheep IgG (= EA2 -RFC) SRBC were stored in Alsevers solution and washed three times with PBS

* The maximal subagglutinating concentration of anti-D IgG to allow rosette formation was determined with a direct Coombs test, using polyvalent anti-human globulin (CLB, Amster- dam, The Netherlands). 49

before use. SRBC were coated with rabbit anti-sheep IgG in the fol- lowing way: - 0.2 ml rabbit anti-sheep IgG; 1:20 diluted*, - 10 x 108 SRBC in 1 ml RPMI, - RPMI-medium added up to 4 ml (final dilution of rabbit anti-sheep IgG: 1:400**). This mixture was incubated for30min. at 37°C, then washed three times 8 and resuspended in 2 ml RPMI (5 x 10 SRBC/ml). EA2 -rosettes were made by mixing lymphocytes and coated SRBC in a 1:50 ratio in RPMI- medium + 10% FCS. The mixture was then centrifuged for 5 min. (150 g, 20°C) and further incubated for 1 hour at 20°C. Next the pellet was carefully resuspended and the percentage of EA2-RFC determined by counting 200 cells in a haemocytometer after addition of 1 drop Brilliant cresyl blue 1% (1:1 diluted in PBS). Only MNC binding 3 or more SRBC were scored as rosettes.

2.2.9 Identification of IgG-Fc receptor bearing cells with EA-rosette formation, using ox red blood cells (ORBC) coated with rabbit anti-ox IgG (= EA3-RFC) ORBC were stored in Alsevers solution and washed three times in PBS before use. ORBC were coated with rabbit anti-ox IgG in the following way: - 10 x 108 ORBC in 1 ml RPMI-medium, - 0.04 ml rabbit anti-ox IgG*, - RPMI-medium added up to 4 ml (final dilution of rabbit anti-ox IgG: 1:100***) This mixture was incubated for 30 min. at 37°C, then washed three times 8 and resuspended in 2 ml RPMI-medium (5 x 10 ORBC/ml). EA3-rosettes were formed by mixing lymphocytes and coated ORBC in a 1:50 ratio in RPMI + 10% FCS. The mixture was then centrifuged for 5 min. (150 g, 20°C) and further incubated for 1 hour at 20°C. Next the pellet was carefully resuspended and the percentage of EA3-RFC determined by counting 200 cells in a haemocytometer after addition of 1 drop Brilliant

* Kindly supplied by Dr. C.J.L.M. Meijer, Dept. of Pathology, University Hospital, Leiden, The Netherlands. ** Maximal subagglutinating concentration was determined with a haemagglutination test. *** Maximal subagglutinating concentration of rabbit anti-ox IgG was determined with a haemag- glutination test. 50

cresyl blue 1% (1:1 diluted in PBS). Only MNC binding 3 or more ORBC were scored as rosettes.

2.2.10 Identification of T-cells with E-rosette formation, using SRBC (= E-RFC) SRBC were stored in Alsevers solution at 4°C and washed three times with PBS before use. To improve rosette stability the SRBC were in- cubated with papain-L-cysteine HCl-solution, according to Wilson et al. (100). Papain was obtained from Sigma Chemical Comp. (St. Louis, Miss., U.S.A.), and L-cysteine HC1 from Merck (Darmstadt, Germany). A papain-L-cysteine HCl-solution was made at 37°C by mixing: - 12.5 ml L-cysteine HC1, - 3.6 ml PBS, - 1.25 ml Na2HPO4 (3.6%), - 0.18 ml papain (25 mg protein/ml) 1 ml of this solution was mixed with 1 ml SRBC in PBS and incubated for8min. at 37°C. After washing three times with PBS, the SRBC were resuspended in 5 ml RPMI, and after counting the cell density adjusted to 10 x 108/ml. E-rosettes (E-RFC) were made by mixing lymphocytes with SRBC in a 1:50 ratio in RPMI + 30% FCS (15 min., 20°C). The mixture was then centrifuged for 5 min. (150 g, 4°C) and further incubated for 1 hour at 4°C. Next the pellet was carefully resuspended and the percentage of E-RFC was determined by counting 200 cells in a haemocytometer, after addition of 1 drop Brilliant cresyl blue 1% (1:1 diluted in PBS). Only MNC binding 3 or more SRBC were scored as rosettes.

2.2.11 Separation of rosette forming cells from non-rosette forming cells For all rosettes or rosette combinations we used the following procedure: after incubating (1 hour, 4°C), the rosettes in the pellet were carefully resuspended. Subsequently 10 ml Ficoll-Isopaque (ds. 1.072 g/ml or ds. 1.077 g/ml, 4°C) was carefully layered underneath the rosette suspension with a Falcon 10 ml pipette. ^ After centrifugation (20 min., 1,000 g, 4°C) the non-rosetting cells were collected, washed three times in RPMI, and resuspended in a-MEM (a-modified Eagle's Minimum Essential Medium, Flow Lab.). MNC in the pellet were collected and the RBC eliminated by isotonic lysis. 51

2.2.12 Teciinique for simultaneous rosette formation During the separation procedures the following rosette combinations were used: 1) E-RFC + EA,-RFC 2) E-RFC + EAx-RFC + M-RFC 3) E-RFC + B-RFC 4) E-RFC + B-RFC + EA2-RFC In all combinations, E-RFC-formation was initially started and after incubation for 15 min. (at 20°C), the other RBC suspensions were added just before the 5 min. centrifugation step (150 g, 4°C). For all RBC sus- pensions, RPMI-medium was used, supplemented with 20% FCS. The following lymphocyte-RBC ratios were used: - E-RFC : 1:50 -EA.-RFC : 1:25 - EA2-RFC : 1:50 - M-RFC : 1:50 -B-RFC : 1:40 For all rosette combinations another 1 hour of incubation at 4°C in 50 ml round bottom glass tubes was used, in which a 25 ml rosette suspen- sion containing 50 x 106 MNC in RPMI + 20% FCS was processed.

2.2.13 Elimination of B-Iymphocytes and monocytes by nylon-wool filtration This was done according to the technique described by Julius et al. (55). Briefly, 0.6 g nylon-wool (Leukopak®, Fenwall Labs, Morton Grave, 111., U.S.A.) was packed to the 6 ml mark in a 20 ml plastic syringe (Mono- ject, Deland, Fla., U.S.A.). The syringes were sealed, autoclaved, and washed with 25 ml PBS and next with 25 ml RPMI + 10% FCS before use. After equilibration of the wet columns for 60 min. (37°C), 75-150 x 106 MNC in 2 ml RPMI + 10% FCS were added and washed into the nylon-wool column with 1 ml RPMI + 10% FCS. The column was then sealed and horizontally incubated for 45 min. at 37°C. Next, non-adherent cells were collected by dropwise elution with 25 ml warm (37°C) RPMI-medium + 10% FCS. Finally the eluate was centri- fuged (450 g, 10 min., 20°C), the cell pellet resuspended in 2 ml RPMI- medium and processed for further analysis. 52

2.2.14 Preparation of and cell processing with continuous Ficoll-Isopaque density gradients Continuous density gradients, isotonic with human serum (osmolarity 294 mOsm, pH 7.4) and ranging from ds. 1.055 g/ml at the top to ds. 1.095 g/ml at the bottom (at 4°C) were prepared from two stock solu- tions of Ficoll-Isopaque + 1% human albumin according to the technique described by Loos et al. (64). Ficoll (polysucrose raw 400,000) was purchased from Pharmacia, Uppsala, Sweden; the Isopaque (1,156 m Metrizoate-solution) from Nyegaard A/S, Oslo, Norway, and the human albumin solution was obtained from the Me'rieux S.A. Institute, Lyon, France. The stock solu- tions were sterilized by filtration (millipore filter, 0.22 ju) and stored at -20°C before use. Linear density gradients of 35 ml in 50 ml corex glass tubes were generated by continuously mixing low and high density Ficoll-Isopaque + 1% albumin solutions with the aid of a roller pump (Auto-analyzer, Mark I), and were immediately used for cell separation procedures. Before studying the density distribution of the various human MNC, the linearity of the gradient system was verified, for which two continuous density gradients were prepared (A and B). A long steel needle was lowered to the bottom of the gradient tubes and 16 fractions of 2.5 ml each were collected from both gradients. From the collected fractions the refractive index (RI) at 20°C was determined and plotted against the fraction number. Fraction 16 was collected from the bottom and therefore had the highest RI. A good linear correlation was found (Fig. 2.1) expressed by the equation Y = 1.3456 + 0.00148 X (r = 0.997, p< 0.05). The relation between density and RI was also determined in several linear gradients. For the gradient solutions used in our laboratory the following formula was calculated: Y = 37.790 + 0.2490 X, where Y = (density at 4°C - 1.000) x 103, and X = (RI at 20°C - 1.3400) x 104. (r = 0.997; p < 0.05). In order to prepare solutions of any desired density within the range of ds. 1.055 — 1.095 g/ml, the mixing ratio (a) of the stock solution was cal- culated with the equation: required density = a (1.095) + (1 -a) 1.055 (64). The required density was verified by measuring the RI at 20°C with an Atago refractometer (Atago, Japan). For cell processing 4.5 x 107 MNC suspended in 5 ml Ficoll-Isopaque (ds. 1.055 g/ml) were carefully layered on the top of the gradient and cen- trifuged for 20 min. (1,000 g, 4°C) with slow acceleration and decelera- 53

Refractive index

1.3700

1.3650 y = 1.3456 + 0,00148 x r= 0.997

1.3600

1.3550

1.3500

1 2 3 i 5 6 7 8 9 10 11 12 13 U 15 16 Fraction number

Fig. 2.1: Relationship between the refractive index and sample fraction (1 <—> 16), obtained from two linear Ficoll-Isopaque density gradients (A, B). Ordinatc: refractive index at 20°C Abscissa: fraction 1 <—* 16 of 2.5 ml each tion of the centrifuge. The cells were then harvested by carefully lowering a long steel needle to the bottom of the gradient tube and subsequently 10 fractions of 4 ml each were collected by pumping out the fluid at a rate of 1 ml/min. The density of each fraction was calculated from the RI of the sample and the MNC were processed for further studies. For each fraction, the number of a given cell type was expressed as percentage of the total number of that cell in all fractions.

2.2.15 Cell staining and counting Nucleated cells were counted in duplicate in a haemocytometer (Biifker) in Turk's-solution (0.01% cristal violet +1% acetic acid in saline). Slides 54 for differential counts were made by cytocentrifuge sedimentation, fixed in absolute methanol for 10 min. at 20°C and stained with May-Griinwald- Giemsa. The slides were then washed in the buffer and air-dried. Mono- cytes were counted by using a a-naphtyl-butyrate-esterase staining tech- nique, described by Ornstein et al. (73).

2.2.16 CFU-c culture technique This was done according to the technique described by Iscove et al. (49). For all experiments methylcellulose (Methocel-MC 4000, Fluka AG, Buchs, Switzerland) was used. The FCS (Microbiological Associates, Bethesda, Md., U.S.A.) was heat-inactivated (56°C, 30 min.) before use. The cells were plated in triplicate in quantities of 5 x 10s cells (macro- technique) or in six-fold in quantities of 2-5 x 104 cells per well (micro- technique).

After culturing in a CO2-incubator (5% CO2, 98% humidity) for 10 days, the colonies were scored using an inverted microscope (Zeiss, Jena, Germany) with a magnification of x 80. Cell aggregates of > 20 were scored as colonies. In this and the next few chapters, several ways to express CFU-c entities are used, which are summarized in Table 2.1.

Table 2.1: Summary of different CFU-c expressions

CFU-c number the mean (± s.e.m.) of scored CFU-c per 5xlOs (=A) or 5xl04 (=B) cells plated (A= cultured in triplicate; B= cultured in 6-fold) CFU-c absolute (number) A= mean cell yield (x 10 ) x mean CFU-c number x 2 B = mean cell yield (xlO6) x mean CFU-c number x 20 5xl0s CFU-c/cell ratio A= 1 mean CFU-c number

mean CFU-c number CFU-c purification _ mean CFU-c number in test suspension x 1 factor (x) * mean CFU-c number in starting suspension (starting suspension = x 1) • u «v\* CFU-c absolute number in test suspension CFU-c absolute number in starting suspension (starting suspension = 100)

* CFU-c for both test and starting suspension stated per 5xlO5 or 5xlO4 cells. 55

2.2.17 Statistical methods The arithmetic mean of samples are expressed with ± 1 standard error of the mean (s.e.m.). The differences between two observations were tested with the two-tailed Student's t-test for paired observations, un- less stated otherwise. Statistical significance was confirmed by a non- parametric rank test (Wilcoxon Mann Whitney). Only p-values of < 0.05 were considered as significant.

2.3 RESULTS

2.3.1 Linear density distribution of monocytes, T-cells, B-cells and CFU-c Buffy coat cells were collected from 500 ml ACD blood of 5 healthy blood donors, in which the mean number of nucleated cells was 1,153 (± 179.3) x 106. After Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation, a mean of 227 (± 33.0) x 106 MNC was obtained, representing 19.6% of the initial cell content. Thereafter, 40 x 106 MNC were layered on each of 5 linear density gradients and processed for further analysis. The whole procedure is schematically shown in Fig. 2.2. After centrifugation the mean cell yield was 56 ± 3.4%. In Fig. 2.3 the density profiles of the different MNC subpopulations of the 5 donors are plotted together.

40x106 MNC

(2O'iOOOg/2O°C) —• interphase MNC top density 1.055 g/ml

• bottom density 1.095 g/ml buffy coat FicoU-Isopaque pellet: linear density gradient ds 1.077 g ml - granulocytes - erythrocytes

Each fraction analysed for : - cell count auto - analyser pump: -T-celts 10 fractions of 4 ml each -Slg© cells - monocytes -CFu-c - refractive - index

Fig. 2.2: Technique for determining the linear density profile of peripheral blood MNC, after isolation from buffy coat cells with a Ficoll-Isopaque (ds. 1.077 g/ml) gradient. Fig. 2.3: Density distribution profile of all MNC (•), T-lymphocytes (•), B-lymphocytes (o), monocytes (*) and CFU-c (A), after MNC isolation from buffy coat cells with a Ficoll- Isopaque (ds. 1.077 g/ml) gradient. (N = 25 buffy coats).

80 80- 90-i All mononuclear cells - T-lymphocytes . monocytes 70 70- 70- A 60 60- 60-

50- 50- 50- » A A (0 (0- t 40 A • A A 30- 30- • • 30- * A A • • - • • A A 20 20- • • 20- *» - A 10 10- • • . • •• 10- A* A* 1 . • • *• • • * . •• • AA* A n ' • 0 •i ilftS** •* • • • * HI Ai^A AA A 1.050 1.060 1.070 1.090 1.090 1.050 1.060' 1.070 1.080 1.090 1.050 1.060 1.070 1.080 1.090 density (g/ml) density (g/ml) density (g/ml)

Mi as B - lymphocytes 5 70- CFu-c/5xX) 130-1 60 110- 50 o o 90 Ml A x A . ° •• 70 J o 30"! o L •i 0 o 50 20- ° „ o o o •• O O X 10 -i A I O Q O 10 A ^ A ft 1.050 1.060 1.070 1.0BO 1.090 1.050 1.060 1.070 1.080 1.090 density (g/ml) density (g/ml)

(". •. o,A, ^-abscissa: density in g/ml (4°C) (•i •» °. *)-ordinate: percentage of cell type, expressed as percentage of the total number of that cell type in all fractions (a)-ordinate: number of CFU-c/5 x 10s cells plated. 57

The distribution of all MNC was found well within the density range 1.055 to 1.095 g/ml, with a mean value of 1.071 ± 0.0014 g/ml. The T-cells were also distributed throughout the whole density range with a mean value of 1.075 ± 0.0012 g/ml. On the other hand, mono- cytes and 3-lymphocytes showed a distribution mainly in the light den- sity region from 1.055 to 1.080 g/ml, with a common mean value of 1.065 ±0.0008 g/ml. The distribution of theCFU-c was also found in the light density region from 1.055 to 1.075 g/ml, with a mean value of 1.063 ± 0.0010 g/ml. Because of the variability in colony-forming capacity, a rather hetero- geneous distribution was found. CFU-c situated in one and the same density position therefore showed a different colony-forming capacity. It is remarkable that although the MNC were initially separated on a density 1.077 g/ml gradient, cells were found in the region 1.077 1.095 g/ml, after linear density centrifugation. This could be the result of a sedimentation inhibition of high density MNC by cell crowding at tize medium Ficoll-Isopaque interphase or by an increase in cell density, caused by the initial Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation step.

2.3.2 linear density distribution of T-cells, B-cells and CFU-c after initial monocyte elimination In the next experiment the same parameters were analysed, but after initial removal of monocytes by carbonyl-iron incubation. This experi- ment was done to see whether the commonly used method for carbonyl- iron treatment could induce alterations in the density of MNC subpopul- ations. From 5 blood donors, buffy coats were obtained and after pre- incubation with carbonyl-iron and centrifugation on a density gradient (ds. 1.077 g/ml), a mean MNC yield of 215 (± 29.0) x 106 was obtained. The mean cell yield after linear density gradient centrifugation was 56 ± 4.2%. The distribution of all MNC (Fig. 2.4) was in the same den- sity region from 1.055 to 1.095 g/ml, but the mean density was clearly shifted to a higher value of 1.076 ± 0.0014 (p < 0.05). The distribution of the T-lymphocytes was basically unaltered, with a mean density of 1.076 ± 0.0021 g/ml. However, the B-cells and non-eliminated monocytes (a total of 1.5%) were clearly altered in their density: both had a scat- tered distribution in the high density region. The CFU-c, however, were not altered in their density profile and were still in the light density region, with a mean value of 1.065 ± 0.012 g/ml. Fig. 2.4: Density distribution profile of all MNC (•), T-lymphocytes (•), B-lymphocytes (o), monocytes (*) and CFU-c (A), after monocyte removal with carbonyl-iron incubation and centrifugation on a Ficoll-Isopaque (ds. 1.077 g/ml) gradient. (N = 5 buffy coats).

V. 80 All mononuclear cells T- lymphocytes monocytes 70 70-

SO- 60-

50 • * 50

40 to

30 30

20- 20

10

0 0 1.050 1.060 1.070 1.0*0 1.090 1.050 1.060 1.070 1.060 1.090 1.050 1.060 1.(770 roeo 1.090 density (g/ml) density (g/ml) density (g/ml)

OO

B-lymphocytes CFu-c/SxiO5 130 T

110

90

70-

50

30

10

1.050 1.060 1.070 I.OtO 1.090 1.050 1.060 1.070 1.0*0 1.090 density (g/ml) density (g/ml)

(•, •. °,A, ^)-abscissa: density in g/ml (4°C) (•» •> °, A)-ordinate: percentage of cell type, expressed as percentage of the total number of that cell type in all fractions (A)-ordinate: number of CFU-c/5 x 105 cells plated. 59

Hence the shift in the peak density of all MNC was mainly caused by an increase in the density of monocytes and B-cells, most probably induced by ingestion of iron particles and/or adhesion of iron particles to the cell membranes.

2.3.3 Comparison of two different densities (ds. 1.072 g/ml; ds. 1.077 g/ml) during the second separation step From the above-described analyses it is obvious that for CFU-c collec- tion a density-"cut"-point must be chosen somewhere between 1.065 and 1.077 g/ml. However, in this range there is substantial "contamina- tion" by other MNC, especially monocytes and B-cells. If the major goal of the separation technique is purification of CFU-c and removal of immunocompetent cells which are involved in cellular immunity (T-cells and monocytes), then it is possible, using a two-step density separation procedure, to remove monocytes and T-cells by carbonyl-iron incubation and E-RFC sedimentation respectively. There- fore the following two-step Ficoll-Isopaque sedimentation procedure was developed (Fig. 2.5). After initial incubation with carbonyl-iron, MNC were collected using a Ficoll-Isopaque gradient (ds. 1.077 g/ml) (step B). The density of 1.077 g/ml was chosen, because then all the CFU-c would collect in the interphase after centrifugation. In the second step the interphase cells were centrifuged on a Ficoll-Isopaque gradient of a 1.072 g/ml density, after previous E-RFC formation. The density of 1.072 g/ml for the second separation step was used because at this density the majority of the CFU-c would be in the interphase after centrifugation. Furthermore, the "contaminating" T-cells (which were not removed by E-RFC sedi- mentation) in the interphase would be slight, because on the ground of their density only, more cells would be centrifuged into the pellet. The disadvantage of the greater cell loss expected to occur with the 1.072 g/ml density, might be compensated by a higher CFU-c purification. The results of this two-step separation procedure will be presented in the next section (2.3.4), after testing this hypothesis. For this purpose, after monocyte depletion with carbonyl-iron during the first step, the interphase cells will be divided into a group (D) which is centrifuged in a density gradient of 1.072 g/ml, and a group (C) which is centrifuged in a density gradient of 1.077 g/ml. For both densities, E-RFC elimination is performed during this second centrifugation step. 60

—( r-» interphase MNC - suspension (20'i000g,20»C)

RcoU-lsopaque pellet: - granutocytes ds 1.077 g/ml - erythrocytes

(W',1000g,20»C) interphase MNC S«l

buffy coat carbonyl-iron Ficoll-lsopaque pellet: -granulocytes ds 1.077 g/ml - erythrocytes - monocytes - carbonyl-iron Formation of: E-RFC

4V1 interphase MNC analysed for: - cell count -T-cells - SIg © cells Ficoll- Isopaque pellet: - monocytes ds 1.072 g/ml T-cells - cFu-c

Fig. 2.5: Technique (B) for sequential monocyte and T-cell elimination, using a combination of density-gradient centrifugation and E-RFC sedimentation. (A) represents the control cell suspension after Ficoll-lsopaque (ds. 1.077 g/ml) centrifugation.

To test the effect of these two densities, MNC obtained from 5 buffy coats were analysed. The characterizations of the interphase cell suspen- sions obtained after the first step, are summarized in Table 2.II. The results obtained after the second separation step arc graphically expressed in Fig. 2.6. As can be seen, the total cell yield was lower after the light

Table 2.11: Cell suspension after carbonyl-iron incubation and subsequent centrifugation on a Ficoll-lsopaque (ds. 1.077 g/ml) gradient (N= 5 buffy coats)

Cell yield (xlO6)* 132±38.6 (=13.2%)** E-RFC% 64 ±2 EA,-RFC% 4.5±1.2 Slg(+) cells % 5.6+0.2 Monocytes % 1.610.4 CFU-c/5xlOs 23.6+4.6 CFU-c absolute 6,230 CFU-c ratio 1:21,186

* calculated from a corrected value of 1x10 nucleated cells in the initial buffy coat ** percentage calculated from the mean value 61

Fig. 2.6: Effect of two different densities during the second Ficoll-Isopaque gradient ccntrifu- gation step (with simultaneous E-RFC removal) on various cell types (N = 5 buffy coats).

cell yield E-RFC Slg®cells EA,-RFC monocy- CFu-c/ xiO6 % % % I tes % i5x105mnc

is -

10- - 100

-BO

5 - -to

- 20 p<0.05 NS NS. NS N.S. p<0.05 -

DC DC DC DC DC DC

Ordinate (left): cell yield (x 106) and % of the different cell types in the final interphase sus- pension Ordinate (right): numbers of CFU-c/5 x 10s cells plated Abscissa: D = Ficoll-Isopaque 1.072 g/ml C = Ficoll-Isopaque 1.077 g/ml Vertical bars represent mean ± s.e.m.

density (1.072 g/ml) separation, compared to ds. 1.077 g/ml (p < 0.05). This was also true for the percentages of T-cells, SIg(+) cells, IgG-Fc- receptor bearing cells and monocytes, but these differences were not sig- nificant. For the CFU-c, however, a significant (p < 0.05) higher value (5 x 10s MNC plated) was found. Since the absolute numbers of CFU-c in the suspensions D and C were equal (Table 2.Ill), the higher yield of CFU-c was relative and probably caused by a cumulative effect of the non- significant reduction in "contaminating cells" in the final cell suspensions. 62

Table 2. Ill: Two step cell separation technique; comparison of absolute cell numbers after the second centrifugation step, using Ficoll-Isopaque gradients of two different densities (N = 5 buffy coats)

ds. 1.072 g/ml (D) ds. 1.077 g/ml (C) Absolute difference

E-RFC (xlOa) 2.7 3.2 0.5 SIg(+) cells (xlOs) 7.6 10 2.4 s EArRFC (xlO ) 8.8 11 2.2 Monocytes (xlO5) 1.5 3.4 1.9 CFU-c absolute 1,463 1,439 24

Step 1 : monocyte removal with carbonyl-iron incubation and subsequent Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells Step 2 : E-RFC removal from the interphase cells obtained after step 1; using Ficoll-Isopaque gradients of two different densities (D and C)

2.3.4 Results of a two-step density gradient separation technique; elimination of monocytes and T-cells and subsequent CFU-c purification Having established that a density of 1.072 g/ml during the second Ficoll- Isopaque centrifugation step, in combination with E-RFC depletion, re- sulted in a more purified CFU-c suspension, this procedure was studied more thoroughly, the results of which are shown in Fig. 2.7. An analysis was done with MNC suspensions obtained from buffy coats of 24 normal blood donors. The mean number of nucleated cells in these initial buffy coats was 1,165 (± 80.6) x 106, of which 500 (± 32) x 106 were MNC. From all the 25 buffy coats a "control" cell suspension was made separately with a Ficoll-Isopaque (ds. 1.077 g/ml) gradient (suspen- sion A), the characteristics of which are summarized in Table 2.IV. After the first separation step (B) a mean of 239 (± 22.6) x 106 MNC were re- covered, which represented 47.8% of the MNC content in the buffy coats. After the second step (D) a mean of 23.6 (± 3.1) x 106 MNC were ob- tained, which represented 4.7% of the MNC content in the buffy coats. As can be seen from Fig. 2.7, the percentage of T-cells showed an initial increase after monocyte depletion from 61.2 ± 2.5 to 67.0 ± 1.5% (p < 0.05) and thereafter a drastic decrease to 3.5 ± 0.5% (p < 0.05). B-cells, on the other hand, showed the opposite phenomenon: a drop from 6.9 ± 0.2% to 5.8 ± 0.1% after monocyte elimination with car- bonyl-iron (NSD)*. *NSD = non significant different 63

Pig. 2.7: Results of a two-step cell separation procedure on CFU-c purification; removal of monocytes and T-cells. (N = 25 buffy coats).

' ' ' ' I / c cell yield E-RFC SIg©cells monocy- CFu-c/5x10° mnc 110 tes •110

100 -100

90 •90

60 80

70 •70

•60

5 - •so

4 - to

3 - 30

2 - 20

1 • 10

T 1 1—'—i 1 r- 0 A8DABDABDABDABD

Ordinate (left o, •) : cell yield, expressed as (mean) percentage of the MNC content in the initial buffy coats. (left •) : also refers to the percentage of E-RFC. Ordinate (right ©) : CFU-c/5 x 10s cells plated. Abscissa; A : "control" cell suspension after Ficoll-Isopaque (ds. 1.077 g/ml) centri- fugation. Step 1-»B : interphase cells after carbonyl-iron incubation and subsequent Ficoll-Iso- paque (ds. 1.077 g/ml) centrifugation of buffy coat cells. Step 2->D : interphase cells after E-RFC removal from suspension 8, using a ds. 1.072 g/ml Ficoll-Isopaque gradient. Vertical bars represent mean ± s.e.m.

After the second step the B-cells showed an enrichment to 11.0 ± 0.2% (p < 0.05). The monocytes displayed the same pattern: an initial decrease from 10.5 ± 0.3% to 1.5 ± 0.2% after the first step (p< 0.05) and a rise after E-RFC depletion to 3.3 ± 0.2% (p < 0.05). The CFU-c in the initial buffy coats had a concentration of 10.4 ± 6.4

Table 2.IV: 'Control' cell suspension obtained from the interphase after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells (N= 25 buffy coats)

Cell yield (xlO6)* 321 ±30.1 (=32.1%)** E-RFC% 61.2 ±2.5 EAi-RFC% 6.8 ±0.9 SIg(+)ceUs% 6.9 ±0.2 Monocytes % 10.5 ±0.3 CFU-c/5xlO5 26.1 ±10.3 CFU-c absolute 16,756 CFU-c ratio 1:19,157

* calculated from a corrected value of lxlO8 nucleated cells in the initial buffy coats ** percentage calculated from the mean value

1.9%/5 x 10s MNC plated and were found to be 26.1 ± 10.3 in the "control" cell suspension (A) (NSD). After monocyte elimination the CFU-c value dropped to 15.4 ± 3.0 (NSD). After the second separation step the CFU-c were ultimately enriched to 82.0 ± 22.0 (p < 0.05). This represented a mean purification factor, compared to the CFU-c concentration of the original buffy coat of 7.8 (p < 0.05). Although the percentage of "contaminating" T-cells and monocytes was acceptable, the percentage of SIg(+) cells was rather high in these final samples. That was because no special effort was made to remove this cell population. A summary of the different cell yields and CFU-c entities of the dif- ferent interphase cell suspensions obtained during this two-step cell separation procedure is given in Table 2.V. It can be seen that although the CFU-c were purified in the final cell suspension (D), a substantial loss of 84.4% in absolute CFU-c number was caused by these two cell separation steps.

2.3.5 Results of a three-step separation technique, including density gradients and nylon-wool filtration; removal of monocytes, T-cells, B-cells, and subsequent CFU-c purification In an attempt to reduce the enormous CFU-c loss as described in section 2.3.4, the effectiveness of nylon-wool filtration on CFU-purification was investigated. Nylon-wool is known to have adhesion properties for B-cells and monocytes, but is unable to bind T-cells. The whole procedure is schem- Table 2. V: Summary of cell yields and CFU-c entities of the different interphase cell suspensions, obtained during a two-step cell separation procedure (N = 25 buffy coats)

Difference Characteristics Buffy coat A B D between

MNC yield*.** (xlO6) 500+32 321±30.6 239122.6 23.6+3.1 A/B B/D A/D (=100%) -> -> -* |(=64%) (=47.8%) (=4.7%) NSD 4- i Total cell yield*1** (xlO6) - 1,000 321 + 30.6 239+22.6 23.613.1 NSD 1 i (=100%) -* |(=32%) (=23.9%) (=2.3%) CFU-c/5xlO5 - 10.4±1.9 26.1110.3 15.4+3 82+22 NSD t t -»• NSD CFU-c absolute - 20,800 16,756 7,361 3,870 - - - O\ CFU-c ratio - 1:48,076 1:19,157 1:32,467 1:6,097 - - - CFU-c purification (x) - 1 2.5 1.4 7.8 NSD t t ->• NSD CFU-c yield (%) - 100 80.5 35.3 18.6 - - - t or I = significant (p<0.05) difference NSD = non significant difference * = cell yield: corrected to l.OOOxlO6 cells in the initial buffy coats ** = cell yield (%): calculated from the mean values suspension A: interphase cell suspension after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells Step 1 -> suspension B: interphase cells after carbonyl-iron incubation and subsequent Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells Step 2 -> suspension D: interphase cells after E-RFC removal from suspension B, using a ds. 1.072 g/ml Ficoll-Isopaque gradient 66

atically shown in Fig. 2.8. In this experiment the initial monocyte re- moval with carbonyl-iron was omitted in order to investigate the monocyte removal capacity of nylon-wool more precisely. 1 3 (2O,W00g,2O»C) gg -* interphase MNC Formation of: e-RFC

buffy coat Ficott-lsopaque pellet: - granulocytes ds 1.077 g/mt - erythrocytes

<2O,WO0g,4'C) interphase MNC nylon-filter

Ficoll- Isopaque pellet: T-cells ds 1.077 g/ml

-» analysed for: - cell count -T-cells ^Stg © cells - monocytes -CFu-c Fig. 2.8: Technique for sequential T-cell, B-ceil and monocyte removal, using a combination of density gradient centrifugation, E-RFC sedimentation and nylon-wool filtration.

Fig. 2.9. presents the results of this three-step separation procedure, with buffy coat cells obtained from 6 blood donors. The cell yield after the last separation step was 1.5%. The T-lymphocytes were depleted from 47.4 ± 4.0% to 5.7 ± 1.0% (p < 0.05) after step C, and then en- riched to 16.8 ± 3.0% after step E (p < 0.05). This confirms the fact that T-lymphocytes do not adhere to nylon-wool. The B-lymphocytes showed the opposite phenomenon: an initial I value of 7.2 ± 1.1%, enrichment to 23.1 ± 1.3% after step C (p < 0.05) and finally depletion to 2.7 + 0.4% after step E (p < 0.05). The monocytes showed the same pattern: an initial value of 11.6 ± 5.2%, increase to 27.7 ± 6.9% after step C (p < 0.05) and decrease to 10.5 ± 1.4% after nylon-wool filtration (p < 0.05). Finally, the CFU-c showed a stepwise increase from 30«8 ± 7:4 to 55.0 ± 14.4 (p < 0.05) to 150.6 ± 20.4 (p < 0.05) per 5 x 10s MNC plated. For the CFU-c it was clearly shown that they pass through the nylon-wool column and do not adhere. 67

Fig. 2.9: Results of a three-step cell separation technique on CFU-c purification; removal of monocytes, T-cells and B-cells. (N = 6 buffy coats).

cell yield E-RFC SIg©cells monocy CFu-c/5x105 • I tes

110

100 -200

90 •180

80 -160

70 •1*0

60 -120

50- 100

40- 80

30- 60

20- to

10- 20

ACEACEACEACEACE

Ordinate (left •) : cell yield, expressed as (mean) percentage of the MNC content in the initial buffy coats. (left •) : also refers to the percentage of different cell types. Ordinate (right ©) : CFU-c/5 x 10s cells plated. Abscissa: Step 1->A : interphase cells after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation Step 2->C : interphase cells after E-RFC removal from suspension A, using a ds. 1.077 g/ml Ficoll-Isopaque gradient. Step 3-i-E : cell suspension after nylon-wool filtration of suspension C. Vertical bars represent mean ± s.e.m.

In Table 2.VI the results of a two-step cell separation procedure as described in section 2.3.4 and those of this three-step cell separation technique are compared, using Student's t-test for two observations. It can be seen that there were no differences in the two starting suspensions, 68 except for the T-lymphocyte percentages. The final suspensions were also comparable as far as the cell yields and CFU-c entities are concerned. However, the three-step cell separation technique resulted in a higher percentage of T-cells and monocytes (p < 0.05), but a lower percentage ofB-cells(p<0.05). If it is the purpose to eliminate especially the immunocompetent T-cells and monocytes, then the two-step cell separation procedure is a quicker approach.

Table 2.VI: Differences between cell yields and composition obtained after a two- and three-step cell separation procedure

Characteristics 2-step separation 3-step separation difference procedure procedure (N= 25 buffy coats) (N= 6 buffy coats)

Starting cell Interphase cells obtained after Ficoll-Isopaque suspension for (ds. 1.077 g/ml) centrifugation of 1x10 both procedures (corrected value) buffy coat cells (= 100%)

Cell yield*>**(xl0°) 321+30.1 (=32.1%) 232±37.8 (=23.2%) NSD E-RFC % 61.2+2.5 47.4±4 4 SIg(+)ceUs % 6.9 ±0.6 7.2+1.1 NSD Monocytes % 10.5 ±1.4 11.6 ±5.2 NSD CFU-c/5xlO5 26.1 ±10.3 30.8±7.4 NSD CFU-c absolute 16,756 14,291 - CFU-c ratio 1.19,157 1:16,233 CFU-c purification (x) 1 1 — CFU-c yield (%) 100 100 -

End cell suspension

Cell yield*'**(xl06) 23.6±3.1 (=2.3%) 15.4 ±1.9 (=1.5%) NSD E-RFC % 3.6±0.5 16.8±3 t SIg(+)cells % 11.1+0.2 2.7 ±0.4 Monocytes % 3.3+0.2 10.5±1.4 t CFU-c/5xl0s 82±22 150.6±20.4 NSD CFU-c absolute 3,870 4,638 - CFU-c ratio 1:6,097 1:3,320 - CFU-c purification(x) 3.1 4.8 NSD CFU-c yield (%) 23 32.4 - t or I = significant difference (p <0.05) NSD = non significant differences * cell yield: corrected to l.OOOxlO6 cells in the initial buffy coat ** cell yield (%): calculated from the mean values 69

2.3.6 The effect of simultaneous E-RFC, EA, -RFC and M-RFC removal during the second separation step on CFU-c purification In the initial experiments the main goal was removal of monocytes and T-lymphocytes and its subsequent effect on CFU-c purification. In the next experiment we also evaluated the effect of removal of B-lymphocyte (-subpopulations). In this experiment the effects on CFU-c entities were evaluated, if cells bearing receptors for M-RFC and IgG-Fc (measured by the EA,-rosette technique) had been removed. To keep the number of Ficoll-Isopaque centrifugation steps limited, simultaneous rosette formation and sedimentation during the second centrifugation step was performed. In the first step (Ficoll-Isopaque, ds. 1.077 g/ml) monocytes were eliminated by carbonyl-iron pre-incuba- tion. For the second centrifugation step (Ficoll-Isopaque ds. 1.072 g/ml) four different approaches were used: 1) Removal of E-RFC; 2) Removal of E-RFC + EA, -RFC; 3) Removal of E-RFC + M-RFC; and 4) Removal of E-RFC + M-RFC + EA, -RFC. Buffy coat suspensions from 6 normal blood donors were investigated. The cell yields obtained after the first and different second cell separa- tion steps are summarized in Table 2.VII.a. The effects on the different cell populations after the various centrifugation steps are summarized in Fig. 2.10. After the first cell separation step, the T-cells were 84.7 ± 4.5%, monocytes 2.8 ± 0.9%, EA,-RFC 3.7 ± 1.4%, M-RFC 1.8 ± 0.8, SIg(+) cells 4.5 + 1.2%, and CFU-c 1.2 ± 0.2 per 5 x 104 cells plated. These values are also presented at the top of Fig. 2.10, while the mean values are presented by dotted lines, except for the E-RFC. For the E-RFC, all second step combinations caused a significant elimination (p < 0.05) of E-RFC. For all the other MNC some differences were found, compared to the values obtained after the first cell separa- tion step (Table 2.VII.b). When for each cell type two second step I combinations were tested against each other with Student's t-test, some differences were found. For instance, for IgG-Fc receptor bearing cells, both combination 2 and 4 caused an elimination when compared to 1 (p < 0.05). For the M-RFC no significant alterations could be detected at all. The effect on the SIg(+) cells showed an increase when combination 2 was tested against 1 (p < 0.05). This was mainly due to removal of the T-lymphocytes and IgG-Fc positive cells. As far as the monocytes were concerned, the EA,-RFC technique seemed to be more sensitive for 70

Table 2. Vila: Cell yields obtained from the interphases during a two-step cell separation procedure, with removal of different rosette combinations during the second step (N= 6 buffy coats)

Cell yield (xlO6) Difference between combination***

Step 1 and all four Step 1 374+70.4* (=37.43 step 2 possibilities: P<0 .05

33.4±11.7 (=3.3%) 1-2 : p<0.05 2-3 : p<0.05 15 ±4.3 (=1.5%) Step 2 3-4 : p<0.05 23.5+5.6 (=2.3%) 1-3 : NSD 13.5+3.6 (=1.3%) 1-4 : NSD 2-4 : NSD

NSD = non significant difference * corrected value; cell yields are adjusted to 1,000x106 cells in the initial buffy coats ** percentages calculated from the mean values *** tested with Student's t-test for paired observations Step 1 -* moncyte removal from buffy coat cells with carbonyl-iron incubation and subsequent centrifugation on a Ficoll-Isopaque (ds. 1.077 g/ml) gradient Step 2-> centrifugation on a Ficoll-Isopaque (ds. 1.072 g/ml) with removal of (from interphase cells obtained after step 1): 1) E-RFC 2) E-RFC + EA.-RFC 3) E-RFC + M-RFC 4) E-RFC + EArRFC + M-RFC

IgG-Fc receptors on monocytes, because when combination 2 was compared to 1, there was a significant decrease in monocytes (p < 0.05). The effect on CFU-c purification was small. All four combinations did not differ from each other when tested as paired observations, but if separately compared to the CFU-c content after the first separation step, I all combinations resulted in significantly higher yields (p < 0.05). 71

Fig. 2.10: Results of a two-step cell separation procedure. Monocyte removal during the first Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation step after initial carbonyl-iron incuba- tion. Removal of E-RFC (1), E-RFC + EA,-RFC (2), E-RFC+ M-RFC (3) and E-RFC + EA,-RFC + M-RFC (4) during the second Ficoll-lsopaque (ds. 1.072 g/ml) centrifugation step. Cell characteristics after this step are presented in the top of the figure, while the mean values are also represented by the dotted lines (except for mean E-RFC %).

84.716.51 3.711.4 [ 1.8+0.8 [ 4.511.2 [ 2.810.9 J 1.210.2 E-RFC EA.-RFC M-RFC SIgfficeLls mono-CFu-c/5x104mnc % I cytes 20

is •15

10- 10

5-

i i i 1234 1234 1234 123 4 12 34 12 34

Ordinate (left) : percentage of various cell types in the final different interphase cell suspensions. Ordinate (right) : CFU-c/5 x 10* cells plated, in the final different interphase cell suspensions. Abscissa : 1,2, 3 and 4 represent the various second separation steps. Vertical bars represent mean ± s.e.m. 72

Table 2.VII.b: Differences between the various cell type percentages, obtained during a two-step cell separaticn procedure, with removal of different rosette combinations during the second step (N = 6 buffy coats)

E- EAi- M- SIg(+) Mono- CFU-c/ RFC% RFC% RFC% cells% cytes% 5xlO4

1 p<0.05 p<0.05 NSD NSD NSD p<0.05 compared to* -\ 2 p<0.05 NSD NSD p<0.05 p<0.05 p<005 Step 1 < • Step 2 ;•* 3 p<0.05 NSD NSD NSD NSD K0.05 * 4 p<0.05 NSD NSD NSD NSD p<0.05

Comparison of* Step 2 combination 1-2 NSD p<0.05 NSD p<0.05 p<0.05 NSD 1-3 NSD NSD NSD NSD NSD NSD 1-4 p<0.05 p<0.05 NSD NSD NSD NSD 2-3 NSD NSD NSD NSD NSD NSD 2-4 NSD NSD NSD NSD NSD NSD 3-4 NSD NSD NSD NSD NSD NSD

Tested with Student's t-test for paired observations NSD = non significant difference For the different cell separation steps: see Table 2.VH.a

2.3.7 Modification of a B-cell rosette technique, using goat anti-human Fab (GahuFab) coated SRBC (= B-RFC), as indicator Since the aim of this study was to purify CFU-c in a null cell fraction (non-B, non-T), a rosette technique, which would make use of SIg on B-lymphocytes (and its subsequent removal by the use of density gra- dients), was introduced. Since SIg are present on all B-lymphocytes and bind to IgG-Fc receptors on monocytes, it could be expected that the non-removed monocytes after the first cell separation step would also be eliminated by this type of rosette. Giuliano et al. (34) described a B-RFC technique in which SRBC were coated with GahuFab by addition of CrCl3. Using the material and methods for this rosette as described by these authors, we were unable to observe proper rosettes because of agglutination. Therefore, with the technique described, using 0.1 ml GahuFab [ 1:25 diluted, simultaneously incubated with 0.1 ml CrCl3 (0.38 M) 1:20 diluted and 0.1 ml SRBC (10 x 108) cells], the effect of different incubation times on the number of coated SRBC and the degree of SRBC coating with GahuFab were tested. 73

The results are presented in Table 2.VIII. It can be seen that the percentage of SRBC coated with GahuFab reaches a plateau after 15 min. of incubation. However, with a longer incubation time, a drastic decrease in coated SRBC yield was observed owing to agglutination. Therefore, in the next experiment serial dilutions of CrCl3 and GahuFab were tested using a fixed incubation time of 15 min. The results are presented in Table 2.IX. From this dilution experiment it can be concluded that a 1:100 dilu- tion for both CrCl3 and GahuFab, with an incubation time of 15 min. was optimal.

Table 2.VIII: Effect of different incubation times on coated SRBC yield and degree of coating with GahuFab; using 0.1 ml (1:20 diluted) GahuFab and 8 0.1 ml (0.38M, 1:20 diluted) CrCl3 and 0.1 ml (10xl0 ) SRBC

Incubation time (min.) SRBC yield % GahuFab coated SRBC (%)*

0 (control) 100 0

4 68.4 0 5 84.6 2 6 64.6 2 7 57.6 5 8 56.2 8 9 40.6 8 10 28.9 15 15 2.4 95 20 0.4 (+++) 100 (+++) 25 0.5 (+++) 100 (+++) 30 0.3 (+++) 100 (+++)

* determined with a direct immunofluorescence test, using 100 jul (1:5 diluted) FITC-con- jugated rabbit anti-goat IgG (30', 20°C) (+++) = agglutination

2.3.8 Testing of the B-RFC technique in a three-step separation experi- ment, in combination with monocyte, E-RFC and EA,-RFC elimination To test the efficiency of GahuFab coated SRBC as a B-RFC technique, buffy coat cells from 3 normal donors were processed to obtain a "con- trol" cell suspension after Ficoll-Isopaque (ds. 1.077 g/ml) centri- fugation (suspension A). The residual buffy coat cells were separated on Table 2.IX: Effect of different dilutions of CrCl3 and GahuFab on the degree of cell yield and coating of SRBC with GahuFab, performed with a standard incubation time of IS min.

15 min. of incubation: lOxlO8 0.1mlCrCl3 SRBC (0.1 ml) with GahuFab CeU GahuFab Agglutina- (0.38M) diluted to: yield coated tion diluted to: (xlO6) SRBC (%)* 1:25 1:50 1:100 1:200

1 1:20 0.1ml 13.4 100 2 1:20 0.1ml 9 100 3 1:20 0.1ml 15 100 i 4 1:20 0.1ml 17.2 100

1 1:50 0.1ml 1.1 (xlO8) 100 2 1:50 0.1ml 0.7 100 3 1:50 0.1ml 0.7 100 4 1:50 0.1ml 0.4 100

1 1:100 0.1ml 7.6 36 2 1:100 0.1ml 7.5 67 - 3 1:100 0.1ml 8 95 4 1:100 0.1ml 7.9 90

1 1:200 0.1ml 12.7 10 2 1:200 0.1ml 10.1 10 - 3 1:200 0.1ml 11.2 35 4 1:200 0.1ml 8.2 20

1 NACL 0.9% 0.1ml 12.7 2 2 NACL 0.9% 0.1ml 11 2 3 NACL 0.9% 0.1ml 8.4 0 - 4 NACL 0.9% 0.1 ml 13.4 0

1 CrCL3 1:20 NACL 0.1 ml 18.4 5 8 2 CrCL3 1:50 NACL 0.1 ml - - 1 (xlO ) 5 8 3 CrCL3 1:100 NACL 0.1 ml 6.5 (xlO ) 10 4 8 * CrCL3 1:200 NACL 0.1 ml 10 (xlO ) 10 determined with a direct immunofluorescence test, using 100//I (1:5 diluted) FITC-conjugated rabbit anti-goat IgC (30\ 20°C) 75

a Ficoll-Isopaque (ds. 1.077 g/ml) density gradient after carbonyl-iron incubation. Subsequently the interphase cells (suspension B) were al- lowed to form E + EAj-RFC and were separated on a Ficoll-Isopaque (ds. 1.077 g/ml) gradient. After the second separation step the interphase cells (C) were allowed to form B-RFC and were then centrifuged on a Ficoll-Isopaque (ds. 1.072 g/ml) gradient. After this last centrifugation step the interphase cells (E) were harvested and analysed. By this, it was expected to obtain a high percentage of B-RFC in suspension C, which could subsequently be removed during the last separation step. With buffy coat cells of 3 normal donors this hypothesis was tested and the results are presented in Table 2.X.

Table 2.X: Testing of the B-RFC technique in a three-step cell separation procedure (N = 3 buffy coats)

Difference Characteristics B C E between B/E

E-RFC % 66.5 ±3.2 ->• 1 5.5 ±1.7 5±2.8 1 NSD EAi-RFC% 5.5±0.2 -> 1 0.5 ±0.3 -» tl2.2±0.2 t B-RFC % 8.5 ±0.3 -»• t 27±3.5 -» 4 2.5 ±0.3 SIg(+)ceUs % 8±0.7 ->• t 24 ±0.7 -* I 3.5 ±0.3 I Cell yield*'**(xlO6) 1,000 _> . 100±2.5 _> . 8.911.6 (=100$) + (=10%) + (=0.8%)

t or -!• = significant difference (p<0.05) NSD = non significant difference * cell yield corrected to 1,000x106 cells in suspension B ** percentages calculated from the mean values Step 1 -*• Suspension B: interphase cells after carbonyl-iron incubation and subsequent Ficoll-lsopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells Step 2 ->• Suspension C: interphase cells after combined E-RFC + EAi-RFC removal from suspension B, using a ds. 1.077 g/ml Ficoll-lsopaque gradient Step 3 -> Suspension E: interphase cells after B-RFC removal from suspension C, using a ds. 1.072 g/ml Ficoll-lsopaque gradient

As can be seen, a significant increase in B-RFC and SIg(+) cells was obtained in suspension C. After B-RFC elimination (suspension E) a significant B-RFC and SIg(+) cell removal was found. However, a red- 76

procal increase was observed with the EAi-RFC. This indicates that the B-RFC does not eliminate cells with IgG-Fc receptors as far as it was tested with this assay.

2.3.9 Testing of the B-RFC technique in a two-step separation experi- ment; effect of B-RFC removal on monocytes The experiments in section 2.3.8 indicated that the B-RFC sedimentation also eliminates monocytes. To substantiate this finding more definitely, an experiment was performed with one buffy coat, in which the initial monocyte elimination with carbonyl-iron was omitted and the B-RFC technique directly carried out on the buffy coat cell suspension. For that reason the contaminating HuRBC in the buffy coat were eliminated by rouleaux-formation using methylcellulose 2% sedimentation (suspension A). Next, B-RFC elimination with a Ficoll-Isopaque density 1.077 g/ml gradient was performed. The interphase cells obtained after this centrifugation (suspension B) were analysed and the results are presented in Table 2.XI.

Table 2.XI: Effect of B-RFC removal on monocytes (N= 1 buffy coat)

Characteristics A B

E-RFC% 33 6.7 EAi-RFC% 5 7.1 B-RFC % 16 3.2 SIg(+) cells % 12 1.5 Monocytes % 22 7 Granulocytcs % 34 5 Cell yield* (xlO6) 1,000 139.3 (=100%) (=13.9%)

CFU-c/5xlO4 0.5 3 CFU-c absolute 10,000 8,340 CFU-c ratio 1:100,000 1:16,666 I CFU-c purification (x) 1 6 CFU-c yield (%) 100 83.4

* cell yield corrected to 1,000x106 cells in suspension A Suspension A: buffy coat cells after methylcellulose 2% sedimentation Suspension B: interphase cells after B-RFC removal from suspension A, using a ds. 1.077 g/ml Ficoll-Isopaque gradient 77

It is obvious that the monocytes, which are normally situated in the light density region, are eliminated by the B-RFC technique. For gra- nulocytes it is difficult to determine the "depletion effect" of the B-RFC technique, because the majority of granulocytes will appear in the peiiet fraction of the tube, only because of their intrinsic density.

2.3.10 The effect of a simultaneous "triple rosette" combination (E- + EA] - + B-RFC) during the second separation step on CFU-c purification

In the next experiment the effect of a triple rosette combination during the second separation step on CFU-c purification was investigated, in an attempt to limit the total number of density gradient separation steps. From the buffy coat cells of 3 donors a "control" MNC suspension (A) was made. After carbonyl-iron pre-incubation and density 1.077 g/ml gradient separation (step 1) the interphase B was allowed to form simultaneous rosettes. Subsequently this cell suspension was centrifuged on a density 1.072 g/ml Ficoll-Isopaque gradient and the cells in the interphase (C) and pellet (D) were investigated. The results are presented in Table 2.XII. The effect of this triple rosette combination on the end-suspension C, compared to suspension A, is an acceptable removal of E-RFC and EAj-RFC, but an unfortunate interaction with B-RFC elimination, resulting in an inadequate B-RFC removal. However, a CFU-c purification factor of x 11 is obtained, with a final cell yield of 2.1%. 78

Table 2.XH: Effect of a combined E-RFC + EAi-RFC + B-RFC removal during the second Ficoll-Isopaque (ds. 1.072 g/ml) centrifugation step (N= 3 buffy coats)

Difference Characteristics A B C D between A/C B/C

E-RFC % 77±1.4 NT 16.6±5.4 82.6+1.3 I -

EArRFC% 8.8±0.9 NT 2+0.8 6.5±2.3 1 - B-RFC % 20.3±4.9 NT 16.6±5.1 16.6±5.8 NSD - SIg(+)cells % 9±0.4 NT 20+7.3 3.8±1.7 NSD - Monocytes % 4±1.2 NT 3±0.4 4.8±1.7 NSD - Granulocytes % 3.3±0.2 NT 3.6±0.6 4±2.1 NSD - Cell yield •.••(xlO6) NT 1,000 21+4.6 850+53.1 - (=100%) (=2.1%) (=85%) CFU-c/5xlO4 NT 0.5±0.2 5.5±1.9 0.3±0.1 - t CFU-c absolute - 10,000 2,310 5,100 - - CFU-c ratio - 1:100,0001:9,090 1:166,666 - - CFU-c purification (x) - 1 11 - - t CFU-c yield (%) - 100 23.1 51 - -

t or I = significant difference (p<0.05) NSD = non significant difference NT = not tested * cell yield corrected to l.OOOxlO6 cells in suspension B ** cell yield (%) calculated from the mean values Suspension A: Inteiphase cells after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation Step 1 - Suspension B: Interphase cells after carbonyl-iron incubation and subsequent Ficoll- Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells ^Suspension C: Interphase cells after a combined E-RFC + EA,-RFC + B-RFC removal from suspension B, using a ds. 1.072 g/ml Ficoll-Isopaque Step 2 gradient "Suspension D: pellet cells

I 2.3.11 Another approach for monocyte removal and subsequent testing in a two- and one-step separation technique; effect on CFU-c purification During the progress of the previous experiments another method for monocyte removal was tested. An earlier described disadvantage of mono- cyte removal with carbonyl-iron pre-incubation and Ficoll-Isopaque den- sity gradient centrifugation is a substantial cell loss of about 50% (see 79

section 2.3.4). In an attempt to reduce this cell loss we tested the fol- lowing approach: 1) concentration of buffy coat cells using methylcellulose 2% sedimen- tation; 2) subsequent incubation of these buffy coat cells with carbonyl-iron and application of a sterile magnet; 3) carefully decanting the non-phagocytic buffy coat cells, so that a Ficoll-Isopague density gradient step can be omitted. To test this method, buffy coat cells from 3 normal donors were pro- cessed as follows: leucocytes were concentrated by methylcellulose sedimentation and then depleted of monocytes by the above-mentioned method (suspension B) or incubated with carbonyl-iron and separated on a Ficoll-Isopaque density 1.077 g/ml gradient (suspension E). Subsequently the cells obtained by the two methods were processed for a simultaneous E-RFC and B-RFC rosette and centrifuged on a den- sity 1.072 g/ml gradient (suspension C and F respectively). The results are presented in Table 2.XIII. As can be seen, the differences between suspension B and E are not significant as far as the various lymphocyte subpopulations, monocytes and CFU-c are concerned. However, the cell yield and the percentage of granulocytes are significantly lower in suspension E than in suspension B. It must be stressed that even with carbonyl-iron pre-incubation and Ficoll-Isopaque centrifugation the elimination of granulocytes is incom- plete. After combined E-RFC + B-RFC rosette elimination the results obtained from suspensions C and F were not different. Although good results were obtained with this combined B-RFC and E-RFC rosette sedimentation after initial monocyte removal, using methyl-cellulose 2% sedimentation and carbonyl-iron incubation, a second attempt was made with a "triple" rosette combination and one Ficoll- Isopaque density gradient separation step. Therefore, leucocytes were concentrated using methylcellulose 2% sedimentation; the cells were then allowed to form rosettes with E-RFC, EAi-RFC and B-RFC. Next these rosettes were centrifuged using a density 1.072 g/ml Ficoll-Isopaque gradient. The results are presented in Table 2.XIV. Again, it is obvious that optimal removal of E-RFC and EAi-RFC is obtained, but that interference with the B-RFC (c.q. SIg(+) cells) re- moval occurs. Besides, the granulocytes are not adequately removed. The interference of this rosette combination with the B-RFC might be caused by binding of the goat anti-human Fab with the Fab portion of the anti-D IgG on the human erythrocytes. 80

Table 2.XIH: Two different techniques for obtaining monocyte-free cell suspensions, followed by a combined E-RFC + B-RFC removal during the second cell separation step (N = 3 buffy coats) Difference Characteristics B C E F between B/E C/F

E-RFC % 74±3.5 -> 4 9+5.0 75+3.5 -+47.7+1.2 NSD NSD EAi-RFC % 4±0.5 -• 16.7±0.6 410.2 ->t6.210.1 NSD NSD B-RFC % 8±0.8 -> 4 2.710.2 11.213.6 -*43i0.6 NSD NSD SIg(+) cells % 5.2±0.4 -> 40.510.2 4.210.5 -40.810.3 NSD NSD Monocytes % 0.6±0.2 0.3+0.1 0.8+0.2 0.810.4 NSD NSD NSD -• NSD Granulocytes % 12±0.5 -> 4,2.711.2 5.710.4 -» 3.911 NSD -* NSD Cell yield *.**(xlO6) 323±29.9 6.5+2.4 104136.6 3.911 I NSD (=32.3%) "* ^(=0.65%) (=10.4%) ""'''(=0.39%) CFU-c/SxlO4 1.6+0.5 -»• 120.7+8.7 2.6+0.9 -+tl3.7+4.2 NSD NSD CFY-c absolute 10,336 2,691 5,408 1,068 - - CFU-c ratio 1:31,250 1:2,415 1:19,230 1:3,649 - - CFU-c purification (x) 1 12.9 1 5.2 NSD NSD CFU-c yield (%) 100 26 100 19.7 - -

t or 4 = significant difference (p< 0.05) NSD = non significant difference * cell yield corrected to l.OOOxlO6 cells in the initial buffy coat ** cell yield (%) = calculated from the mean values Suspension B: Buffy coat cells after methylcellulose 2% sedimentation and sub- sequent carbonyl-iron incubation. Cells were carefully decanted. No Step 1 Ficoll-Isopaque centrifugation step Suspension E: Buffy coat cells after carbonyl-iron incubation and Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation Step 2 Simultaneous E-RFC + B-RFC removal, using a ds. 1.072 g/ml Ficoll-Isopaque gradient Suspension B -> C I Suspension E -> F For that reason two other EA-rosette assays were introduced; EA2 -RFC (using sheep red blood cells, coated with rabbit anti-sheep IgG) and EA3-RFC (ox red blood cells, coated with rabbit antiox IgG). To invest- igate these two EA-rosette techniques and especially the EA2-RFC, buffy coat cells of three normal donors were analysed. The results are presented in Table 2.XV. 81

Table 2.XIV: Effect of a simultaneous 'triple-rosette' elimination (N = 3 buffy coats)

Difference Characteristics B C D between B/C

E-RFC % 64.5 ±0.3 2±0.7 71.5±0.3 I

EArRFC % 2±1.4 0±0 l±0 I B-RFC% 8±1 13±0.2 11.5+1.4 t SIg(+)cells % 6.5±0.4 9±0.3 3.5+0.3 t Monocytes % l±0 4±0.3 l±0 t Granulocytes 19.5±0.3 18±2.4 23.5±5.3 NSD Cell yield *.**(xlO6) 4.6 730.9 (=100%) (=0.46%) (=73%) CFU-c/5xlO4 NT NT NT t or = significant difference (p<0.05) NSD = non significant difference * cell yield corrected to i,000xl06 cells in suspension B ** cell yield (%): calculated from the mean values Suspension B: Buffy coat cells after methylcellulose 2% sedimentation and sub- sequent carbonyl-iron incubation; cells were carefully decanted. No Ficoll-Isopaque centrifugation step Suspension C: Interphase cells after a combined E-RFC + EAi-RFC + B-RFC re- moval from suspension B, using a ds. 1.072 g/ml Ficoll-Isopaque Ste v gradient Suspension D: Pellet cells

Initially leucocytes were concentrated by using methylcellulose 2% sedimentation (suspension A). It can be seen that the EA2-RFC had a percentage of 41.1 ± 4.4, so this assay probably also detects IgG-Fc- receptors on monocytes and/or granulocytes. Another possibility is that the rabbit anti-sheep IgG does not properly coat the SRBC, by which an interference with the E-RFC is obtained. The EA3-RFC percentage was 24.6 ± 2.9 and therefore gives a more realistic estimate of the percentage of IgG-Fc positive cells. After monocyte removal (suspension B) no significant differences were observed in the EA2- and EA3-RFC, which might indicate that these EA-assays do not detect IgG-Fc-receptors on monocytes. After subsequent EA2-RFC removal (suspension B) using a density 1.077 g/ml Ficoll-Isopaque gradient (suspension C), it is very interesting to observe that contrary to a relative enrichment of E-RFC as was ob- served after EAi -RFC elimination, a decrease in E-RFC was found. This Table 2.XV: Effect of sequential EA2-RFC and E-RFC + B-RFC removal on CFU-c purification (N = 3 buffy coats)

Difference Characteristics A B C D E F between C/E B/E A/E

E-RFC % 52.3±3.8 -> T633±3.6 •* 121.6±4 67.8±0.1 2+1.2 34.3+6.7 4 4 4

EA2-RFC % 41.1+4.4 38.8±4.9 -* 49.3+2.2 39.6+5.7 410.2 12.1±2 4 4 4 ->• NSD EA3-RFC % 24.6+2.9 22.3±1.4 -* 48.6+1.7 15.8+3.3 5.6±O.S 9.3+1.3 NSD 4 4 -* NSD B-RFC % 32±0.5 29+9.9 30±7.3 14±3.2 4.6+0.2 33+0.2 4 4 4 -*• NSD ->• NSD SIg(+) cells % 3.8±0.3 4.5+0.6 -* t24±3.8 1.3+0.2 4.2+0.1 23+5.3 4 NSD NSD -* NSD Monocytes % 13.6+4.1 -»• 10.5+0.2 0.1±0.1 0.6+0.1 010 0.1+0.1 NSD NSD 4 •* NSD Granulocytes % 39±12.3 -*• 427.3+8.9 -> 45.6±1.5 25.6±9.5 5.7+0.3 5-6 ±1.9 NSD 4 4

Cell yield *•** 641±76.5 466184.6 22.4+9.3 302.3±26.2 0.9+0.1 15.8+7.5 4 4 4 OO 6

might indicate that the EA2-RFC overlaps the E-RFC-assay, or detects IgG-Fc receptors on T-lymphocytes. Furthermore, it had to detect IgG-Fc positive cells, because the EA3-RFC were also significantly lower in suspension C than in suspension B. On the other hand, the percentage of SIg(+) cells after this centrifugation step was increased to 24 ± 3.8, while the B-RFC were not significantly different. Since there was no significant alteration in the percentage of mono- cytes between suspension B and C, it could be that the B-RFC also detects SIg on granulocytes. Therefore, it is reasonable to assume that the fact that the B-RFC percentage was not changed from suspension B to C, was caused by an increase in SIg(+) cells and a decrease in the percentage of granulocytes. Furthermore, it was interesting to observe a significant purification of CFU-c of x 14.8, which indicates that CFU-c do not bear IgG-Fc- receptors, if measured with this EA2-RFC technique. When the interphase cells obtained after centrifugation step 1 were then processed for a simultaneous E-RFC + B-RFC removal (suspension E), there was a significant decrease of mobt cell types. However, the granulocytes were difficult to eliminate completely. An ultimate CFU-c purification of x36.8 was reached with a CFU-c ratio of 1:1,170. How- ever, this was achieved at the cost of a CFU-loss of 94.9% and an ulti- mate total cell yield of 0.1 %.

2.3.12 The effect of a two-step density gradient technique, using mono- cyte removal during the first step and simultaneous E- and B-RFC elimination during the second step From the previous experiments it is obvious that the combined removal of E-RFC and B-RFC has a considerable impact on CFU-c purification. Therefore, this approach was studied more thoroughly. From the buffy coat cells of 13 donors a control leukocyte suspension was made by methylcellulose 2% sedimentation. For the first step these buffy coat cells were centrifuged, using a Ficoll-Isopaque density 1.077 g/ml gradient after pre-incubation with carbonyl-iron (suspension B). Interphase cells obtained after this first step were allowed to form simul- taneous rosettes, after which they were centrifuged using a density 1.072 g/ml Ficoll-Isopaque gradient. The results are given in Table 2.XVI. After the first step, which involves removal of monocytes and granulo- cytes, there was an increase in E-RFC and EA3-RFC as was observed before. The EAj-RFC were decreased, because they detect mainly the 84

Table 2. XVI: Effect of a combined E-RFC + B-RFC removal during the second cell separation step on CFU-c purification (N = 13 buffy coats)

Difference Characteristics A B C D between A/C

E-RFC % 47.2±3.1 ->t65.1±3.8-*-4-9±1.4 70.9±3.3 EA,-RFC% 4.2+0.6 -^1.7±0.5 -*t3.5±1.2 3.2±0.5 NSD

EA2-RFC % 44.7+3.1 ^•t61.9±3.7-*|37.5±3 59.7±2.5 NSD

EA3-RFC% 15.5±2 19±3.5 24.2±0.8 22.5 ±3.9 t -»• NSD -*• NSD B-RFC % 17.5±2.6 -*;8.6±1.1 -»44.5±0.7 7.6±0.7 I SIg(+)ceUs % 5.3±0.6 3.6±0.3 -9-11.1 ±0.2 5.7±0.5 1 -• NSD Monocytes % 10.2+1.2 ->4 0.4 ±0.1 0.7+0.4 l±0.2 ->• NSD Granulocytes % 43.2±3.8 ->424.6±5.5 13.3±3.8 25.6 ±4.5 •+ NSD Cell yield *• **(xlO6) 1,000 . 450±56.3 7.7±1.4 375±37.5 I (=100%) (=0.47%) (=37.5%) CFU-c/5xlOs 6.2±1.1 -+tl6.1±3.4 -4216.8154.4 1.8 ±0.75 t CFU-c absolute 12,400 14,490 3,338 1,350 - CFU-c ratio 1:80,645 1:31,055 1:2,306 1:277,777 — CFU-c purification (x) 1 2.5 34.9 - t CFU-c yield (%) 100 116.8 26.9 10.8 - t or + = significant difference (p<0.05) NSD = non significant difference * cell yield corrected to l.OOOxlO6 cells in suspension A ** cell yield (%): calculated from the mean values Suspension A: Buffy coat cells after methylcellulose 2% sedimentation Step 1 -*• Suspension B: Interphase cells after carbonyl-iron incubation and subsequent Ficoll- Isopaque (ds. 1.077 g/ml) centrifugation of suspension A cells . Suspension C Interphase cells after a combined removal of E-RFC + B-RFC from Step 2 . suspension B, using a ds. 1.072 g/ml Ficoll-Isopaque gradient Suspension D: Pellet cells

IgG-Fc receptors on monocytes and furthermore an expected decrease also occurred in the B-RFC. The CFU-c purification increased by a factor x2.5. After subsequent combined E-RFC and B-RFC removal in step two, an ultimate CFU-c purification of x 34.9 was found, with a final cell yield of 0.47%. 85

When suspension A was compared to the final suspension C, a significant decrease in T-lymphocytes, B-RFC, monocytes and granulocytes was ob- served, while no significant difference was found for the EA,-RFC and EA2-RFC, and an increase in EA3-RFC. The CFU-c purification which was ultimately reached is comparable with the normal CFU-c content of human bone marrow, with a CFU-c ratio of 1:2,306. However, this purification could only be achieved by accepting a CFU-c loss of 73.1 %.

2.3.13 The effect of a two-step density gradient technique, using mono- cyte removal during the first step and simultaneous E-, B- and EA2 -RFC elimination during the second step From previous experiments it was clear that a combined triple rosette elimination, using E-, EA!- and B-RFC, induced an interference with the B-PFC removal. Our final approach was to accomplish a triple rosette combination, using E-RFC + EA2-RFC + B-RFC during the second separation step, while the monocytes and granulocytes were mainly removed during the first step. It was to be expected that the remaining IgG-Fc positive cells in the final suspension, when E-RFC + B-RFC were used only (see section 2.3.12), could be eliminated by adding the EA2-rosette separa- tion. Since the EA2-indicator cell is a SRBC coated with rabbit anti- sheep IgG, no interference with the B-RFC was expected. For this final experiment the buffy coat cells of 10 normal donors were used and pro- cessed as follows. A control suspension A was obtained after leucocyte concentration using methylcellulose 2% sedimentation. The first step involved Ficoll- Isopaque density 1.077 g/ml centrifugation after pre-incubation with carbonyl-iron. The interphase cells obtained after the first step were then allowed to form simultaneous E-RFC + B-RFC + EA2-RFC rosettes. Thereafter they were centrifuged on a Ficoll-Isopaque density 1.072 g/ml gradient. The results are presented in Table 2.XVII. The alterations observed in suspension B are well known from the previous experiments. After the triple rosette elimination it can be seen from suspension C that there is an overall decrease in every cell type. The ultimate cell yield is 0.26%. The CFU-c purification is x48.4, with a CFU-c ratio of 1:2,515. The ultimate CFU-c loss is 87.3%. In conclusion, applying the above-mentioned technique it is feasible to obtain a null cell suspension which is highly purified for CFU-c, by 86

using two density gradient separation steps and combined rosette elimi- nation.

Table 2.XVII: Effect of a combined E-RFC + EA2-RFC + B-RFC removal during the second cell separation step on CFU-c purification (N = 10 buffy coats)

Difference Characteristics A B C D between A/C

E-RFC % 56.3±1.2 -*-t69.1±3.5 -*47.11il.8 67.8+3.9 1

EA2-RFC % 45.3±2.8 ->t56.7+2.5 ->4-12.3±2.8 49.8+2.6 I

EA3-RFC % 19.3±2 18.1±2.9 -> 19.9+1.5 15+3.3 I -> NSD B-RFC % 13.7±1.7 -•J.7.710.8 -•4,4.210.5 10.912.1 I SIg(+) cells % 5.410.3 3.5±0.5 -41.2+0.3 710.5 -• NSD Monocytes % 10.6±1.7 -s-10.610.1 0.5+0.2 1.5+0.2 -* NSD Granulocytes % 35±2.7 -+J26+2.3 -44.5+2.3 30.6+4.5 Cell yield *>**(xlO6) 1,000 470+45.3 2.610.2 367140.5 (=100%) ~)4(=0.26%) (=36.7%) CFU-c/5xl0s 4.1±0.8 ->tl6.7±0.3 ->tl98.8l30.2 2.810.8 t CFU-c absolute 8,200 15,698 1,045 2,055 - CFU-c ratio 1:121,951 1:29,940 1:2,515 1:178,571 - CFU-c purification (x) 1 4 48.4 - t CFU-c yield (%) 100 191 12.7 25 - t or •!• = significant difference (p <0.05) NSD = non significant difference * cell yield corrected to 1,000x10* cells in suspension A ** cell yield (%): calculated from the mean values Suspension A: Buffy coat cells after methyIcellulose 2% sedimentation Step 1 Suspension B: Interphase cells after carbonyl-iron incubation and subsequent Ficoll- Isopaque (ds. 1.077 g/ml) centrifugation of suspension A cells

, Suspension C: Interphase cells after a combined E-RFC + B-RFC + EA2-RFC re- moval from suspension B, using a ds. 1.072 g/ml Ficoll-Isopaque gra- Step 2 dient ' Suspension D: Pellet cells 87

2.4 CONCLUSIONS AND DISCUSSION

Conclusions:

1) In human blood, 1,165 (± 80.6) x 106 leucocytes containing 500 (± 32) x 106 MNC were present in buffy coats obtained from 440 ml. In these leucocyte suspensions the CFU-c content per 5 x 105 cells plated was 10.4 ± 1.9, which correlated with a mean absolute CFU-c concentration of 20,800 per 1,000 x 106 leucocytes. After Ficoll-Isopaque (ds. 1.077 g/ml) gradient centrifugation, the CFU-c concentration was 26.1 ± 10.3 per 5 x 10s MNC plated. 2) Linear density gradient centrifugation of MNC demonstrated that cells were still present in the high density region, which were mainly T-lymphocytes. The latter showed a density distribution throughout the whole density region from 1.055 to 1.095 g/ml. On the other hand, the B-lymphocytes, monocytes and CFU-c were mainly distri- buted in the light density region. Linear density gradient centrifu- gation of MNC after monocyte removal with carbonyl-iron revealed a shift of the mean density to a higher value. This was caused by an increase in the cell density of B-lymphocytes and non-removed monocytes. 3) None of the used rosette sedimentation techniques allowed absolute elimination of the cells under study. By lowering the Ficoll-Isopaque density to 1.072 g/ml, the percentage of non-eliminated monocytes, B-lymphocytes and T-lymphocytes was somewhat lower, which re- sulted in a higher CFU-c purification. 4) CFU-c did not adhere to nylon-wool columns, neither did they bear Fc receptors for IgG, as studied with various EA-rosette techniques. It seemed that the EA^RFC assay was more sensitive to Fc-IgG- receptors on monocytes. The results with the M-RFC were not con- clusive in terms of presence of this receptor on CFU-c. 5) The rosette technique for B-lymphocytes (B-RFC) proved to be re- producible and did not only eliminate the majority of B-lymphocytes, but also most monocytes and granulocytes. However, the B-RFC dis- played reciprocal interaction with the EAj-RFC, which means that the EA,-RFC technique probably detects a B-lymphocyte sub- population.

6) The "triple" rosette technique (E- + EAr + B-RFC), developed in an attempt to reduce the total number of Ficoll-Isopaque gradient steps, resulted in an interaction with the B-RFC and prevented its sedimentation. 88

7) The method to remove monocytes by using methylcellulose 2% sedimentation combined with carbonyl-iron pre-incubation and avoidance of a Ficoll-Isopaque gradient separation step, resulted in a higher cell yield as well as in a higher residual percentage of gra- nulocytes. 8) The EA2 -RBC technique proved to be more specific for IgG-Fc- receptors on B-lymphocytes, but also had an overlap with T-lympho- cytes. 9) By various "trial and error" separation techniques, the final approach, comprising monocyte removal during the first step and a combined removal of B-lymphocytes, T-lymphocytes and IgG-Fc bearing cells (EA2 -RFC-technique), resulted in a CFU-c purification of x48.4. This implied an increase in CFU-c ratio from 1:121,951 (starting buffy coat suspension) to 1:2,515 (null cell suspension). However, the final cell yield was only 0.26% and the ultimate CFU-c loss was high: 87.3% ! 89

The various CFU-c entities and cell yields obtained from the most illus- trative cell separation procedures are summarized in Table 2.XVIII.

Table 2.XVIII: Summary of all cell separation experiments

Total Total TypeofMNC Cell CFU-c/MNC CFU-c Purification of cell ofFicoll- eliminated yield ratio loss % factor sepaiation Isopaque (or concentrated*) % steps steps

1 Monocytes (2) 2.3 1:6,097 81.4 x7.8 2 T-lymphocytes

1 "MNC-concentrated* from buffy coat"

(2) 2 Monocytes +B- 1.5 1:3,320 67.6 x7.9 lymphocytes with nylon-wool 3 T-lymphocytes

1 Monocytes (2)( 2.1 1:9,090 76.9 xll 2 B-lymphocytes T-lymphocytes IgG-Fc bearing cells (EAi-RFC)

1 Monocytes 0.6 1:2,415 74.0 xl2.9 2 B-lymphocytes T-lymphocytes

1 Monocytes (2) 0.4 1:2,306 73.1 x34.9 2 B-lymphocytes T-lymphocytes

1 Monocytes * 2 B-lymphocytes (2). T-lymphocytes 0.1 1:1,170 94.9 x36.8 3 IgG-Fc bearing cells (EA2-RFC)

1 Monocytes (2)^ 0.26 1:2,515 87.3 x48.4 2 B-lymphocytes T-lymphocytes IgG-Fc bearing cells (EA2-RFC) 90

Discussion:

In this study a rapid technique for the separation of the nucleated cells in human blood on the basis of altered cell density in order to obtain an enriched CFU-c fraction is described. The separation of lymphocyte suspensions on the basis of differences in density must fulfil particular requirements as regards the material to be used for the preparation of the gradients. The material must be dense enough to produce gradients covering the normal lymphocyte density range (1.055 - 1.095 g/ml). Besides, the material must allow generation of gradients which are iso-osmotic and have a constant pH throughout the gradient. Moreover, the material must not influence the cells with respect to aggregation, density or viability, nor should it be antigeneic. Its viscosity should be low in order to allow short centrifugation times. Finally, the gradients should give high cell yields and reproducible patterns. Several materials have been used for the generation of density gra- dients, continuous as well as discontinuous (63, 64). It was found that the relation between concentration and osmolarity of polyethylene glycol was non-linear. Moreover, a solution with the required density of 1.090 g/ml far exceeded the osmolarity of 290 mOsm (64). Gum- acacia also showed a non-linear relation between concentration and os- molarity. Although this compound is dense enough, its viscosity was too high. When Isopaque is mixed with an isotonic salt solution, a linear density gradient ranging from 1.000 to 1.063 g/ml and isotonic with human serum (290 mOsm) is obtained. However, this density region is too low for the separation of lymphocyte subsets. Owing to its high water-binding capacity, Ficoll shows an exponential increase in osmolarity with increasing concentrations. With Ficoll only it is not possible to obtain a linear density gradient wKh a constant os- molarity. However, an iso-osmotic gradient with low viscosity and a constant pH of 7.4 throughout the gradient, can be made by mixing Ficoll, Isopaque, Tris-HCl (0.175 M, pH 7.4 at 4°C) and a Krebs-Ringer- Tris solution (= 125 mM NaCl, 5 mM KC1, 1.2 mM MgSO4, 35 mM Tris, 1 mM Na phosphate (pH 7.4 at 4°C)), which has a linear increase in density (64). This method is reproducible and has no effect on the in vitro proliferative capacity of lymphocytes and MLC-tests, while it is non-toxic and non-stimulatory. However, rebanding experiments demon- strated that, to a small extent, Ficoll (-Isopaque) was either ingested by or adhered to the cells, causing an increase in cell density (64). 91

The Gaussian distribution profile after rebanding experiments showed a skewness which indicates that some, but not all cells, increase in den- sity. Similar changes have been reported for mouse lymphoid cells on albumin gradients (81). In linear density gradients equilibrium is reached within 30 min of centrifugation at 20°C, using 400 g. However, the density of mono- cytes increases continuously during density centrifugation (62). Cel- lular overload, which causes interphase impedence, is observed if the number of cells is not restricted to < 8 x 106/cm2. Furthermore, the re- fractive index of each sampled fraction is not influenced if the cell con- tent is approximately 107 per fraction (62). Using (linear) Ficoll-Isopaque density gradients for the separation of human MNC we found a mean cell yield of 56 ± 3.4%. Other investigators, using a variety of density gradient systems for leucocyte separation, have reported recoveries of 50-90% (29, 33, 59, 67, 79, 81, 89, 104, 107). The linear density distribution profiles of the different MNC subpopulations showed Gaussian curves with single peaks. Although in several other studies it was not specifically reported, inspection of the density distribution curves also suggested Gaussian characteristics of density profiles (27, 29, 46, 59, 68, 99, 110). The distribution of all MNC was found in the density range 1.055 - 1.095 g/ml with a peak at 1.071 g/ml, which correlates well with the value of 1.072 found by Loos and Roos (64) and 1.074, described by Huber et al. (47). The distribution of B- and T-lymphocytes showed two different peak densities, but with a substantial overlap. The B-lymphocytes were found in the light density region, as was described earlier by Huber (47), Yu (107), Zier (108) and Zipursky (109). The monocytes were also found in the light density region, confirming the results of other investigators (62, 64). As far as CFU-c's are concerned, most density distribution studies have been done with human bone marrow, where the CFU-c are found in the light density region from 1.055 to 1.070 with a peak around 1.060 g/ml (45, 50, 67). The CFU-c obtained from the peak density fraction also showed the largest colonies when cultured in vitro (45). From our study it became clear that in human peripheral blood the CFU-c are also situated in the light density region. In contrast to a shift in density for B-cells and non-removed monocytes after carbonyl-iron pretreatment, no such alteration was observed for the CFU-c. From these density distribution profiles it was clear that with density 92

separation no absolute separation of CFU-c could be obtained owing to a substantial overlap with B-lymphocytes, T-lymphocytes and monocytes. Therefore, CFU-c could only be purified by negative selection, e.g. removal of unwanted MNC. Various authors have described multi-step approaches to obtain null cell suspension, enriched for CFU-c. Richman and Chess (82) were the first who presented evidence that the number of CFU-c is increased in a suspension of null lymphocytes obtained from human peripheral blood by Ficoll-Isopaque density gradient centrifugation, followed by sequen- tial Sephadex G-200 anti-Fab column chromatography and E-rosette depletion. Not only the CFU-c were found in the null cell fraction, but also the red cell precursor BFU-e (22, 71). Since then, several investigators confirmed this observation and worked out more details about the null cells and their CFU-c content. The most important experiments are summarized in Table 2.XIX. Most

Table 2.XIX: Characterization of null cell suspensions

E- EA- EAC- Mono- Cell Authors Ref. RFC% cells RFC% RFC% cytes CFU-c yield

MacDermott 65 <8 9 60-80 9 9 etal. 1975 <1 r Chess et al. 16 1974,1976, 1977 17,18 4 2 9 30-60 2 9 1

&hl etal. 1977 72 24 0.8 7 13 9 297/lxlOs 4 (range 80-615) Richman 83 2 50 2 26/2xl0s 9 etal. 1978 <10 ? Klein etal. 1978 58 8 9 18 20 9 9 10 Breardetal. 1978 13 < 5 <2 ? 60 9 26/2xl0s < 10 Zwaan, 1980 this 7.1 1.2 9.9 20 0.5 198.8/5xlOs 0.26 thesis (see I chapter III)

investigators used multi-step cell separation techniques, which mostly implied removal of Brlymphocytes (and monocytes) by rabbit anti- human-Fab immuno-absorbent column chromatography, E-rosette depletion to remove T-cells using Ficoll-Isopaque gradients, and/or nylon-wool adherence to remove phagocytic cells. Mostly 3-5 cell sepa- ration steps were used to obtain null cells. 93

With our method, described in this chapter, a null cell suspension highly enriched for CFU-c could be obtained very rapidly. The cell characteristics are comparable to those observed by others (see Table 2.XIX). Our study also confirms the observation that CFU-c do not adhere to nylon-wool and do not bear IgG-Fc receptors on their cell surfaces. Furthermore, it was found by Richman et al. (83) that CFU-c do not bear complement receptors, but require the presence of peripheral blood mononuclear cell feeder-layers for maximal CFU-c proliferation. If cultured in vitro using conditioned medium, no cell growth was observed. A similar observation was done when null cell suspensions were cul- tured for BFU-e. In the presence of erythropoetin only, no BFU-e growth was observed; however, if T-lymphocytes were added or con- ditioned medium obtained from stimulated T-cells, then approximately 80 BFU-e per 1 x 10s null cells were found (71). The most obvious disadvantage of our technique to purify peripheral blood CFU-c, is the tremendous cell loss. Therefore, instead of a negative selection procedure, a positive selection of CFU-c using a specific anti- gen(s) on the CFU-c cell membrane, might be a better approach. One such an antigen could be the P23.30, which is present on 20% of the null cells (87). Recent experiments indicate that the functional activity of myeloid precursors (CFU-c) can be inhibited by hetero-antisera to the B-cell antigen P23.30, in the presence of complement (23, 103). More direct evidence comes from the observation that CFU-c can be separated as anti-P23.30 positive cells by means of the fluorescence activated cell sorter (37). However, with such an experiment it should be remembered that the P23.30 antigen is also present on most of the B-cells, some monocytes and some T-cells. The null cell suspension obtained by our two-step cell separation technique can be used for immunological studies and experiments in which the haemopoietic precursor cells are to be studied more precisely. 94

2.5 REFERENCES

1. Aiuti, F, Cerottini JC, Coombs RRA, Cooper M, Dickler HB, Fr^Iand S, Fudenberg HH, Greaves MF, Grey HM, Kunkel HG, Natvig J, Preud'homme JL, Rabellino E, Ritts RE, Rowe DS, Seligmann M, Siegal FP, Stjernsward J, Terry WD and Wybran J: Special tech- nical report: identification, enumeration and isolation of Band T lymphocytes from human peripheral blood. Report of a WHO/IARC-sponsored workshop on human B and T cells, London, July 1974. Scand. J. Immunol., 3: 521,1974. 2. Bach JF, Delrieu F and Delbarre F: The rheumatoid rosette. A diagnostic test unifying seropositive and seronegative rheumatoid arthritis. Amer. J. Med., 49: 213,1970. 3. Barr RD, Whang-Peng J, and Perry S: Hemopoietic stem cells in human peripheral blood. Science, 190:284,1975. 4. Basten A, Miller JFAP, Sprent J and Pye J: A receptor for antibody on B lymphcytes. I. Method of detection and functional significance, j. Exp. Med., 135: 610,1972. 5. Beaumariage ML and Focan C: Double test of spontaneous rosettes with sheep and mouse erythrocytes. Acta Haemat.,59: 321,1978. 6. Bentwich Z, Douglas SD, Siegal FP and Kunkel HG: Human lymphocyte sheep erythro- cyte rosette formation: some characteristics of the interaction. Clin. Immunol. Immuno- pathol., 1:511,1973. 7. Bianco C, Patrick R, and Nussenzweig V: A population of lymphocytes bearing a mem- brane receptor for antigen-antibody-complement complexes. J. Exp. Med., 132: 702,1970. 8. Bianco C and Nussenzweig V: Theta-bearing and complement-receptor lymphocytes are distinct populations of cells. Science, 173: 154,1971. 9. Boxel JA van, and Rosenstreich DL: Binding of aggregated 7-globulin to activated lympho- cytes in the guinea pig. J. Exp. Med., 139: 1002,1974. 10. B#yum A: Separation of leukocytes from blood and bone marrow. Scand. J. Clin. Lab. Invest., 21: Supplement 97,1968. 11. Brain P, Gordon J, and Willets WA: Rosette formation by peripheral lymphocytes. Clin. Exp. Immunol., 6: 681,1970. 12. Branganza CM, Stathopoulos G, Davies AJS, Elliott EV, Kerbel RS, Papamichail M, and Holborow EJ: Lymphocyte-erythrocyte (L.E.) rosettes as indicators of the heterogeneity of lymphocytes in a variety of mammalian species. Cell, 4: 103,1975. 13. Breard J, Chess L, and Schlossman S: The heterogeneity of the human null cells subclass. INSERM Symposium No. 8. Editors: B. Serrou and C. Rosenfeld. Elsevier/North-Holland. Biochemical press, 143,1978. 14. Brown G and Greaves MF: Cell surface markers for human T and B lymphocytes. Eur. J. Immunol., 4: 302,1974. 15. Chervenick P and Boggs D: In vitro growth of granulocytic and mononuclear cell colonies from blood of normal individuals. Blood, 37: 1,1971. 16. Chess L, MacDermott RP, Sondel PM, and Schlossman SF: Characterization of cells in- volved in human cellular hypersensitivity. Progress in Immunology II, Vol. 3. Editors: L. Brent and J. Holborow. North Holland Publishing Company, 125,1974. I 17. Chess L, Levine H, MacDermott RP, and Schlossman SF: Immunologic function of isolated human lymphocyte subpopulations. VI. Further characterization of the Ig-negative, E-rosette negative (null cell) subset. J. Immunol., 115: 1483,1975. 18. Chess L, Evans R, Humphreys RE et al.: Inhibition of antibody-dependent cellular cyto- toxicity and immunoglobulin synthesis by an antiserum prepared against a human B-cell la-like molecule. J. Exp. Med., 144: 133,1976. 19. Chiao JW, Pantic VS, and Good RA: Human peripheral lymphocytes bearing both B-cell complement receptors and T-cell characteristics for sheep erythrocytes detected by a mixed rosette method. Clin. Exp. Immunol., 18: 483,1974. 95

20. Chiao JW, Pantic VS, and Good RA: Human lymphocytes beating both recpetors for complement components and SRBC. Clin. Immunol. Immunopathol., 4: 545, 1975. 21. Claman HN and Chaperon EA: Immunologic complementation between thymus and bone marrow cells - a model for the two-cell theory of immunocompetence. Transplant. Rev., 1:92,1969. 22. Clarke BJ and Housman D: Characterization of an eiythioid precursor cell of high pro- liferative capacity in normal peripheral blood. Proc. Natl. Acad. Sci. U.S.A., 74: 1105, 1977. 23. Cline MJ and Billing R: Antigens expressed by human B-lymphocytes and myeloid stem cells. J. Exp. Med., 146: 1143,1977. 24. Coombs RRA, Gurner BW, Wilson AB, Holm G, and Lindgren B: Rosette-formation be- tween human lymphocytes and sheep red cells not involving immunoglobulin receptors. Int. Arch. Appl. Immunol., 39: 658,1970. 25. Dicier HB and Kunkel HG: Interaction of aggregated 7-globulin with B-lymphocytes. J. Exp. Med., 136: 191,1972. 26. Oickler HB: Studies of the human lymphocyte receptor for heat-aggregated or antigen complexed immunoglobulin. J. Exp. Med., 140: 508,1974. 27. Dilla MA van, Fulwijler MJ and Boone IU: Volume distribution and separation of normal human leukocytes. Proc. Soc. Exp. Biol. Med., 125: 367,1967. 28. Dwyer JM: Identifying and enumerating human T and B lymphocytes. Progr. Allergy, 21: 178,1976. 29. El-Arini MO and Osoba D: Physical properties of mouse spleen cells involved in the mixed leukocyte reaction and graft-versus-host disease. J. Immunol., 110: 1476,1973. 30. Porni L and Pernis B: Interactions between Fc-receptors and membrane immunoglobulins on B lymphocytes. In INSERM Symposium I. Ed.: Seligmann M, Preud'Homme JL and Kourilsky FM, New York: American-Elsevier Co., 193,1975. 31. Finland SS and Natvig JB: Identification of three different human lymphocyte popula- tions by surface markers. Transplant. Rev., 16: 114,1973. 32. Finland SS, Wisl(4ff F, and Michaelsen TE: Human lymphocytes with receptors for IgG. A population of cells distinct from T and B-lymphocytes. Int. Arch. Appl. Immunol., 47: 124,1974. 33. Gatien JG, Schneeberger EE, and Merler E: Analysis of human thymocyte subpopulations using discontinuous gradients of albumin: precursor lymphocytes in human thymus. Eur. J.Immunol.,5: 312,1975. 34. Giuliano UJ, Jasin HE, Hurd ER, and Ziff M: Enumeration of B-lymphocytes in human peripheral blood by a rosette method for the detection of surface-bound immunoglobulin. J.Immunol., 112: 1494,1974. 35. Gmelig FHJ: Characterization of human lymphocyte subpopulations by cell surface markers. Thesis, Utrecht, 1977. 36. Greaves MF and Brown G: Purification of human T and B lymphocytes. J. Immunol., 112:420,1974. 37. Greaves MF: Membrane antigens and enzymatic phenotypes in leukaemia: correlates of normal haemopoietic differentiation? INSERM Symposium No. 8. Ed.: Gerrou B, and Rosenfeld C. Elsevier/North-Holland. Biomedical press, 253,1978. 38. Grey HM, Rabellino E and Pirofsky B: Immunoglobulins on the surface of lymphocytes. IV. Distribution in hypogammaglobulinemia, cellular immune deficiency, and chronic lymphatic leukemia. J. Clin. Invest., 50: 2368,1971. 39. Gupta S and Grieco MH: Rosette-formation with mouse erythrocytes: probable marker for human B lymphocytes. Int. Arch. Appl. Immunol., 49: 734,1975. 40. Gupta S, Good RA, and Siegal FP: Rosette-formation with mouse erythrocytes. II. A marker for human B and non-T lymphocytes. Clin. exp. Immunol., 25: 319,1976. 96

41. Gupta S, Good RA.andSiegalFP: Rosette-formation with mouse erythrocytes. III. Studies in patients with primary immunodeficiency and lymphoproliferative disorders. Clin. exp. Immunol., 26: 204,1976. 42. Gupta S, Ross GD, Good RA, and Siegal FP: Surface markers of human eosinophils. Blood, 48: 755,1976. 43. Haegert DG, Hallberg T, and Coombs RRA: B and T lymphocyte subpopulations in human peripheral blood. Int. Arch. Appl. Immunol., 46: 525,1974. 44. Hallberg, T, Gurner BW, and Coombs RRA: Opsonic adherence of sensitized red cells to human lymphocytes as measured by rosette formation. Int. Arch. Appl. Immunol., 44: 500,1973. 45. Haskill JS, McKnight RD, and Galbraith PR: Cell-cell interaction in vitro: studied by density separation of colony forming stimulating and inhibiting cells from human bone marrow. Blood, 40: 394,1972. 46. Hilal SK, Mosser DG, Loken MK, and Johnson RW: A new technique for high-resolution density gradient separation of bone marrow cells. Ann. N.Y. Acad. Sci., 114: 661,1964. 47. Huber C, Zier K, Michlmayr, G, Rodt H, et al.: A comparative study of the buoyant den- sity distribution of normal and malignant lymphocytes. Brit. J. Haemat, 40: 93, 1978. 48. Huber H, and Fudenberg HH: Receptor sites of human monocytes for IgG. Int. Arch. Allergy appl. Immunol., 34: 18,1968. 49. Iscove NN, Senn JS, Till JE, and McCulloch EA: Colony formation by normal and leukae- mic human marrow cells in culture: effect of conditioned medium from human leukocytes. Blood, 37: 1,1971. 50. Iscove NN, Messner H, Till JE, and McCulloch EA: Human marrow cells forming colonies in culture: analysis by velocity sedimentation and suspension culture. Ser. Haemat. Vol. V., 2: 37,1972. 51. Jondal M, Holm G, and Wigzell H: Surface markers on human T and B lymphocytes. I. A large population of lymphocytes forming non-immune rosettes with sheep red blood cells. J. Exp. Med., 136: 207,1972. 52. Jondal M, and Klein G: Surface markers on human B and T lymphocytes. II. Presence of Epstein-Barr virus receptors on B lymphocytes. J. Exp. Med., 138:1365,1973. 53. Jondal M-. Surface markers on human T and B lymphocytes. III. A marker for lymphoid adherence. Scand. J. Immunol., 3: 269,1974. 54. J^nsson V, and Christensen BE: Distribution of B, T, and O lymphocytes in blood and tissues of normal humans, reflecting a kinetic model. Scand. J. Haem., 18: 185, 1977. 55. Julius MH, Simpson E, and Herzenberg LA: A rapid method for the isolation of functional thymus-derived murine lymphocytes. Eur. J. Immunol., 3: 645,1973. 56. Kaplan ME, and Clarke C: An improved resetting assay for detection of human T lympho- cytes. J. Immunol. Methods, 5: 131,1974. 57. Klein E, Klein G, Nadkarni JS, Nadkarni JJ, Wigzell H, and Clifford P: Surface IgM-kappa specificity on a Burkitt lymphoma cell in vivo and in derived culture lines. Cancer Res., 1300,1968. 58. Klein E, Bakacs T, Poros A et al.: Natural killer effect of human blood lymphocytes. INSERM Symposium No. 8. Ed.: Serrou B, and Rosenfeld C. Elsevier/North-Holland. Biomedical press, 181,1978. 59. Ladinsky JL and Westring DW: The effect of anticoagulants on the volume of normal and leukemic leukocytes. Cancer Res., 27: 1689,1967. 60. Lay WH, Mendes ND, Bianco C, and Nussenzweig V: Binding of sheep red blood cells to a large population of human lymphocytes. Nature, 230: 531,1971. 61. Lin PS, and Hsu CCS: Human leukaemic T cells with complement receptors. Clin. and Exp. Immunol., 23: 209,1976. 62. Loos JA, Blok-Schut B, Doom R van, et al.: A method for the recognition and separation of human blood monocytes on density gradients. Blood, 48,731,1976. 97

63. Loos JA and Roos 0: Isopycnic centrifugation of antigen-activated human blood lym- phocytes in the early phase of in vitro stimulation. Exp. Cell. Res., 86: 342,1974. 64. Loos JA and Roos D: Ficoll-Isopaque gradients for the determination of density dis- tribution of human blood lymphocytes and other reticulo-endothelial cells. Exp. Cell Res., 86: 333,1974. 65. MacDermott RP, Chess L, and Schlossman SF: Immunologic functions of isolated human lymphocyte subpopulations. Clin. Immunol. Immunopathol., 4: 41S, 1975. 66. Mendes NF, Miki SS, and Peixinho ZF: Combined detection of human T and B lympho- cytes by rosette formation with sheep erythrocytes and zymosan-C3 complexes. J. Im- munol., 113: 531,1974. 67. Messner H, Till JE, and McCulloch EA: Density distributions of marrow cells from mouse and man. Ser. Haemat. V., 2: 22,1972. 68. Metcalf D, Moore MAS, and Shortman K: Adherence column and buoyant density separa- tion of bone marrow stem cells and more differentiated cells. J. Cell. Physiol., 78: 441, 1971. 69. Moretta L, Ferrarini M, Durante ML, and Mingari MC-. Expression of a receptor for IgM by human T cells in vitro. Eur. J. Immunol., 5: 565,1975. 70. Moretta L, Webb SR, Grossi CE, Lydyard PM, and Cooper MD: Functional analysis of two human T-cell subpopulations: help and suppression of B-cell responses by T-cells bearing receptors for IgM or IgG. J. Exp. Med., 146: 184,1977. 71. Nathan DG, Chess L, Hillman D et al.: Human erythroid burst-forming unit: T-cell re- quirement for proliferation in vitro. J. Exp. Med., 147: 324,1978. 72. Oehl S, Schaefer UW, Boeckner WR, Kalden JR, and Peter H: Human peripheral blood "null-lymphocytes", a highly enriched pool for granulocytic stem cells (CFU-c). Z. Im- mun. Forsch. 152: 423,1977. 73. Ornstein L, Hudson A, and Saunders A: Improving manual differential white cell counts with cytochemistry. Blood cells, 2: 557,1976. 74. Papamichai] M, Brown JC, and Holborow EJ: Immunoglobulins on the surface of human lymphocytes. Lancet, II: 850,1971. 75. Pegrum GD and Evans CA: Significance of mouse red cell rosette forming lymphocytes in chronic lymphocytic leukaemia. Acta Haemat., 60: 36,1978. 76. Pellegrino MA, Ferrone S, and Theofilopoulos AN: Rosette formation of human lymphoid cells with monkey red blood cells. J. Immunol., 115:1065,1975. 77. Psilmann P, Perlmann H, and Wigzell H: Lymphocyte mediated cytotoxicity in vitro. Induction and inhibition by humoral antibody and nature of effector cells. Transplant. Rev., 13: 91,1972. 78. Pernis B, Forni L, and Amante L: Immunoglobulins as cell receptors. Ann. N.Y. Acad. Sci., 190: 420,1971. 79. Pretlow TG and Laberoff DE: A new method for separating lymphocytes and granulocytes from human peripheral blood using programmed gradient sedimentation in an isokinetic gradient. Immunol., 24: 85,1973. 80. Raff MC: Two distinct populations of peripheral lymphocytes in mice distinguishable by immunofluoiescenge. Immunol., 19: 637,1970. 81. Raidt DJ, Mishell RI, and Dutton RW: Cellular events in the immune response: Analysis and in vitro response of mouse spleen cell populations separated by differential flotation in albumin gradients. J. Exp. Med., 128: 681,1968. 82. Richman C, and Chess L: Concentration of human peripheral blood stem cells in the null cell fraction. Blood, 46: 1011 (Abstr.), 1975. 83. Richman, and Chess L, and Yankee RA: Purification and characterization of granulocytic progenitor cells (CFU-c) from human peripheral blood using immunologic surface markers. Blood, 51: 1,1978. 98

84. Roitt IM, Greaves MF, Torrigiani G, Brostoff J, and Playfair JHL: The cellular basis of immunological responses. Lancet, II: 367,1969. 85. Roos D, and Loos JA: Changes in the carbohydrate metabolism of mitogenically stimulated human peripheral lymphocytes. I. Stimulation by phytohaemagglutinin. Biochem. Biophys. Acta, 222: 565,1970. 86. Ross GD and Polley MJ: Specificity of human lymphocyte complement receptors. J. Exp. Med.,141: 1163,1975. 87. Schlossman SM, Chess L, Humphreys RE, and Strominger JL: Distribution of la-like mole- cules on the surface of normal and leukaemic cells. Proc. Natl. Acad. Sci., 73:1288,1976. 88. Serrou B, Touraine JL, Caraux J et al.: Attempted characterization of a sub-population of human T-lymphocytes with Fc-receptor and la-like antigen responsible for Con-A res- ponsiveness, ADCL and MLC. Human lymphocyte differentiation, its application to can- cer. INSERM Symposium No. 8. Ed.: Serrou B and Rosenfeld C. Elsevier/North-Hol- land. Biomedical press, 101,1978. 89. Shortman K: Separation of different cell classes from lymphoid organs. II. The purifi- cation and analysis of lymphocyte populations by equilibrium density centrifugation. Austr. J. Exp. Med. Sci., 46: 375,1968. 90. Siegal FP, Pernis B and Kunkel HG: Lymphocytes in human immunodeficiency states: a study of membrane-associated immunoglobulins. Eur. J. Immunol., 1:482,1971. 91. Siegal, FP, Voss R, Al-Mondhiry H, Polliack A, Hansen JA, Siegal M, and Good RA: Association of a chromosomal abnormality with lymphocytes heaving both T and B markers in a patient with lymphoproliferative disease. Am. J. Med., 60: 157,1976. 92. Siegal FP and Good RA: Human differentiation markers and their application to immune deficiency and lymphoproliferative diseases. In: Disorders of and lym- phoid function. Clin. Haemat. 6 (Fudenberg HH, Ed.) Saunders, London, 355, 1977. 93. Stathopoulos G, and Elliott EV: Formation of mouse or sheep rosettes by lymphocytes from normal and leukaemic individuals. Lancet, I: 600,1974. 94. Uhr JW: Passive sensitization of lymphocytes and macrophages by antigen-antibody complexes. Proc. Natl. Acad. Sci. USA, 54: 1599,1965. 95. Uhr JW and Vitetta ES: Synthesis, biochemistry and dynamics of cell surface immuno- globulin on lymphocytes. Fed. Proc., 32: 35,1973. 96. Unanue ER: The regulatory role of macrophages in antigenic stimulation. Adv. Immunol., 15:95,1972. 97. V r MS, Bianco C and Nussenzweig V: Enhanced binding of neuraminidase-treated p erythrocytes to human T lymphocytes. Blood, 42: 939,1973. 98. trcrnet P: Human la-type allo-antigens: methods of detection, aspects of chemistry and biology, markers for disease states. Transplant. Rev., 30: 271,1975. 99. Williams N, Moore MAS, Shortman K, Condon L, Pike B, and Nossal GJV: Density separa- tion and characterization of human blood leukocytes including T and B lymphocytes. Austr. J. Exp. Biol. Med. Sci., 52: 491,1974. 100. Wilson AB, Haegert DG and Coombs RRA: Increased sensitivity of the rosette-forming reaction of human T lymphocytes with sheep erythrocytes afforded by papain treatment of the sheep cells. Clin. Exp. Immunol., 22: 177,1975. 101. Wilson JD and Nossal GJV: Identification of human T and B lymphocytes in normal peripheral blood and in chronic lymphocytic leukaemia. Lancet, II: 788,1971. 102. Winchester RJ and Ross G: Methods for enumerating lymphocyte populations. In: Manual of clinical immunology (Rose NR and Friedman H, Eds.) Am. Soc. Microbiol., 64,1976. 103. Winchester RJ, Ross GD, Jarowski CI, Wang CY, et al.: Expression of la-like antigen molecules on human granulocytes during early phases of differentiation. Proc. Natl. Acad. Sci., USA, 74: 4012,1977. 104. Worton RG, McCulloch EA, and Till JE: Physical separation of hemopoietic stem cells from cells forming colonies in culture. J. Cell Physiol., 74: 171,1969. 99

105. Wybran J, Carr MC, and Fuder.berg HH: The human rosette-forming cells as a marker of a population of thymus-derived cells. J.Clin. Invest., 51: 2537,1972. 106. Yoshida TO and Andersson B: Evidence for a receptor recognizing antigen-complexcd immunoglobulin on the surface of activated mouse thymus lymphocytes. Scand. J. Im- munol., 1:401,1972. 107. Yu DTY, Peter JA, Paulus HE, and Nies KM: Human lymphocyte subpopulations: study of T and B cells and their density distribution, din. Immunol. Immunopathol., 2: 333, 1974. 108. Zier KS, Huber CH, and Braunsteiner H: Linear density gradient separation of human lymphocyte subsets. Eur. J. Immunol., 7: 366,1977. 109. Zipursky A, Bow E, Seshadri RS, and Brown EJ: Leukocyte density and volume in normal subjects and in patients with acute lymphoblastic leukaemia. Blood, 48: 361,1976. 110. Zucker RM, and Cassen B: The separation of normal human leukocytes by density and classification by size. Blood, 34: 591,1969. 100

Chapter 3

PURIFIED CFU-C SUSPENSIONS OBTAINED FROM HUMAN PERIPHERAL BLOOD; SEVERAL "IN VITRO" CHARACTERISTICS

3.1 INTRODUCTION

Circulating haemopoietic stem cells undoubtedly play a role in the re- generation of the bone marrow after local damage (27, 44). However, little is known about the properties and physiologic importance of cir- culating stem cells and about the control of their appearance in and disappearance from the circulation (70). In the mouse, circulating stem cells differ from bone marrow stem cells with regard to several properties (25). These differences might be interpreted as the result of each haemopoietic compartment having its own type of stem cell, developing under the influence of micro-environ- ment characteristics of each compartment. This assumption however, fails to explain why circulating stem cells which, according to present knowledge, mainly develop in and migrate out of the bone marrow (22, 50), should differ from bone marrow stem cells. Recent investigations suggest that bone marrow stem cells might be a heterogeneous population (24, 64). Stem cell fractions of various den- sities can be isolated by cell fractionation methods, which fractions are different in regard to cell renewal capacity and turnover rate. Another explanation, apart from acquired changes in the stem cell in the peri- pheral blood, could be that migration from the bone marrow into the peripheral blood occurs selectively and not at random (25). The present experiments were designed to investigate some properties of CFU-c circulating in the peripheral blood of man. The cell suspensions studied were enriched for CFU-c and depleted for monocytes, T- and B- lymphocytes. CFU-c properties such as radiosensitivity and the fraction of the population in S-phase of the cell cycle were studied. These proper- ties were chosen since they have been extensively studied and are well known for bone marrow CFU-c. Furthermore, the CFU-c-enriched null cell suspensions were analysed for 101

their cytological aspects, cell volume distribution and immunologic capacities such as lymphocyte transformation and mixed lymphocyte culture.

3.2 MATERIALS AND METHODS

3.2.1 Cytochemical staining techniques of cytocentrifuge smears For preparation of cytocentrifuge smears, the cell concentration of the samples was adjusted so that a monolayer of cells was obtained on the glass microscopic slides. Cell smears were processed for May-Griinwald- Giemsa and cytochemistry (Sudan Black B, periodic acid schiff (PAS)), using standard techniques as described by Dacie and Lewis (15). The non-specific esterase reactions were performed according to Loffler (40) for a-naphtyl-acetate-esterase and Ornstein et al. (61) for ot-naphtyl- buty rate-esterase.

3.2.2 Lymphocyte transformation tests and mixed lymphocyte culture Lymphocyte transformation tests (LTT) 0.1 ml aliquots of the cell suspension under study (3 x 106 MNC/ml) were mixed with 0.1 ml of diluted mitogen in flat-bottomed micro- culture plates and cultured for two days at 37°C in a humidified 5% CO2 atmosphere. Cultures were pulse-labelled with 2 juCi 3H-TdR (specific activity: 2 Ci/mmol, the Radiochemical Centre, Amersham, England) in 50 fil medium (RPMI-1640, Gibco) and harvested onto glass fibre filters. Incorporated 3H-TdR was measured in a liquid scintillation counter. Each test was performed in triplicate, and in a few cases cultures in duplicate. Phytohaemagglutinin (PHA, Burroughs, Wellcome, London, England) was used as a mitogen in a final dilution of 1:400. I In some of the LTT experiments monocytes were added to the null cell suspensions. These monocytes were isolated by adherence to plastic petri dishes and removed by exposure to lidocaine as described by Rinehart (63). The monocytes were added to the null cell suspensions in a ratio of 1:80. The difference in c.p.m. (counts per minute) between test and control cultures represents the net response of the cells to mitogenic stimulation. Results were calculated as mean c.p.m. ± 1 standard error of the mean. 102

Mixed lymphocyte culture (MLC) 0.1 ml aliquots of responder cells at a concentration of 4 x 10s cells/ml were mixed in round-bottomed plates with 0.05 ml (2 x 106 MNC/ml) of 2,000 Rad irradiated stimulator cells, obtained from single donors (X, Y, or Z) or a lymphocyte pool (containing cells from at least three different donors). The medium used was RPMI-1640 (Gibco) supple- mented with 20% (complement-inactivated) AB-serum. In control cul- tures, lymphocytes were cultured in the presence of irradiated autologous cells. All cultures were performed in triplicate. The cells were cultured for four days and further processed as described for the LTT.

3.2.3 Determination of the turnover rate of CFU-c with the 3-H-thymi- dine suicide technique To determine the proliferative rate of CFU-c the tritiated thymidine suicide technique as described by Iscove et al. (30), with minor modifi- cations, was used. One ml of the cell suspension containing 0.5 x 106 MNC was incu- bated with 1 ml of serum-free culture medium (RPMI-1640) containing 12.5 juCi of 3H-TdR (methyl-3H-thymidine, specific activity 24 Ci/mmol; The Radiochemical Centre, Amersham, England) at 37°C for one hour and occasionally mixed. Incubations of cell suspensions in a serum-free culture medium containing 12.5 juCi of 3H-TdR and 100 /ug of non- radioactive thymidine and of cell suspensions in similar medium without both radioactive and non-radioactive thymidine were performed in parallel as control experiments. At the end of the incubation period, 5 ml of cold culture medium containing 10% fetal calf serum (FCS) and 100 jug/ml non-radioactive thymidine was added to stop the incorporation of 3H-TdR into the cells. Following sedimentation by centrifugation, the cells were washed twice with 3 ml medium containing non-radioactive thymidine (10^g/ml) and 10% FCS. The washed cells were resuspended in 1 ml of culture medium + 10% FCS and further processed for CFU-c culture according to the technique described in section 2.1.17. In our study three more control cell suspensions were included: 1) Cells in serum-free culture medium, containing 100 jug of non-radio- active thymidine, which were further processed as described above, and 2) Cells in serum-free culture medium, which were washed with RPMI- 1640 medium plus 10% FCS to control for the effect of non-radio- active thymidine addition. 103

3) Cells in serum-free culture medium, which were not incubated but processed for CFU-c culture immediately to control for the incuba- tion effect. The percentage of CFU-c kill (= % of CFU-c in S-phase) was determined as follows:

3 control— H-TdR exposed , , , mean CFU-c counts .nnn* ;—- ; where control = ? = 100%. control 5

3.2.4 Determination of CFU-c radiosensitivity (Do-value) Radiosensitivity was determined by the fractional survival of in vitro irradiated CFU-c. For that purpose 0.2 ml of the MNC-suspension (1 x 106 MNC in 1 ml icecold medium to avoid hypoxia) in plastic tubes (12 x 75 mm, Falcon 2058, Falcon Plastics, Oxnard, California, U.S.A.) were irradiated with various doses of 6 ° cobalt gamma-rays in a homogeneous field at room temperature. The irradiation was performed with a 240 kV X-ray machine, 15 mA tube current, HVL of 3.1 mm Cu; exposure dose rate of 600 Rad/min. The unirradiated control suspensions were kept at room temperature for periods equal to the time periods required for irradiation. The control and irradiated cell suspensions were then processed for CFU-c culture, ac- cording to the technique described in section 2.1.17. The CFU-c counts from irradiated samples were expressed as a percent- age of the number of colonies counted from the cultured non-irradiated cell suspensions. The characteristic response of tissue culture cells exposed to graded doses of X-radiation is termed a survival curve. An initial "shoulder" oi region of low slope is seen, followed by a more rapid decrease of the surviving fraction which is exponential within experimen- tal error. Although many mathematical relationships have been used to fit such survival curves, one is as follows: kD n S=l _(1 _e- ) where S is the surviving fraction; D, dose in Rads; k, slope of the expo- nential part of the curve; n, extrapolation number. The value of n is obtained by extrapolating the exponential part of the curve to its intersection with the ordinate. The reciprocal of k, the D3 7 or Do value, represents the dose necessary to reduce the surviving frac- tion S by a factor of 0.37 on the exponential part of the curve. These parameters are used to characterize and to compare the response in dif- ferent cell systems. 104

Although all mammalian cells in tissue culture show, in general, the same curve, individual variations can be marked. Also the equation given must not be construed to indicate other than a satisfactory fit of the experimental data shown. The shoulder on the curve is usually taken to indicate that damage must be accumulated before a cell can be lethally affected. However, the exponential nature of the curve indicates that, in principle, there is a probability of some cells remaining viable even after relatively high doses. Perhaps most important, the curve indicates the proiiferative capacity of the individual cells. Since the cultures are scored several days after plating, the colonies indicate only those cells capable of continued and sustained growth. The "surviving" fraction does not include cells that cannot divide, cells that can divide only once or twice, or the remains of cells that are actually dead.

3.2.5 Cell volume analysis Cell volumes were determined electronically by pulse-height analysis, using an electronic cell counter (Coulter counter®, model F, Coulter Electronics, Harpenden, Hertfordshire, England) in combination with a 64-channel pulse-height analyser (Coulter®, model P64) and an X-Y plotter. The coulter counter, with a 100 mju aperture tube, was adjusted to a count range of 512, while optimal conditions for volume analyses were the attenuator at 1, the transducer aperture at 16, and the threshold at 16. A second volume distribution profile was made from each sample, using an attenuation of 0.7 and an aperture of 32. The volume distribution analysis was preceded by calibration of the multi-channel analyser and coulter counter, using human red blood cells of a known mean corpuscular volume (MCV). The cell sample under study (20 Ml cells; 10 x 106 cells/ml) was diluted in 10 ml coulter Isoton®, so that the cell density was 0.2 x 106 cells/10 ml Isoton®. Red blood cells were previously removed from the test sample by isotonic lysis and not by saponin because this agent causes shrinkage of leucocytes (57, 75). The fraction of cells, which corresponded with a certain volume (= channel), is expressed as a percentage of the peak value. The lymphocytes showed a single distribution curve, from which the peak (= modal) volume could be read by eye. 105

3.2.6 Statistical methods Differences in experimental results were analysed with the Student's t-test for paired observations, and with a non-parametric Rank-test (Wilcoxon Mann Whitney). Results of experiments in which counts/ min. (3H-TdR) were involved, were analysed using the signed Rank-test (Wilcoxon one-sample test). Radiation survival parameters were obtained by least-squares linear regression analysis. All values are given as the a- rithmetical mean ± s.e.m. P-values less than 0.05 were considered as significant.

3.3 RESULTS

3.3.1 Cytological aspects of "CFU-c purified" null cell suspensions In the May-Griinwald-Giemsa staining, the lymphocytes revealed round nuclei with condensated chromatin and a rather abundant cytoplasm. The majority of the lymphocytes had a blue-grey cytoplasm with no granulation or vacuoles. Compared to a rarely present eosinophil, the lymphocytes were rather large, with an estimated diameter of 12-15 mju. Two distinct populations were possibly present; a large lymphocyte with blue cytoplasm and a fragile chromatin structure (minority) and lymphocytes with blue-grey cytoplasm with a more condensated chro- matin structure. The null lymphocytes were negative in the peroxidase and PAS- staining, while in the latter not even a tinge was present. The acid phos- phatase staining revealed the presence of 5-7% T-lymphocytes by dense unipolar localised dot-like positivity, which was also observed in the a-naphtyl-esterase staining. In the latter staining, some cells revealed a scattered granular positivity, which is in accordance with their B-lym- phocyte nature. The a-naphtyl-butyrate-esterase staining revealed no positivity whatsoever. With these used staining techniques no specific feature of null lympho- cytes could be demonstrated.

3.3.2 Null cells: their capacity to respond to PHA; and MLC reactivity In this experiment null cell suspensions were studied with respect to their proliferative response to PHA, and compared to unseparated MNC- and 106

T- plus B-lymphocyte suspensions. The latter cell suspension was ob- tained from the pellet after the second cell separation step, as described in section 2.3.13. Null cell suspensions obtained from 6 healthy volunteers were studied; the characteristics of these suspensions are summarized in Table 3.1. As can be seen, the percentage of C3-bearing lymphocytes was also deter- mined with the EAC-RFC technique*.

Table 3.1: In vitro characteristics of the 'null cell1 suspensions, used for MLC-tests and PHA-stimulation (N=6)

E-RFC % • 4.7± 1.1 EAt-RFC% 5.1±2.4 EA3-RFC% . 16.9±7.4 EAC-RFC% 14.4+2.2 B-RFC % 2.5±0.6 SIg(+) cells % 1.2±0.2 Monocytes % 0.1±0.1 Granulocytes % 3.5± 1.0 CFU-c/5xlO4 15.7±3.1

(All values as mean % ± 1 standard error (s.e.m.))

From 6 individuals, triplicate counts per experiment were pooled and expressed as mean counts per min. (± s.e.m.). The results are presented in Fig. 3.1. The "control" MNC suspension yielded a count of 30,352 (± 3,772), which is significantly higher than the counts obtained for both the null cell and null cell plus monocyte suspensions (p < 0.05). No dif- ference (p = 0.3) was found between the null cells and null cells plus monocytes. This mixing experiment was done because PHA-responding cells are supposed only to give maximal response in the presence of monocytes. This was confirmed by the fact that the mean counts for T- plus B-lymphocytes (monocyte depleted) were lower (p < 0.05) than those obtained from the control MNC suspension. However, it should be noted that some monocytes were still present in the T- plus B-lymphocyte cell suspension as well as in the null cell suspensions. It can be concluded that the CFU-c enriched null cell suspensions have a low reactivity to PHA, which is, according to some investigators, one of the parameters for acute GVHD initiating cells (2, 16). Next the capacity of null cell suspensions to respond and stimulate in

•Using the EA2 -RFC technique with addition of mouse-complement (1:5 diluted). 107

Fig. 3.1: Proliferative response to phytohaemagglutinin (PHA) stimulation of various peripheral blood mononuclear cell suspensions.

cprn 3 (x10 ) H^j Null cells 36 H I ) Null cells + monocytes

T+B-lymphocytes 30- „ Control" mononuclear cells

20-

10-

Counts per min. (x 103) expressed as the mean (± s.e.m.) of pooled triplicate experiments from 6 individuals. the MLC reaction was investigated. In this experiment three different ceil suspensions from each of 6 blood donors were used: null cells, T- plus B-lymphocytes, and a "control" MNC suspension. All the triplicate or duplicate counts obtained from stimulation of or response to single individuals (X, Y, Z) or cell pools were taken together and expressed as mean counts per min. (± s.e.m.). The results are presented in Fig. 3.2. For the three cell suspensions studied, no differences in the response capacity in the MLC were observed. However, the capacity to stimulate in the MLC for null cells was found to be very high: 21,821 (± 7,205) counts/min., which is significantly higher than the counts obtained for T- plus B-lymphocytes (p < 0.05) or the "control" MNC suspensions (p < 0.05). No differences were found between the T- plus B-lymphocytes and the "control" MNC suspensions with regard to their capacity to stimulate in the MLC. 108

Fig. 3.2: Response and stimulatory capacity in the mixed lymphocyte culture (MLC)-test of various peripheral blood mononuclear cell suspensions.

Q Null cells cpm P^ T + B - lymphocytes (x1 30 (1 ..Control" mononuclear t—J cells

20-

10 -

-=i 1

Res.cap. Stim.cap.

Counts per min. (x 103) expressed as the mean (± s.e.m.) of pooled triplicate experiments from 6 individuals. Stimulation/response to single individuals and cell pools (from at least 3 individuals).

To investigate this high stimulating capacity of null cells in more detail two experiments were performed. First it was of interest to know if this high stimulating capacity was bound to a threshold effect. Therefore, null cells and "control" MNC from two individuals were tested against a third party. The number of responding cells was kept constant (40 x 103 per well), whereas the number of stimulator cells varied from 1.25 to 160 x 103. The results are presented in Fig. 3.3. It is obvious that the "control" MNC suspensions showed a threshold up to 10 x 103 stimulator cells, after whic/i an exponential increase in counts was observed (Y = — 17,863 + 17,643 log X). Null cell suspensions, however, showed an immediate stimulating effect starting at 1.25 x 103 cells and a plateau at 80 x 103 cells. The regression of concentration of null cells against counts/min. was calculated to be Y = 394 + 16,289 log X. The slope of this line was not statistically different from the line for control MNC (calculated for additions of 10 x 103 or more). From the difference in intercepts it was clear that for comparable numbers of stimulator cells the counts for null cells did have a constant higher value 109

Fig. 3.3: Dose-response curves of the stimulatory capacity of null cells and "control" mono- nuclear cells in the mixed lymphocyte culture (MLC)-test.

cpm (x103) 30 -t

20 •

10-

1.25 2.5 5 10 20 <0 Stimulator cells* (x103)

Counts permin. (x 103) of responding cells from two individuals are expressed as the mean (± s.e.m.). Respouder cell numbers were kept constant (40 x 103 per well), while stimulator cells (* = irradiated) from one individual (• • = null cells; A A = control mononudear cells; o o = background) were increased from 1.25 to 160 x 103 per well.

than for "control" MNC suspensions (p < 0.05). Therefore, for the range studied, (1,25 to 80 x 103 "null" stimulator cells) no threshold effect was observed. The second experiment was done to find out if the high stimulatory capacity of null cells in the MLC could be caused by elimination of in- hibiting cells (« suppressor cells) during the cell separation procedure. For that purpose 40 x 103 irradiated null cells from one donor were allowed to stimulate 40 x 103 MNC of both individual X and Y, while an increasing number of T-lymphocytes were added. These T-lympho- cytes were obtained from the pellet after E-RFC sedimentation and were from the same individual from whom the stimulating null cells were obtained. All experiments were performed in triplicate and the mean value for individual X plus Y is graphically represented in Fig. 3.4. The mean counts, plotted against the number of T-cells added, showed a rather horizontal course (regression equation Y = 17.08 + no

Fig. 3.4: The effect of T-lymphocyte addition to the stimulatory capacity of null cells in the mixed lymphocyte culture (MLC)-test.

0 *

added T cells* (x 103 )

Counts per min. (x 103) of responding cells from two individuals are expressed as mean values. Responder cell and irradiated stimulator null cell numbers were kept constant (40 x 103 per well), while added T-cells (* = irradiated) from the "null cell" donor [A A = responder cells + (sti- mulatory null cells + added T-cells), x x = responder cells + (added T-cells], were increased from 0 to 40 x 103 per well.

0.0514X). If a suppressor cell effect should be present, a rather steep negative slope should have been found. However, the slope of 0.0514 is not significantly different from 0, when tested with the 95% confidence limit. When increasing numbers of isolated T-lymphocytes, in the ab- sence of null cells, were mixed with a fixed number (40 x 103) of responder cells, a dose-related increase in responding capacity was ob- served (Y = 0.1985 + 0.2376X, r = 0.99), meaning that isolated lympho- cytes, to some extent, are capable to stimulate in the MLC-reaction. Ill

3.3.3 Determination of the turnover rate of CFU-c with the 3H-thymi- dine suicide technique Drew and Painter demonstrated that cells exposed to high activities of 3H-TdR during DNA-synthesis, may undergo a loss of colony-forming ability, owing to the lethal effect of the incorporated thymidine (19). The CFU-c survival fraction after the 3H-thymidine suicide is an of the number of CFU-c in S-phase of the mitotic cycle. With this tech- nique the CFU-c present in null cell suspensions of eight healthy adults were evaluated. The null cells were obtained by the technique described in section 2.3.12. In addition, the proliferative state of CFU-c in peri- pheral blood and bone marrow was compared by studying the fractional survival of bone marrow CFU-c from four healthy donors. For logistical reasons it was not possible to obtain bone marrow and peripheral blood suspensions from the same donors. To make the bone marrow and peri- pheral blood null cell suspensions mutually comparable, mononuclear bone marrow cells were cultured after Ficoll-Isopaque (ds. 1.077 g/ml) gradient centrifugation. By this procedure the majority of mature mye- loid cells, which have an established inhibitory effect on CFU-c growth in vitro (4, 7, 8), were removed. Furthermore, we were interested to see if additional removal of monocytes from the bone marrow mononuclear cell suspension would alter the fraction of surviving CFU-c. The results of this experiment are presented in Fig. 3.5. For bone marrow A (marrow cells obtained from the interphase after Ficoll-Iso- paque, ds. 1.077 g/ml, centrifugation) 52.1 ± 6.5 CFU-c per 2 x 104 cells plated were found. For bone marrow B (the same processing as bone marrow A, but with monocyte elimination by carbonyl-iron pre-incuba- tion) a significantly higher (p < 0.05) number of CFU-c were found: 102.8 ±11.9 per 2 x 104 cells plated. The abovementioned values were obtained from 5 triplicate cultures from each of four normal bone marrows. After 3H-TdR suicide the fractional survival for bone marrow A was 85.0 ± 4.3%, for bone marrow B a value of 80.5 ± 5.5% was found. The difference between these two fractional survival percentages was not significant. For the null cell suspensions a fractional survival of 94.6 ± 3.1% was found, which is significantly higher (p < 0.05) than the fractional survival for bone marrow A and B. It was stated by Liu et al. (39) that the fractional survival of clusters was higher than for CFU-c in human peripheral blood. Therefore the 3H- 112

Fig. 3.5: Percentage of CFU-c in DNA-synthesis, estimated as the CFU-c surviving fraction after 3 H-TdR-suicide in vitro of human bone marrow and peripheral blood null cell sus- pensions.

CFu-C Control surviving fraction 3H-TdR

100

80 I

60-

40- I 20- I •/'•' 1 ''A A B Null-cells Bone marrow

Surviving fractions after 3 H-TdR-suicide expressed as mean (± s.e.m.) percentages, compared to control (= 100%) experiments. Bone marrow A: mononuclear bone marrow cells of 4 individuals, collected from the interphase after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation. Bone marrow B: mononuclear bone marrow cells from the same 4 individuals, collected from the interphase after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation after carbonyl- iron incubation.

TdR suicide for clusters (cell aggregates > 3 < 20 cells) in null cell suspensions was also tested. A mean of 2.5 ± 0.2 clusters per 5 x 104 cells plates was observed in vitro cultures of null cell suspensions. The fractional survival of these clusters was found to be 91.7 ± 5.1, which is not significantly different from the fractional survival for CFU-c present in peripheral blood. The fractional survival of clusters for both bone marrow A and B was found to be 90.2 ± 3.0, which is not significantly different from the fractional survival of CFU-c in bone marrow and from clusters or CFU-c in peripheral blood.

3.3.4 Determination of CFU-c radiosensitivity (Do-value) In our experiments the radiosensitivity parameters for human peripheral 113

progenitor cells were studied and compared to those obtained from nor- mal human bone marrow. From four healthy donors, mononuclear bone marrow cells (A) were collected from the interphase after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation, while from the same donors monocyte-free bone marrow cells were obtained by pre-incubation with carbonyl-iron and subsequent Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation (B). The individual survival curves of bone marrow (A), after irradiation with various doses of 6 ° cobalt gamma-rays (dose rate 42 Rad/min.) are presented in Fig. 3.6, while the cumulative data for bone marrows A and

Fig. 3.6: Radiation-survival curves of bone marrow CFU-c from 5 individuals. Bone marrow samples were irradiated in vitro with * °Co-7-rays (dose rate 42 Rad/min).

CFu-c surviving, fraction () 100 -*£:'"

50-

20-

10-

5 -

33 67 100 133 167 200 Rad Abscissa: irradiation dosage (in Rads) to which the bone marrow samples were exposed. Ordinate: Fractional survival of CFU-c expressed as percentage of the "control" non-irradiated samples. Each point indicates the individual surviving fraction at each level of irra- diated dosage. 114

B are presented in Fig. 3.7. The calculated Do-values for CFU-c and clusters as well as the various extrapolation numbers (n) are summarized in Table 3.II.

Fig. 3.7: Mean radiation survival curves of bone marrow CFU-c from 4 individuals. Bone mar- row samples were irradiated in vitro with • °Co-7-rays (dose rate 42 Rad/min).

CFu-c surviving fraction (%)

100 -1

50-

20-

10 H

5 -

33 67 100 133 167 200 Rad

Abscissa: irradiation dosage (in Rads) to which the bone marrow samples were exposed. Ordinate: fractional survival of CFUc, expressed as percentage of the "control" non-irradiated samples. Each point represents the mean (± s.e.m.) surviving fraction at each level of irradiation dosage. • • : mononuclear bone marrow cells. o o: mononuclear bone marrow cells, from which the monocytes were removed.

A mean CFU-c Do-value of 82.9 ± 10.7 was found for bone marrow A with an n-value of 3.5 ± 0.7. Bone marrow clusters were much more radioresistant than CFU-c: Do 191.2 ± 28.7, with an n-value of 1.5 ± 0.1. 115

Table 3.II: Radiosensitivity of human bone marrow CFU-c/clusters. 60Co-irradiation (42 Rad/min.)

Do-value* (Rad) Extrapolation number** (n) # bone marrow sample CFU-c clusters CFU-c clusters

1 52.1 103 6.1 1.8 2 83.1 199 3.2 1.7 A 3 83.6 264 2.5 1.3 4 113.0 199 2.3 1.4

mean ±s.e.m. 82.9±10.7 191128.7 3.5±0.7 1.510.1

101.9 280 1.1 1.4 109.8 367 1.3 1.0 143.1 389 1.1 1.7 145.6 295 1.2 1.6

mean Is.e.m. 125.1+9.7 332+23.1 1.1+0.04 1.4+0.1

A = Inter phase bone marrow cells after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation B= Interphase bone marrow cells after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation, with initial carbonyl-iron incubation

*,** Do-value = — ; n = ; both derived from the calculated linear regression slope 100 line of e?ih individual survival curve (irradiation range 100-200 Rad)

These differences are significant when tested with Student's t-test for paired observations (p < 0.05). The same differences between the Do- value of CFU-c and clusters were found for bone marrow B (p < 0.05), but no significant difference was found between the extrapolation num- bers for CFU-c and clusters. When bone marrow A was compared to bone marrow B by Student's t-test for paired observations, a lower radiosensitivity for CFU-c and clusters for bone marrow without monocytes was found (p < 0.05). Also a lower extrapolation number for CFU-c was found (p < 0.05), but there was no difference for the extrapolation number of clusters. So the remo- val of monocytes (and also more granulocytes) by carbonyl-iron pre- incubation induced a significant alteration in the radiosensitivity of bone marrow progenitor cells. Next the radiosensitivity parameters were studied for human peripheral blood CFU-c present in null cell suspensions, as well as in control MNC suspensions. The radiosensitivity of CFU-c present in null cells was 116

Fig. 3.8: Mean radiation survival curves of peripheral blood CFU-c. The cell samples were irradiated in vitro with 60Co-y-rays (dose rate 42 Rad/min, o o and A A) and 'a 7Cs (dose rate 660 Rad/min, • •).

CFu-c surviving.fraction

Abscissa: irradiation dosage (in Rads) to which the peripheral blood cell suspensions were exposed. Ordinate: fractional survival of CFU-c, expressed as percentage of the "control" non-irradiated samples. Each point represents the mean (± s.e.m.) surviving fraction at each level of irradiation dosage. peripheral blood mononuclear cells (N = 2). peripheral blood null cells, exposed to high dose rate irradiation (N = 6). peripheral blood null cells, exposed to low dose rate irradiation (N = 5). determined with low dose rate irradiation (6 ° cobalt X-rays, dose rate 42 Rad/min.) and with high dose irradiation (137Cesium: 660 Rad/min.). This dual experiment was done to study the effect of dose rates on survival of CFU-c's irradiated in vitro. The survival curves are presented in Fig. 3.8. The individual Do-values and extrapolation numbers for CFU-c and clusters are summarized in Table 3.III. 117

Table 3. Ill: Radiosensitivity of human peripheral blood CFU-c/clusters present in null cell suspensions

Do-value* (Rad) Extrapolation number** (n) # cell suspension CFU-c clusters CFU-c clusters

1 68.3 115.0 1.0 1.0 2 55.2 85.0 1.4 1.0 3 52.2 71.0 1.1 1.2 4 65.0 77.2 1.0 1.3 5 46.4 77.3 1.0 1.0

mean ±s.e.m. 57.4±3.6 85.1±6.9 l.l±0.06 l.l±0.01

1 80.1 115.6 1.0 1.3 2 95.2 108.4 1.0 1.2 3 44.0 67.2 1.0 1.0 4 55.6 59.6 1.1 1.0 5 83.9 92.0 1.0 1.0 6 77.8 93.7 1.0 1.0

mean ±s.e.m. 72.7±7.1 89.4±8.2 1.0±0.01 1.0+.0.04

Suspensions C + D 65.7+4.8 87.4 ±5.5 1.0+0.03 1.0+0.01 mean ±s.e.m.

1 84.2 108 E 2.3 1.6 2 73.9 217 1.9 1.0

mean ±s.e.m. 79.0±3.6 162.5±38.5 2.1±0.1 1.3±0.2

C = null cells, irradiated with 60Co (42 Rad/min.) D = null cells, irradiated with 137Cs (660 Rad/min.) E = control MNC, cells irradiated with 60Co (42 Rad/min.) N.B.: group C, D and E are different individuals. 1 2 72'ntercept *.** Do-value =• ; both derived from the calculated slope ' n" =• 100 linear regression line of each individual survival curve (irradiation range 100-200 Rad)

For low dose irradiation a mean Do-value of 57.4 ± 3.6 for CFU-c was found, while the Do-value for clusters was 85.1 ± 6.9 (p < 0.05). No dif- ferences were found for the extrapolation numbers. For high dose irra- diated CFU-c and clusters Do-values of 72.7 ±7.1 and 89.4 ± 8.2 were found respectively (p < 0.05). As observed before with bone marrow, the clusters were more radioresistant than CFU-c. In analogy with the 118

Fig. 3.9: Mean radiation survival curves of peripheral blood CFU-c from two individuals. The cell samples were irradiated with 60Co-7-rays (dose rate 42 Rad/min). Differences in the low dose irradiation-range for CFU-c survival.

CFu-c surviving fraction (%)

8 16 24 33 50 67 Rad.

Abscissa: irradiation dosage (in Rads) to which the peripheral blood cell suspensions were exposed. Ordinate: fractional survival of CFU-c, expressed as percentage of the "control" non-irradiated samples. Each point represents the mean (± s.e.m.) surviving fraction at each level of irradiation dosage. peripheral blood mononuclear cells. peripheral blood null cells. peripheral blood T- + B-lymphocyte suspensions. irradiation survival data, no differences were found between the extra- polation numbers for CFU-c and clusters. When the Do-values and extra- polation numbers for CFU- and clusters obtained from the low irradia- tion dose group were compared to the high irradiation dose group with Student's t-test for two observations, no significant differences were found. Hence, for both groups together the calculated Do-value for CFU-c was 65.7 ± 4.8 with an extrapolation number of 1.0 ± 0.03, while for clusters a Do-value of 87.4 ± 5.5 and an extrapolation number of 1.0 ± 0.01 was found. In Table 3.IV the collected data from bone marrow and peripheral blood are summarized; the overall picture is that bone marrow CFU-c 119 and clusters have higher Do-values and extrapolation numbers than found for CFU-c and clusters in peripheral blood null cell suspensions. From Table 3.Ill it is clear that the control MNC suspensions did not differ in their mean Do-value for CFU-c, when compared to CFU-c present in null cells, but had a higher mean extrapolation number (p < 0.05). So the removal of monocytes results in lower extrapolation numbers as was already observed for bone marrow. For clusters in two "control" MNC suspensions a higher Do-value (p < 0.05) was found when compared to null cells, but no difference in extrapolation numbers. Since these two "control" MNC suspensions were not derived from the null cell donors and the assays were not carried out as paired ob- servations, the following experiment was performed.

Table 3.IV: Summary of the various radiosensitivity parameters of CFU-c/clusters in human bone marrow and peripheral blood

Bone marrow (A) Null cells Differences*

Do-CFU-c 82.9+10.7 65.714.8 NSD n-CFU-c 3.5±0.7 1.0+0.03 p<0.05 Do-clusters 191.2±28.7 87.415.5 p<0.05 n-ctusters 1.510.1 1.0+0.01 p<0.05

Bone marrow (B) Null ceils

Do-CFU-c 125.119.7 65.714.8 p<0.05 n-CFU-c 1.1+0.04 1.010.03 NSD Do-clusters 332 123.1 87.415.5 p<0.05 n-clusters 1.4 + 0.1 1.0+0.01 p<0.05

Bone marrow A = Interphase bone marrow cells after Ficoll-lsopaque (ds. 1.077 g/ml) centrifu- gation Bone marrow B = Interphase bone marrow cells after Ficoll-lsopaque (ds. 1.077 g/ml) centrifu- gation, with initial carbonyl-iron incubation All values as: mean 11 standard error (s.e.m.) * tested with Student's t-test for two observations.

To substantiate the different radiosensitivity of CFU-c after monocyte removal in the low dose irradiation range, buffy coat cells of two normal donors were processed to obtain 3 cell suspensions. 1) Interphase MNC after Ficoll-lsopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells; 120

2) Interphase MNC after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation of buffy coat cells with initial carbonyl-iron incubation (removal of monocytes and surplus of granulocytes); 3) Interphase null cells, after combined B-RFC + E-RFC sedimentation from interphase suspension 2. Samples of these 3 cell suspensions from each of the two donors were irradiated in vitro with 8.2, 16.5, 24.7, 33.5 and 67 Rad. The results are presented in Fig. 3.9. It is clear that removal of monocytes (+ granulocytes) by carbonyl- iron incubation and an extra Ficoll Isopaque step, eliminates the shoulder in the low dose survival region. Both monocyte-free and null cells showed an exponential course and an already demonstrated CFU-c kill of about 30-40% at 8.2 Rad. The presence ot monocytes seems to protect CFU-c from irradiation damage in the low dose irradiation region.

3.3.5 Volume profile of MNC-subpopulations The final experiments on "in vitro" characteristics of CFU-c enriched null cell suspensions dealt with the volume profiles of various MNC sub- populations, obtained during the two-step cell separation technique as is described in section 2.3.13. From 6 healthy volunteers buffy coat cells were processed. Suspen- sion A contains buffy coat cells obtained after methylcellulose 2% sedi- mentation. Suspension B contains MNC after pre-incubation with car- bonyl-iron and subsequent Ficoll-Isopaque (ds. 1.072 g/ml) centrifuga- tion. Suspension C represents cell suspension B depleted of E-RFC plus B-RFC by Ficoll-Isopaque (ds. 1.072 g/ml) centrifugation, while suspension D contains the pellet cells obtained after this centrifugation step. The characteristics of these suspensions, with regard to lymphocyte subpopulations, are presented in Table 3.V. The model volume (in jtzm3) of these different cell suspensions are summarized in Table 3.VI.

Table 3.V: In vitro characti Tistics of the various cell suspensions used for MNC volume profile determination

4 E-RFC EA3-RFC EAORFC B-RFC SIg(+) cells Monocytes Granulocytes CFU-c/SxlO

1) Suspension A 53.6+6.5 22.2±4.2 29.5 + 2.4 13.2±3.9 3.9+0.6 15+2.7 26.5+3.8 0.8+0.2 2) Suspension B 68.6+4.8 17.6 + 3.3 24+0.7 7+2 3.7±1 0.3+0.2 13.5±3.1 1.1 + 0.3 3) Suspension C 2.1 ±0.4 30.2+4.9 13.5 + 1 3.6±2.S 0.1+0.1 0 5±2.1 I0.S+ 2.8 4) Suspension D 70.7+6.5 I7.g±3.5 27+5.6 8.8+2.4 3+0.6 0.5±0.1 15+3.2 0.6+0.2

1) Suspension A = buffy coat cells obtained after methylcellulose 2% sedimentation 2) Suspension B = MNC after pre-incubation with carbonyl-iron and Ficoli-lsopaque (ds. 1.077 g/ml) cenlrifugation 3) Suspension C = suspension B, after removal of B-RFC + E~RFC by Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation 4) Suspension D = pellet cells obtained after centrifugation step 3. All values as mean % ± 1 standard error (s.e.m.) 121

Table 3.VI: Modal lymphocyte volumes (in (Mm ) determined from various types of cell suspensions (N=6 donors)

Coulter- Type of cell suspension Donor # adjustment* A B C D 1 240 247 253 214 I 2 237 256 265 256 1 - 227 279 221 II 2 237 292 219 1 - 247 279 253 III 2 247 283 256 1 274 247 256 IV 247 2 229 237 247 237 1 227 247 265 V t o t o 2 228 257 270 0 " - 1 221 240 286 240 VI 2

modal volume 230±2.1 244+2.5 270±4.1 235±4.5 (±s.e.m.)in jttm3 (N=7)

Suspension A = Buffy coat cells obtained after methylcellulose 2% sedimentation Suspension B = MNC after pre-incubation with carbonyl-iron and Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation Suspension C = suspension B, after removal of B-RFC + E-RFC by Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation Suspension D = pellet cells obtained after centrifugation step 3.

1) attenuator 1 ; aperture 16 - threshold 16 ; count range 512 2) attenuator 0.7 ; aperture 32 - threshold 16 ; count range 512 (using Student's t-test for paired observations: no difference between 1 and 2).

The modal volume (in fj.m3) for buffy coat cells (A) was found to be 230 ±2.1, while removal of monocytes and granulocytes (suspension B) resulted in an increase of the modal volume to 244 ± 2.5 (p < 0.05). After subsequent removal of T- plus B-lymphocytes, the null cell sus- pension C again demonstrated an increment in the modal volume: 270 ± 4.1, which is significantly higher than suspension A (p < 0.05) and B (p < 0.05). The pellet cells (mainly T- and B-lymphocytes) revealed a modal volume of 253.6 ± 4.5, which is not significantly different from suspension A or B, whereas it is significantly lower than suspension C (p < 0.05). 122

These results demonstrate that the CFU-c enriched null lymphocytes have a higher modal cell volume than the total lymphocyte population, as is represented by suspension B. An example of the volume profiles of the various cell suspensions obtained from one blood donor is presented in Fig. 3.10.

Fig. 3.10: Example of the different volume distribution profiles, determined from various peri- pheral blood cell suspensions, from one individual.

100 -i

90 -

80 -

70 -

60 -

50 -

40 -

30-

20 -

ID-

65 130 195 260 325 390 455 520 585 650 Volume (Aim3)

Suspension A: buffy coat cells after methylcellulose 2% sedimentation. Suspension B: mononuclear cells after pre-incubation with carbonyl-iron and subsequent Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation. Suspension C: cell suspension B, depleted of E-RFC and B-RFC by combined rosette sedimen- tation in a Ficoll-Isopaque (ds. 1.072 g/ml) gradient. Suspension D: pellet cells, after the afore-mentioned centrifugation step.

In suspension A the broad peak in ihe higher volume region represented granulocytes and monocytes, but this peak is no longer present in the volume distribution curve of suspension B. The null cell suspension (C) 123 not only had a higher modal volume, but also revealed a skewness to the right, which implied that a substantial fraction of the null cells have a very high volume. However, in this high volume region 5% granulocytes were still present, as can be seen in Table 3.V. From Fig. 3.9, it is obvious that, although a shift to the right in the modal density of null cells was observed, there was a substantial overlap with other lymphocytes. This implies that with a cell separation proce- dure on the basis of volume only, no absolute discrimination from other lymphocytes can be achieved.

3.4 CONCLUSIONS AND DISCUSSION

Conclusions

1) The lymphocytes present in null cell suspensions did not reveal a specific pattern in the various cytochemical staining techniques. 2) The CFU-c enriched null cell suspensions obtained by the two-step density separation technique, as is described in chapter 2, showed a low reactivity to PHA-stimulation, which was mainly caused by eli- mination of T-lymphocytes. This low PHA-stimulation was not only the consequence of monocyte removal, as was demonstrated by mixing experiments. The null-cell suspensions did have a normal capacity to respond in MLC-tests, but had a very high stimulating capacity. This high stimulating capacity did not show a threshold phenomenon and was not abolished by co-cultivation with isolated T-lymphocytes. Therefore, it is very unlikely that the high stimulating capacity in the MLC should have been caused by elimination of suppressor cells during the cell separation technique. 3) With the 3H-TdR suicide technique it was found that 15-20% of the CFU-c in human bone marrow were in S-phase of the mitotic cycle. The removal of monocytes (and granulocytes) from the bone marrow suspension by pre-incubation with carbonyl-iron and Ficoll- Isopaque centrifugation did not alter the fractional survival of bone marrow CFU-c's. The CFU-c present in the null cell suspension had a much lower percentage of cells in S-phase: ± 5%. However, it must be stated that statistic evaluation was not done on paired observations of bone marrow and peripheral blood cells obtained from the same donors. 124

Finally, no differences in fractional survival for CFU-c and clusters in human peripheral blood could be observed. 4) Radiosensitivity studies revealed that for both bone marrow and peri- pheral blood, clusters were much more radioresistant than CFU-c. CFU-c in human peripheral blood showed a higher radiosensitivity than CFU-c in human bone marrow, especially when compared with monocyte-depleted bone marrow. The dose rate did not alter the Do-value or extrapolation numbers. Elimination of monocytes (and granulocytes) from the test sus- pensions induced an alteration in radiosensitivity parameters. For human bone marrow this resulted in disappearance of the shoulder in the survival curves and consequently a lower extrapolation number. Moreover, the Do-values increased, which indicates a lower radio- sensitivity. Also for peripheral blood progenitor ceils the removal of mono- cytes (and granulocytes) from the test suspensions induced an alteration in radiosensitivity, especially in the low-dose irradiation region. The shoulder in the survival curve disappeared, which resulted in a lower extrapolation number. In general, it can be concluded that the radiosensivitity of haemo- poietic stem cells is strictly dependent on the presence or absence of other haemopoietic cells in the test suspensions. 5) Finally, the volume profile determination of the different MNC sus- pensions revealed that the CFU-c enriched null cells had a relatively high mean volume compared to unseparated MNC or T- + B-lympho- cyte suspensions. However, there was a substantial overlap in volume distribution with other lymphocyte populations.

Discussion

The development of semi-solid culture techniques for growing colonies of differentiating neutrophils, monocyte/macrophages and eosinophils has permitted an analysis of the properties and behaviour of individual progenitor cells in the marrow or peripheral blood of humans. A striking feature of haemopoietic colonies grown in vitro is the variability in size and shape, indicating the probable existence of a con- siderable degree of heterogeneity of progenitor cells within even one haemopoietic family. Application of physical separation techniques in mice, such as velocity 125

sedimentation, density gradient separation and cell adherence procedures, has confirmed that CFU-s and CFU-c are different cells, while it has further shown that each compartment comprises several subpopulations that differ in capacity of self replication or response to growth regulators in vitro (43, 54, 69, 73). This diversity of human granulopoietic precur- sor cells in human bone marrow, in analogy with that of the mouse, has been described in detail by Jacobson et al. (32), Johnson et al. (34) and Iscoveetal. (31). Cell separation experiments with mouse bone marrow suspensions have yielded a very detailed picture of the pluripotent haemopoietic stem cell, such as buoyant density (73) and velocity sedimentation rate (69). Despite its unique biological properties, the stem cell shares several physical properties with other cell types. Useful criteria for stem cell recognition may be found in the occurrence of specific membrane components, such as membrane surface antigens. Van den Engh et al. (20) studied mouse CFU-s and CFU-c membrane antigens by incubating mouse bone marrow with antisera of known specificity and complement and subsequently measuring the suppression on the CFU-c and CFU-s formation. The CFU-s were found to express H-2k, H-2d and an antigen associated with brain tissue (BAS). The dif- ferentiation antigens la (H-2') and Thy-1 could not be demonstrated. None of these antigens were found on the committed CFU-c. In mice sperm-associated antigens were also demonstrated on the CFU-s (36). There is no assay system for the detection of multipotent stem cells in human bone marrow or peripheral blood, so only the committed pro- genitor cells can be investigated for surface antigens. In the previous chapter it was already summarized that none of the "immunological" markers (IgG-Fc, C3, receptors for mouse and sheep erythrocytes) were present in human CFU-c. In man, the only antigens that have been proved to be present on CFU-c are HLA-antigens (23) and the la-like antigen P23.30 (13, 33, 72). However, these unique glyco- proteins are also present on most B-lymphocytes (12), Fc-receptor bearing T-lymphocytes, monocytes and macrophages (21, 72), immature myeloid cells, and erythroid precursor cells (72). Also, 25% of the megakaryo- cytes bear the P23.30 antigen, but this antigen is not detectable on platelets (72). When B-lymphocy tes are maturing to plasma cells the P23.30 antigen disappears (72). Furthermore, on the committed progenitor cells CFU-e and CFU-c, the blood group antigen i was demonstrated (28). However, with more refined serological techniques it can be expected that stem cell specific antigens will be detected in the near future, which 126

will provide the possibility of selective purification of these stem cells by fluorescence activated cell sorter techniques. The possibility to study properties of progenitor cells in man with techniques that are also avail- able for animal studies can be extended, in order to determine parallels in the biological properties of these circulating progenitor cells. The CFU-c-enriched null cell suspensions in our study demonstrated a very slight response to PHA, which is one of the parameters for the absence of acute GVHD inducing cells (2, 16). Addition of monocytes to the null cell suspensions did not enhance the stimulating effect of PHA. It has been shown by Mookerjee (52) that monocytes (macrophages) are necessary for the lymphocyte response to phyto mitogens, due to release of humoral factors into the culture supernatant fluids. The fact that no increase in PHA response was found after monocyte addition indicates that the low response to PHA was not a result of eli- minated monocyte "helper" function, but purely by the fact that in- sufficient T-lymphocytes were present. This is in complete accordance with the findings by Lohrman et al. (41). It is generally agreed that the cells responding in MLC are T-cells (1, 11, 41, 66). Conflicting evidence has been presented, however, as to which lymphoid cell subpopulation expresses MLC-stimulating deter- minants. While B-lymphocytes are generally considered capable of MLC stimulation (1, 59), conflicting reports have been published on T-lym- phocytes (59, 66). The presence of monocytes, derived from either the stimulating or responding cell population, is essential for a proliferative response to take place. Monocytes have been shown to express MLC- stimulating determinants (1). We demonstrated that null cell suspensions which are virtually depleted of T-lymphocytes, B-lymphocytes and monocytes, but have a relative high percentage of complement and IgG-Fc bearing cells, are highly stimulatory in the MLC. This could be explained by the fact that DR-like antigen-bearing cells in the null cell suspension are relatively enriched in their concentration, resulting in a higher stimulatory capacity. The fact that the null cell suspensions displayed a normal response capacity compared to unseparated mononuclear cells, is in contrast with the observations of Chess et al. (11). The high stimulatory capacity of null cell suspensions in the MLC reaction did not show a threshold phenomenon and was not caused by absence of suppressor cells, which latter is in accordance with the experi- ments of Van Oers and Zeilemaker (59). Despite the vast demands for the daily replacement of blood cells, the 127 proliferative rate of the pluripotent haemopoietic stem cell in mice (CFU-s), the ultimate source of these cells, is less than 10% (5). The high cell output is affected by two interrelated amplifying transit populations: the committed precursors and the "recognizable" precursors. Under con- ditions of haemopoietic regeneration or in cases of sudden greatly in- creased (or sustained) differentiation demands, the proliferative rate of the stem cell is increased to such an extent that 30-40% of the cells may be in DNA-synthesis (5, 14, 38). In contrast, more CFU-s in peripheral blood of mice are normally already in S-phase: 25-36.3% (25). In mouse marrow the red cell progenitor cell CFU-e showed a S-phase percentage of 74, irrespective of being tested in normal or in regenerating bone marrow (51). From progenitor cells of megakaryocytes (CFU-m) in mouse bone marrow 3040% of the population is in S-phase of the mitotic cycle (49). Most committed progenitor cells in peripheral blood do have a lower percentage of cells in S-phase, as was demonstrated for the CFU-c in dogs (58) and for mixed erythroid-granulocytic colonies in mice (35). For human bone marrow we found a percentage between 20 and 25% of bone marrow CFU-c in S-phase, which is in accordance with results found by Moore (52), Iscove (29), Rickard (62) and Jacobsen (32). Measured with hydroxyurea, approximately the same percentage of cells in S-phase was found in human bone marrow (6, 18, 55, 67). However, CFU-c from human bone marrow scored on day 7 or day 14 were not only found to have different sedimentation velocities, but also different percentages of cells in S-phase. From CFU-c scored on day 14, 21 ± 7% were killed with 3H-TdR, whereas day 7 scored CFU-c demon- strated a cell kill of 48 ± 4% (32). This observation indicates a tremen- dous heterogeneity of haemopoietic precursor cells in humans. This was further illustrated by the studies of Dresch et al. (17), who demonstrated that eosinophilic precursor cells had a lower percentage in S-phase than neutrophilic precursor cells, 20 and 50% respectively. For human peripheral circulating CFU-c we found a very low percent- age of cells in S-phase of the mitotic cycle. This is not only in accordance with studies done in animals (35, 58), but also with previous studies in humans. Tebbi et al. (68) studied 6 normal volunteers and found that 12% of colony-forming cells in the blood were in DNA-synthetic phase. Less than 15% was found by Dresch and Faille (17) for CFU-c in human peripheral blood. It is remarkable that these authors also tested umbilical cord blood in which they found 66% of the CFU-c in S-phase. Also the 128 red cell progenitor cell BFU-e in human peripheral blood shows a very low percentage of cells in DNA-synthesis (60). Our results are in contrast to those observed by Liu et al. (39), who found a 26 ± 10% reduction in colony formation after 3H-TdR exposure for human peripheral blood CFU-c. They found no difference in the magnitude of reduction in colony formation following exposure to 3H-TdR for CFU-c in blood and those in bone marrow. However, it should be emphasized that 2 out of these 6 studied persons had a higher CFU-c kill in the bone marrow than in the peripheral blood samples studied in parallel. Furthermore, these authors used peripheral blood lymphocyte suspensions, filtered through nylon-wool and subsequently centrifuged in Ficoll-Isopaque gradients. The cell composition therefore is different from that used by us. Although the monocytes and B-lym- phocytes were eliminated, T-lymphocytes were still present in their test samples. It is conceivable that the presence or absence of helper T-lymphocytes can influence the mitotic cycle of CFU-c. Variations in the number and type of MNC in the test suspensions can be responsible for the difference in percentage of cells in S-phase, as reported by several groups of inves- tigators. In this connection it has been demonstrated that mature granulo- cytes have inhibiting properties on colony formation by CFU-c (4, 7,8) and that the monocytes (or macrophages) can have stimulatory capacities by the production of colony stimulating activity (CSA) (9, 37) or inhibitory capacities by the production of prostaglandin-E (9). Therefore, it is absolutely necessary in these kinds of studies to make an exact statement of the cell composition of test suspensions. Furthermore, the degree of cells killed in S-phase by hydroxyurea or 3H-TdR is also dependent on the type of CSA in the "in vitro" culture system (10). The same strict criteria should be applied for studies on radiosensiti- vity of human CFU-c in bone marrow and peripheral blood. In this field too, the results reported are conflicting. In the most thoroughly studied animal, the mouse, irradiation experiments in vitro as well as in vivo have been performed to determine the radiosensivity of different classes of progenitor cells. For the CFU-s in the bone marrow of mice, a Do-value of 100 Rad was found by Gidali et al. (25) and 86 Rad by Nakeff et al. (56). It was found by Siegers et al. (65) that the observed Do-value for CFU-s in mouse bone marrow was related to the stage of the mitotic cycle. Cells in S-phase had a Do-value of 80 Rad (n = 1.23), while cells in all other stages of the mitotic cycle had a Do-value which was significantly higher: 185 Rad (n= 1.14). 129

The committed haemapoietic progenitor cells in mouse bone marrow have Do-values of approximately 120 Rad for CFU-c (46) and CFU-m (56). The CFU-e in mouse bone marrow have a higher radiosensitivity: Do-dose of 66 Rad. Greenberger et al. (26) observed for bone marrow CFU-c in the mouse, that the Do-value was increased from 120 to 210 Rad, if the number of plated cells per well were increased from 2 x 10s to 5 x 10s. Furthermore, McCulloch et al. (46) found that for CFU-c in the bone marrow of mice no difference in Do-value was seen when the stem cells were irradiated in vitro or in vivo, but a significant difference was ob- served in extrapolation numbers, which were higher when the stem cells were irradiated in vitro. In animals the data on radiosensitivity of haemopoietic progenitor cells in peripheral blood are scanty. Gidali et al. (25) compared the radiosensitivity of CFU-s and CFU-c in the peripheral blood of mice and found a Do-value for CFU-s in peri- pheral blood of 140 Rad, which is significantly higher than the Do-value of 100 Rad for CFU-s in bone marrow. The most relevant studies concerning human bone marrow CFU-c have been published by Broxmeyer et al. (6). Their most important observa- tion was were that the potency CSA used to stimulate cultures of irra- diated cells, determined the Do-value obtained. The higher the level of CSA, the greater the Do-value. Depending on the CSA-concentration, Do-values between 45-165 Rad for human bone marrow CFU-c were found. In contrast to observations in mice (26) they found no alterations in Do-values by higher plating doses of marrow cells per well. Secondly, they found that preferentially CFU-c in S-phase of mitosis were affected by irradiation in the lower dose region. Our observation that the Do-value for human bone marrow CFU-c was increased by removing monocytes and/or granulocytes from the test suspension, while the CSA-activity in the conditioned medium re- mained constant, implies that not only the amount of CSA is of im- portance, as was found by Broxmeyer et al. (6), but also the cell com- position of the test suspensions. From the differences found in extrapolation numbers it can be as- sumed that the presence of monocytes interferes with the lethal effect of low-dose irradiation to CFU-c in S-phase. This phenomenon was also found in experiments using peripheral blood CFU-c present in cell suspensions with or without monocytes (and granulocytes), but too few data concerning the radiosensitivity of human peripheral blood CFU-c have been reported to make a sensible comparison. 130

It is known that other MNC, such as monocytes and T-lymphocytes (9, 37, 47), can influence the CFU-c growth capacity in vitro. From animal studies it is known that both monocytes and T-lymphocytes are more radioresistant (3, 48). When present in the irradiated test suspen- sion a non-lethal radiation effect might increase the likelihood that these cells become more differentiated and/or stimulated and release stimula- tory and/or colony-inhibiting substances. As was previously stated for the 3H-TdR suicide results, an adequate description of the cell composi- tion of the test suspensions is warranted. Furthermore, it is known that other circumstances such as anoxia during the irradiation (46) as well as the number of cells in DNA-synthesis (45,65) can influence the Do-value and extrapolation number. As far as the lymphocyte volume distribution profiles are concerned our results with unseparated mononuclear blood cells are in accordance with the results published by Westring et al. (71), Loos et al. (42), Zucker et al. (75) and Zipursky et al. (74). The observation by Zipursky et al. (74), that the lymphocyte volume is inversely correlated with the cell density, is in accordance with our observation that the CFU-c-enriched null cells, which are present in the low density region (see chapter 2), have a relatively high modal volume.

3.5 REFERENCES

1. Albiechtsen D and Lied M: Stimulating capacity of human lymphoid cell subpopulations in mixed lymphocyte cultures. Scand. J. Immunol., 7: 427,1978. 2. Amato D, Cowan DH and McCulloch EA: Separation of immunocompetent cells from human and mouse hemopoietic cell suspensions by velocity sedimentation. Blood, 39: 472,1972. 3. Anderson RE, Olson GB, Autry JR et al.: Radiosensitivity of T- and B-lymphocytes. IV. Effect of whole body irradiation upon various lymphoid tissues and numbers of lecirculating lymphocytes. J. Immunol., 118: 1191,1977. 4. Baker PL, Broxmeyer HE and Galbraith PR: Control of granulopoiesis in man. HI. Inhibi- tion of colony formation by dense leukocytes. J. Cell. Physiol., 86: 337,1975. 5. Becker AJ, McCulloch EA, Siminovitch L, and Till JE: The effect for differing demands for blood cell production on DNA-synthesis by hemopoietic colony-forming cells of mice. Blood, 26: 296,196S. 6. Broxmeyer HE, Galbraith PR, and Baker FL: Relationship of colony-stimulating activity to apparent kill of human colony-forming cells by irradiation and hydroxyurea. Blood, 47: 403,1976. 7. Broxmeyer HE, Moore MAS, and Ralph P: Cell free granulocyte colony inhibiting activity derived from human polymorphonuclear neutrophils. Exp. Hemat., 5: 87,1977. 8. Bruch C, Kovacs P, Ruber E, and Fliedner TM: Studies on the inhibitory effect of granulo- cytes on human granulocytopoiesis in agar culture. Exp. Hemat., 6: 337,1978. 9. Bruch C, Kovacs P and Ruber E: The role of phagocytic cells in human blood leukocyte suspensions for in vitro colony-forming cells. Exp. Hemat., 6: 346,1978. 131

10. Byrne GV, Heit W, and Kubanek B: The proliferative states of density subpopulations of granulocyte-macrophage piogenitoi ells. Exp. Hemat., 7: 105,1979. 11. Chess L, MacDermott RP, Sondel PM, and Schlossman SF: Characterization of cells in- volved in human cellular hypersensitivity. Progress in Immunology II, Vol. 3. Ed.: L. Brent and J. Holboiow. North Holland Publishing Company, 125,1974. 12. Chess L, Evans R, Humphreys RE, Strominger JL, and Schlossman SF: Inhibition of anti- body-dependent cellular cytotoxicity and immunoglobulin synthesis by an antiserum pre- pared against a human B-cell la-like molecule. J. Exp. Med., 144: 113,1976. 13. Cline MJ and Billing R: Antigens expressed by human B-lymphocytes and myeloid stem cells. J. Exp. Med., 146:1143,1977. 14. Croizet H, Frindel E, and Tubiana M: Proliferative activity of the stem cells in the bone marrow of mice after single and multiple irradiations (total or partial body exposure). Int. J. Radiat. Biol., 18:347,1970. 15. Dacie JV and Lewis SM: Practical Haematology, 5th edn. Churchill Livingstone, Edinburgh, 1975. 16. Dicke KA, Tridente G, and Van Bekkum D\V: The selective elimination of immunologically competent cells from bone marrow and lymphocyte cell mixtures. III. In vitro tests for detection of immunocompetent cells in fractionated mouse spleen cells and primate bone marrow suspensions. Transplantation, 8: 422,1969. 17. Dresch C and Faille A: Kinetics of granulo-monocyte colony forming cells from bone mar- row, blood and cord blood in man. Exp. Hemat., 7: suppl. 6, 31,1979. 18. Drcsch C, Faille A, Poirier I, Balitrand N, and Najaen Y: Hydroxyurea suicide study of the kinetic heterogeneity of colony forming cells in human bone marrow. Exp. Hemat., 7: 337,1979. 19. Drew RM and Painter RB: Action of tritiated thymidine on the clonal growth of mammalian cells. Radiat. Res., 11: 535,1959. 20. Engh van den G, Russel J, and Cicco de D: Surface antigens of hemopoietic stem cells: The expression of BAS, Thy-i, and H1 antigens on CFU-s. In: Baum SJ and Ledney GD (eds), Exp. Hematol. Today, New York, Springer Verlag, 9,1978. 21. Faldetta TJ, Howe RC, Rogan KM et al.: Internal synthesis of P23.30 by several lymphoid malignancies. Exp. Hemat., 7: 95,1979. 22. Feher I and Antal S: Effect of phytohaemagglutinin on CFU-regeneration in mice. Eighth Annual Meeting of the European Society for Radiation Biology, Baska Polje, Yugoslavia, 1971. 23. Fitchen JH and Cline MJ: Human CFU-c express HLA-antigens. Exp. Hemat., 6: suppl. 3,47,1978. 24. Gidali J, Feher I, and Varteresz V: Differences in the growth and differentiation of the CFU fraction surviving sublethal irradiation. Radiat. Res., 52: 499,1972. 25. Gidali J, Feher I, and Antal S: Some properties of the circulating hemopoietic stem cells. Blood, 43: 573,1974. 26. Greenberger JS, Weichselbaum RR, Botnick LE, Sakakeeny M, and Moloney WC: Cell biological effects of total body irradiation on growth and differentiation of acute myelo- genous leukemia cells compared to normal bone marrow. Exp. Hemat., 7: suppl. 5, 279,1979. 27. Hanks GE: In vitro migration of colony forming units from shielded bone marrow in the irradiated mouse. Nature, 203: 1393,1964. 28. O'Hara C, Shumak KH, and Price GB: Colony formation by human myeloid progenitors affected by exposure to anti-i. Exp. Hemat., 6: suppl. 3, 39,1978. 29. Iscove NN, Till JE, and McCulloch EA: Proliferative state of human granulopoietic pro- genitor cells. Blood, 36: 828 (abstract) 1970. 30. Iscove NN, Till JE, and McCulloch EA: The proliferative status of mouse granulopoietic progenitor cells. Proc. Sos. Exp. Biol. Med., 134: 33,1970. 132

31. Iscove NN, Messner H, Till JE, and McCulloch EA: Human marrow cells forming colonies in culture: Analysis by velocity sedimentation and suspension culture. Ser. Haemat. V. 37,1972. 32. Jacobsen N, Broxmeyer HE, Grossbard E, and Moore MAS: Diversity of human granulo- poietic precursor cells: separation of cells that form colonies in diffusion chambers (CFU-d) from populations of colony forming cells in vitro (CFU-c) by velocity sedimentation. Blood, 52: 221,1978. 33. Janossy G, Greaves MF, Capellaro D, Roberts M, and Goldstone AH: Membrane marker analysis of "lymphoid" and myeloid blast crises in Ph' positive (chronic myeloid) leukaemia. In: Thierfelder S, Rodt H and 7,'hiel E (eds): Immunological diagnosis of leukaemias and lymphomas. 97, Springer Verlag, Berlin, 1977. 34. Johnson GR, Dresch G, and Metcalf D: Heterogeneity in human neutrophil, macrophage and eosinopb'l progenitor cells, demonstrated by velocity sedimentation separation. Blood, 50: 823,1977. 35. Johnson GR and Metcalf D: Growth of adult mouse bone marrow erythroid and mixed erythroid colonies in vitro. Exp. Heimt., 7: suppl. 6,3,1979. 36. Krogsrud RL and Price GB: Hemopoietic stem cells bear sperm associated antigens. Exp. Hemat., 7: 377,1979. 37. Kurland JI, Pelus LM, Meyers PA, and Moore MAS: Differential in vitro modulation of erythropoiesis and myelopoiesis by macrophages. Exp. Hemat., 7: suppl. 6, 26, 1979. 38. Lahiri SK and Van Putten LM: Location of the Go-phase in the cell cycle of the mouse haemopoietic spleen colony forming cells. Cell Tissue Kinet., 5: 365,1972. 39. Liu YK, Stallard SS, Koo V, and Dannaher CL: The proliferative states of circulating gra- nulopoietic stem cells in man. Scand. J. Haemat., 22: 258,1979. 40. Lofflei H: Cytochemischer Nachweis von unspezifischer Esterase in Ausstrichen. Klinische Wochenschrift, 39: 1220,1961. 41. Lohrmann HP, Novikovs L, and Graw Jr RG: Cellular interactions in the proliferative re- sponse of human T- and B-lymphocytes to phytomitogens and allogeneic lymphocytes. J. Exp.Med., 139: 1553,1974. 42. Loos H, Blok-Schut B, Kipp B et al.: Size distribution, electronic recognition, and counting of human blood monocytes. Blood, 48: 743,1976. 43. Lord BI: Growth kinetics of CFU of different densities. Annual Report of the Paterson Laboratories (Christie Hospital and Holt Radium Institute, Manchester), 102,1970. 44. Maloney MA and Patt HM: Migration of cells from shielded to irradiated marrow. Blood, 39: 804,1972. 45. Maruyama Y, Magura C, and Feola J: Corynebacteiium parvum induced radiosensitivity and cycling changes of heamtopoietic spleen colony forming units. J. Natl. Cane. Inst., 59: 173, 1977. 46. McCulloch EA and Till JE: The sensitivity of cells from normal mouse bone marrow in vitro and in vivo. Radiat. Res., 16: 822,1962. 47. Messner HA, Till JE, and McCulloch EA: Interacting cell populations affecting granulopoietic colony formation by normal and leukaemic human marrow cells. Blood, 42: 701, 1973. 48. Metcalf D, Wilson JW, Shortman K, Miller JFAP, and Stocker J: The nature of the cells generating B-lymphocyte colonies in vitro. J. Cell. Physiol., 88: 107,1975. 49. Mizoguchi H, Kubota K, Miura Y, and Takaku F: The proliferative status of mouse mega- karyocytic progenitor cells. Exp. Hemat., 6: suppl. 3,65,1978. 50. Monette FC, Morse BS, Howard D, Niskanen R, and Stohlman Jr F: Hemopoietic stem cell proliferation and migration following Bordella pertussis vaccine. Cell. Tissue Kinet., 5: 121,1972. 51. Monette FC, Kent RB, Weiner EJ, Livanis EC, and Lydon PJ: Cell cycle properties and proliferation kinetics of late erythroid progenitor cells. Exp. Hemat., 6: suppl. 3,12,1978. 133

52. Mookerjee BK and Ballard J: Functional characteristics of monocytes. II. Further obser- vations of lymphocyte-monocyte collaboration in the response to phytomitogens. Trans- plantation, 27: 59,1979. 53. Moore MAS, Williams N, and Metcalf D: In vitro colony formation by normal and leukemic human hematopoietic cells. J. Natl. Cancer Inst., 50: 603,1973. 54. Moore MAS and Williams N: Functional, morphologic, and kinetic analysis of the granulo- cyte-macrophage progenitor cell. In: Hemopoiesis in culture (ed. Robinson WA), 17, DHEW publication no. (NIH) 74-205,1973. 55. Morardet N and Parmentier C: Description of a suicide technique in vitro for granulocytic stem cells (CFU-c) by hydroxyurea on normal human bone marrow. Biomedicine, 27: 349,1977. 56. Nakeff R, McLellan WL, Bryan J, and Valeriote FA: Response of megakaryocyte, ery- throid, and granulocyte-macrophage progenitor cells in mouse bone marrow to gamma- irradiation and cyclophosphamide. Exp. Hemat. Today, New York, Springer-Verlag, 99, 1979. 57. Neff RD and Cassen B: Relative radiation sensitivity of circulating small and iaige lympho- cytes. J. Nuc. Med., 8,402,1968. 58. Nothdurft W, Calvo W, Fliedner TM, and Gerhartz HH: Quantitative and qualitative aspects of CFU-c mobilization in normal and splenectomizcd dogs. Exp. Hemat., 7: suppl. 6,117, 1979. 59. Oers van MHJ and Zeylemaker WP: The mixed lymphocyte reaction (MLR) stimulatory capacity of human lymphocyte subpopulations. Cell. Immunol., 31: 205,1977. 60. Ogawa M, Grush OC, O'Dell RF, Hara H, and MacEachern MD: Circulating erythropoietic precursors assessed in culture: characterization in normal men and patients with hemo- globinopathies. Blood, 50: 1081,1977. 61. Ornstein L, Hudson A, and Saunders A: Improving manual differential white cell counts with cytochemistry. Blood cells, 2: 557,1976. 62. Rickard KA, Brown RD and Kronenberg H: Studies on the proliferative capacity of the in vitro colony forming cell in normal human marrow. Aust. NZ. J. Med., 3: 361,1973. 63. Rinehart JJ, Gormus BJ, Lange P, and Kaplan ME: A new method for isolation of human monocytes. J. Immunol. Methods, 23: 207,1978. 64. Schofield R: A comparative study of the repopulating potential of grafts from various haemopoietic sources: CFU repopulation. Cell. Tissue Kinet., 3: 119,1970. 65. Siegers MP, Feinendegen LE, Lakiri SK and Cronkite EP: Relative number and proliferation kinetics of hemopoietic stem cells in the mouse. Blood cells, 5: 211,1979. 66. Sondel PM, Chess L, and Schlossman SF: Immunologic functions of isolated human lympho- cytes subpopulations. IV. Stimulation of MLC and CML by human T-cells. Cell. Immunol., 18:351,1975. 67. Standen G, Philip MA, and Fletcher J: The proliferative state of granulocytic progenitor cells in human blood and marrow. Cell. Tissue Kinet., 13: 105,1980. 68. Tebbi K, Rubin S, Cowan DH, and McCulloch EA: A comparison of granulopoiesis in cul- ture from blood and marrow of non-leukaemic individuals and patients with acute leukemia. Blood, 48: 235,1976. 69. Visser J, Engh G. van den, Williams N, and Mulder D: Physical separation of the cycling and non-cycling compartments of murine haemopoietic stem cells. In: Baum SJ and Ledney GD (eds), Exp. Haematol. Today, New York, Springer Verlag, 21,1977. 70. Vos O, Buurman WA, and Ploemacher RE: Mobilization of haemopoietic stem cells (CFU) into the peripheral blood of the mouse: effects of endotoxin and other compounds. Cell. Tissue Kinet., 5: 467: 1972. 71. Westring DW, Ladinsky JL, and Feick P: The volume distribution of human lymphocytes. Proc. Soc. Exp. Biol. Med., 131: 1077,1969. 134

72. Winchester RJ, Ross GD, Jarowski CI, Wang CY, Halper J, and Broxmeyer HE: Expression of la-like antigen molecules on human granulocytes during early phase of differentiation. Proc. Natl. Acad. Sci. U.S.A. 74: 4012,1977. 73. Worton RG, McCulloch EA, and Till JE: Physical separation of hemopoietic stem cells differing in their capacity for self renewal. J. Exp. Med., 130: 91,1969. 74. Zipursky A, Bow E, Seshadri RS, and Brown EJ: Leukocyte density and volume in normal subjects and in patients with acute lymphoblastic leukemia. Blood, 48: 361,1976. 75. Zucker RM and Cassen B: The separation of normal human leukocytes by density and classi- fication by size. Blood, 34: 591,1969. 135

Chapter 4

QUALIFICATION AND QUALIFICATION OF COLONY FORMATION BY HUMAN PERIPHERAL BLOOD HAEMOPOIETIC PROGENITOR CELLS IN PLASMA CLOT DIFFUSION CHAMBERS*

4.1 INTRODUCTION

The development of the in vitro agar colony technique and its applica- tion to study the colony formation ability of human haemopoietic progenitor cells has been a major advance in experimental haematology (16,46). There are two main features of this system, which form the basis of its potential clinical application. It allows the quantitation of granulo- cyte-macrophage progenitor cells in human marrow and blood, and it opens the possibility to study the biochemical regulators which control blood cell proliferation and differentiation. Comparable in vitro techniques are used for the detection of committed erythroid progenitor cells in human peripheral blood and bone marrow (46). However, these methods have several limitations. The cell popula- tion being assayed is not that of the multipotential stem cell, but com- mitted progenitor cells. The n mber, type and size of the formed colonies can be influenced by slight alterations in culture conditions. Moreover, the removal of colonies for examination is difficult and time-consuming. A culture system which would prevent either the entry or exit of cellular elements, but permits exposure of the cultured cells to humoral factors, would be of additional value in analysing the control of haemo- poietic cells. The diffusion chamber culture technique appears to fulfil many of these requirements. The physiological conditions provided by the chamber host may produce a more realistic cell proliferation and

*A modified version of this chapter is published; Zwaan FE, Te Velde J, Puliga A, Goselink H and Lobatto R: Stem cells in human peripheral blood: A method for histologic analysis of colony formation in plasma clot diffusion chambers. In: Diffusion chamher culture, Eds.: Cronkite EP and Carsten AL. Springer-Verlag, Berlin, Heidelberg, New York; 157,1980. 136

differentiation, since the cells are not subject to the vagaries of in vitro culture, which can alter the number, size, and type of the colony formed. The diffusion chamber technique was introduced by Algire et al. (1) in a study designed to distinguish cellular from humoral mechanisms of homograft rejection. The cells under study were enclosed in a chamber constructed from millipore filters, with a pore diameter small enough to prevent passage of intact cells, and a supporting plastic ring. The cha- mbers were then surgically implanted into the peritoneal cavity of host animals (mostly mice) and were removed at various intervals for analysis. In 1959 Berman and Kaplan (6) used diffusion chambers to culture culture mouse bone marrow cells in syngeneic hosts. Cells, capable of colonizing and therefore promoting the survival of lethally irradiated mice, persisted in the cultures for up to 30 days. In 1966, Nettesheim et al. (48) found that the clot formed within the chambers could be dis- solved by incubating the unopened chambers in a 0.5% pronase solution, without damage to the enclosed cells. This pronase treatment thus per- mitted the harvest of cells in suspension. Benestad (5) and B<^yum and Borgstr^m (10) subsequently developed methods for using pronase in the quantitative evaluation of haemopoietic cells cultured in diffusion chambers. B^yum et al. (12) reported an in- crease in myelopoiesis and numbers of CFU-c in diffusion chambers im- planted in irradiated hosts. This observation suggests that the environ- ment of the irradiated host triggered additional precursor cells into cycle, and once triggered they continue to proliferate even after re-implantation into a normal environment. The same year Breivik et al. (13) reported a decline in the number of CFU-s, to about 60% within two hours after implantation of normal mouse bone marrow. Thereafter, the numbers of CFU-s increased within the chambers and reached a level of about 150% compared to that of the original inoculum by the 4th to 5th day of culture. Also the number of committed myeloid progenitor cells (CFU-c) increased in the diffusion chamber (61). Hence, the diffusion chamber technique seems to be a useful technique for studying the regulation of haemopoietic progenitor cells. With the "in vivo" diffusion chamber technique, normal human bone marrow (8, 11, 18, 59) or peripheral blood progenitor cells (2, 15, 34) give rise to recognizable granulopoietic and erythropoietic cells and megakaryocytes. This technique offers a method to demonstrate all three committed progenitor cells simultaneously. This culture system has recently been modified to facilitate the study of colony formation by normal human haemopoietic progenitor cells in either semi-solid agar (26, 27) or fibrin clots (36, 37, 38, 39, 40, 41). 137

The plasma clot diffusion chamber offers a supporting semi-solid matrix, so that differentiating progeny cells remain localized and develop into discrete and quantifiable colonies. This system could circumvent problems of cell migration as was noted in CFU-c cultures (46) and the CFU-s assay (57). The number and morphological characteristics of cells comprising each colony provide a means to evaluate the proliferative and differentiating potential of progenitor cells. To obtain more specific information about the colony formation capacity of human peripheral progenitor cells cultured in plasma clot diffusion chambers, we adapted the methyl-methacrylate embedding technique described by Te Velde et al. (66). With this technique the diffusion chambers are in toto embedded in plastic, so that manipulation of the plasma clot can be avoided. The present study provides some quantitative information on progenitor cell proliferation and differentia- tion in diffusion chambers and attempts to describe the different types and structures of colonies.

4.2 MATERIAL AND METHODS

4.2.1 Mice The mice used were from the inbred strain NMRI. The diffusion chamber recipients were females, 8 to 10 weeks of age. The mice were given a commercial pellet diet (Hope Farms, Woerden, The Netherlands) and tap water ad libitum. The irradiated animals were given water acidified with hydrochlorid acid (6 ml HC1/1; pH 2.9) to prevent intestinal infections. The mice were kept in cages in groups of 4 to 8 during the experi- ments; otherwise up to 20 mice per cage. To harvest the diffusion cham- bers, the mice were killed by an ether overdose.

4.2.2 Irradiation of mice The mice received 650 Rad total body irradiation (TBI) by a Tele-cobalt 60-source (source-skin distance: 60 cm, dose rate 100 Rad/min.), 24 hours before diffusion chamber implantation.

4.2.3 Phenylhydrazine pretreatment of mice When indicated, NMRI-mice were given phenylhydrazine-hydrochloride 138

(PHZ), 60 mg/kg b.w. i.p. on two occasions, one day before and at irra- diation. Diffusion chambers were implanted 24 h later and harvested at various intervals.

4.2.4 Diffusion chamber technique "Conventional" chambers were constructed using commercial millipore plexiglass rings, millipore filters (type GS, porosity 0.22 fx) and millipore MF-cement (Millipore Corporation, Bedford, Mass., U.S.A.) according to techniques described earlier (5,6,10,11,12). The plexiglass ring had an external diameter of 13 mm, an internal diameter of 10 mm and a thick- ness of 2 mm. All chambers were sterilized for 12 h at 80°C and tested for leaks before use. 5 x 105 MNC (suspension A) in 100 jul RPMI were injected into each chamber using a 20-Gauge needle; the filling orifice was thereafter closed with a nylon rod and subsequently heat-sealed. The sealed chambers were stored in a 96 mm Falcon plastic culture dish (Falcon Plastics, Oxnard, Calif., U.S.A.) with 4 ml medium at 4°C until implantation. Two chambers were implanted into the peritoneal cavity of each mouse under ether anaesthesia. Approximately 0.5 ml RPMI-medium containing 1% penicillin and streptomycin was instilled and the wound closed with metal suture clips. At various times after implantation some of the animals were killed, the chambers removed, wiped clean with tissue paper and processed for cell analysis. After 7 days of culture, as a routine, all chambers were removed, cleaned and re-implanted into newly irradiated mice. To dissolve the fibrin clot in the chambers, they were shaken (1 h, 37°C) in a 5 ml solution of 0.5% pronase (pronase, B-grade, 45,000 p.u.k./ g, Calbiochem, San Diego, Calif., U.S.A.) and 5% Ficoll (Pharmacia, Uppsala, Sweden) in RPMI-medium. One membrane was pierced with a pasteur pipette, the chamber contents were aspirated into a 1.0 ml syringe and the chamber was thereafter rinsed twice to get all cells in suspension. The chamber contents and washings were transferred to a small pre-weighed conical plastic tube. The total volume was determined by weighing and the cells counted in a haemocytometer, so that the total cell amount per chamber could be calculated. Finally, the tubes were centrifuged (5 min., 200 g), the cell pellet re- suspended in a 20% albumin solution and smears prepared for cytological analysis. 139

4.2.5 Cell staining and counting Nucleated cells obtained after conventional diffusion chamber culture were counted in duplicate in a haemocytometer (Biirker) in Turks solu- tion (0.01% cristal violet + 1% acetic acid in saline). Cell smears were fixed in absolute methanol for lOmin. at 20°C and thereafter stained with May-Grunwald-Giemsa (diluted to 10% in a pH 6.8 buffer) for 10 min. The slides were then washed in the buffer and air-dried. Mono- cytes were determined by the use of the a-naphtyl-butyrate-esterase staining technique, described by Ornstein et al. (51). Cytochemistry (Sudan-Black-B, periodic acid Schiff (PAS)) was performed using standard techniques described by Dacie et al. (19).

4.2.6 Plasma clot diffusion chamber technique Plasma clot diffusion chambers were constructed according to a modifi- cation of the technique described by Steinberg et al. (60). Each chamber was seeded with 5 x 10s MNC (suspension B) in 50 nl RPMI, containing 40% citrated bovine plasma. The chamber was then filled to capacity with RPMI-medium containing 0.4 I.U. thrombin/ml (Hoffmann- La Roche, Basel, Switzerland). Each chamber was immediately sealed and shaken, to ensure a uniform distribution of the cells within the chamber. Each chamber was set aside for 5 min. until the clot was formed and then stored in 4 ml medium at 4°C in a Falcon plastic culture dish until implantation.

4.2.7 Methyl-methacrylate embedding technique and histological pro- cessing of plasma clot diffusion chambers After harvesting, the plasma clot diffusion chambers were cleaned with tissue paper and immediately fixed and embedded in methyl-methacrylate as described by Te Velde et al. (66). By using this technique there is no manipulation of the clot, the tissue shrinkage is minimal, and the resulting semi-thin sections are of high histological quality. Plastic blocks containing one chamber can be cut at 1.0-30 ju thickness on a Zeiss heavy duty microtome. All routine histochemical staining methods can be used, but some require slight modification of the staining times, depending on the thickness of the sections. 300 sections of 3.0 n thickness were made throughout the midportion 140 of each chamber, and the sections were automatically collected on a long cellophane tape. Three consecutive sections were thereafter mounted on a glass slide and stained with Gallamine-Giemsa or PAS. Then every sixth section was scored for different types of cell aggregates (CA) of > 3 cells (see Fig. 4.1).

Methyl - methacrylate embedded plasma-clot diffusion chamber

300 sections of 3.0/u •from midportion e' the chamber plasm* clot

plastic colony

sections each midsection on slide automatically analysed for O 00 on plastic type and number tape of cell-aggregates mounted on of ?- 3 cells glass-slide and stained

Fig. 4.1: Diagram of the histological processing of methyl-methacrylatc embedded plasma clot diffusion chambers.

It has to be stressed that in this study a cell aggregate is defined as cells lying in close proximity in a histological section and are not equal to a colony. From several whole colonies three dimensional reconstructions were made. For that purpose, serial sections of 5.0 [i thickness were made through whole colonies, which sections were subsequently analysed for total cell numbers, types of cells and spatial position of the cells. All slides were interpreted (magnification: 40 x, oil-immersion) by the same investigator.

4.2.8 Human peripheral blood mononuclear cell suspensions used in diffusion chamber cultures 440 ml blood was collected from healthy volunteers in 60 ml ACD (2.7% w/v anhydrous disodium citrate and 2.3% w/v anhydrous glucose). After centrifugation (1,200 g, 8 min., 20°C) plasma and cells were se- 141

parated and 50 ml of the upper layer of the packed cells was collected (= buffy coat). This buffy coat was diluted to 200 ml with RPMI-medium (Flow Laboratories, Irvine, Ayrshire, Scotland). To obtain mononuclear cells (MNC), 25 ml buffy coat was layered onto a 10 ml Ficoll-Isopaque density gradient (ds. 1.077 g/ml) and cen- trifuged for 20 min. (1,000 g, 20°C) (9). All cell separations were per- formed with a Sorvall-RC-3 centrifuge. The MNC in the interphase were collected and washed (10 min., 450 g, 20°C) three times with RPMI- medium in a Sorvall-GLC-1 centrifuge. These cells (suspension A) were used for culturing in conventional diffusion chambers. To obtain more 'purified' CFU-c suspensions, MNC were depleted of monocytes and T-lymphocytes (= suspension B). To remove phagocytic cells, 50 ml buffy coat cells were diluted to 100 ml in RPMI and incuba- ted with 500 mg sterile carbonyl-iron powder (type SF, Aristofarma. Delft, The Netherlands) in siliconized Erlenmeyer glass flasks (45 min. at 37°C, under continuous agitation) in a volume of 25 ml per flask. Thereafter a sterile magnet was added and the cell suspension was shaken

2 STEP CELL SEPARATION TECHNIQUE

Carbonyl-iron Buffy coat incubation (30 min, 37°C)

Ficoll-Isopaqu£ e (ds -1.077 g/ml) Step I Sedimentation Neutrophils, monocytes ,erythrocytes

E-Rosette Formation

Ficoll - Isopaque (ds :1.072 g/ml) Step II Sedimentation T- lymphocytes

650 R

5 x 10scells/Diffusion-chamber -Cultivation 13 days

Fig. 4.2: Diagram of the CFU-c "purification" technique and subsequent culture in diffusion chambers. 142 for another 5 min. Then 25 ml of the cell suspension in RPMI was layered onto a 10 ml Ficoll-Isopaque (ds. 1.077 g/ml) gradient and the MNC in the interphase were collected after centrifugation (1,000 g, 20 min., 20°C). The MNC-suspension was subsequently allowed to form E-RFC according to the technique described in section 2.2.10. After incubation (1 h, 4°C) the rosettes were resuspended and 10 ml Ficoll-Isopaque (ds. 1.072 g/ml) was carefully layered underneath the rosette suspension with a 10 ml pipette. After centrifugation (20 min., 1,000 g, 4°C) the non-rosetting cells were collected, washed three times in RPMI and processed for further diffusion chamber culture (Fig. 4.2). The various characteristics of these cell suspensions, used for culture in conventional diffusion chambers (suspension A) or plasma clot dif- fusion chambers (suspension B), are summarized in Table 4.1.

Table 4.1: Several 'in vitro' characteristics of the cell suspensions used for diffusion chamber culture

Initial buffy coat Suspension-A Suspension-B

Mononuclear cell yield (xlO6) 500 321130.1 23.61 3.1 (= 100%) (=64%) (= 4.7%) E-RFC % 47.213.1 61.21 2.5 4.91 0.8 Monocytes % 10.2 ±1.2 10.5 1 1.4 3.4 1 0.5 SIg(+) cells % 5.310.6 6.91 0.6 11.1 1 1.5 CFU-c/5 x 10s MNC 10.4 11.9 26.1 110.3 83.7 122.0 Mean CFU-c 'purification' factor (x) 1.0 2.5 8.0

Suspension A: Buffy coat cells after Ficoll-Isopaque (ds. 1.077 g/ml) centrifugation only, without specific monocyte elimination Suspension B: Buffy coat cells after two Ficoll-Isopaque separation steps (see Fjg. 4.1), with sequential monocyte and T-cell elimination Alle values are given as the mean 1 s.e.m. The number of mononuclear cells in the initial buffy coats is recalculated to 500 x 106 (=100%) and the subsequent mononuclear cell yields of suspension A and B consequently corrected.

4.2.9 Statistical methods The results are expressed as arithmetic mean values ± 1 standard error (± s.e.m.). Differences in experimental results were anlysed with Student's two-sided t-test, for two samples or paired observations. Statistical sig- 143 nificance was confirmed by a non-parametric rank test (Wilcoxon Mann Whitney). Homogeneity of the CA-distribution in 50 sections per cham- ber was tested with Spearman's rank correlation test. Only p-values of less than 0.05 were considered as significant.

4.3 RESULTS

4.3.1 Effect of irradiation and/or phenylhydrazine pretreatment on the survival and peripheral blood counts of mice Besides the total body irradiation (TBI) applied to diffusion chamber host mice, phenylhydrazine (PHZ) pretreatment was also instituted in a Fig. 4.3: Survival of NMRI-mice after total body irradiation. Survival % 100 »- o—o650Rad 90- •—• 750 Rad

80 o—i

70-

60- O—i

50- O 1

40 -

30-

20- O—i

10- -1 •n • • - o- 2 U 6 8 10 12 u Days after irradiation 20 mice were irradiated with 650 Rad (o o) and 20 mice with 750 Rad (• 144

group of mice in an attempt to stimulate human erythropoiesis in diffusion chambers (see section 4.3.7). Irradiation of the host mice is essential to allow an optimal growth of human cells. The total dose should be as high as possible, but the mouse must live at least 7 days to make re-implantation of the diffusion cham- bers possible. Two groups of 20 mice each were exposed to TBI of 650 Rad and 750 Rad respectively; the survival curves are presented in Fig. 4.3. From this experiment we adapted the 650 Rad TBI dose to diffusion chamber bearing NMRI-mice. Thereafter an experiment was performed using three groups of mice (10 mice in each group), which received 650 Rad, 650 Rad + PHZ, or PHZ only. The peripheral blood counts were obtained by sampling free-flow tail blood. The results are presented in Fig. 4.4. From the leucocyte curves (Fig. 4.4a) it is obvious that PHZ induced a leucocytosis in the host mice, since with 650 Rad + PHZ higher values were obtained than with 650 Rad only. The haematocrit (Fig. 4.4b) showed a very rapid decrease after PHZ only, with a nadir on day 2 and normalization on day 9. The 650 Rad + PHZ curve therefore showed a lower course than 650 Rad only, but no clear dip. As far as the reticulocytes were concerned (Fig. 4.4c), PHZ induced an initial decrease, nadir on day +1, followed by an overshoot with a maxi- mum on day +5. However, a second dip in reticulocyte-counts was observed on day +6. As expected, the 650 Rad + PHZ treatment showed higher values than 650 Rad only, with a small peak on day 6. This suggests that, although the marrow is irradiated, it is still capable to respond to an erythropoietic challenge. 145

Fig. 4.4: The influence of iriadiation with 650 Rad (o o); irradiation + phenylhydrazine (• •), or phenylhydrazine only (^ &) on the peripheral blood values of leuco- cytes (Fig. 4.4a), haematocrit (Cig. 4.4b) and reticulocytes (Fig. 4.4c).

twcocyUt (>10Sj() 650R*RH2. 650 R RH.Z. 3C.0

W.O

3.5 3.0- 1.5 JB 1.5-

3 t 5 I 7 Diyi after trutmtnt M (V.) 650R + RH.Z. 70 650 R E5 P.H.Z. 60 Si 50 IS to- 35- 30- 25- 20- 15- 10- 5- 0- 15 6 7 0r/% aftar traatirant Rttic (%)

IS- 1.0-

RHZ. RHZ,

For each group of treatment 10 NMRI-mice were used and the mean values are given. Vertical bars indicate ± s.e.m. 146

4.3.2 Proliferation and differentiation of human peripheral blood mono- nuclear cells in conventional diffusion chambers. Cytological aspects and growth profiles The change in the total number of nucleated cells per chamber with time after implantation of 5 x 105 MNC of suspension A, is illustrated in Fig. 4.5.

Fig. 4.5: Change in total number of nucleated cells per chamber with time after implantation of 5 x 10s mononuclear peripheral blood cells (suspension A) from 6 healthy individuals.

Rttransplantation

5 7 9 13 15 Days after implantation Chambers were implanted into irradiated (650 Rad) hosts and re-implanted on day 7 into newly pre-irradiated mice. Mean values of 4-6 chambers from each individual at each time harvest arc given. Vertical bars indicate ± s.e.m.

In all 6 normal persons studied, the growth pattern was similar, con- sisting of an initial decrease in absolute cell count of 50%, with a nadir on day 5, followed by an increase in cell number to a maximum around day 13, after which the cell count decreased again. However, great individual variation in growth capacity was observed. 147

On day 13, cytological investigation (Fig. 4.6a-d) showed that lym- phoid blast cells and macrophages predominated the picture. There was abundant vacuolization in the majority of cells. However, non-prolifer- ating and proliferating granulopoietic cells were observed, which ex- hibited strong affinity for Sudan black B. Megakaryocytes were also present in all chambers and the majority of them were multinucleated. Megakaryocytes could be easily differentiated from large macrophages by their negativity in the a-naphtyl-butyrate-esterase staining. Occasionally eosinophils and erythropoietic cells were observed. The use of the MNC-suspensions for chamber culture has the advantage that the subsequent appearance of recognizable haemopoietic cells can be taken as evidence for their development in the chamber. The appear- ance of these cells therefore suggests differentiation of committed pro- genitor or even pluripotent stem cells in these diffusion chambers.

4.3.3 Effect of preculturing haemopoietic progenitor cells in diffusion chambers on CFU-c forming capacity in vitro From the above experiment it was clear that haemopoietic progenitor cells, when cultured in diffusion chambers, gave rise to recognizable progeny cells of different types. Besides the differentiating capacity of these progenitor cells, it was also of interest to know if the committed progenitor cells (e.g. CFU-c) were subject to quantitative alteration induced by preculturing in dif- fusion chambers. Therefore, 5 x 105 MNC were implanted into conven- tional chambers and harvested at different time intervals. After harvest, the chamber contents were processed for CFU-c culture according to the technique described in section 2.2.16. The results obtained with MNC from 6 healthy donors are presented in Fig. 4.7. It is clear that, although the profiles were different from person to person, a substantial increase in the total number of CFU-c/ 2 x 104 MNC occurs in time, in 4 out of 6 tested individuals. The other two individuals demonstrated rather constant CFU-c levels, but on day 0 they already had a relatively higher initial CFU-c number. This increase in CFU-c numbers could be due to several mechanisms. First of all, the micro-environment in the chamber allows more primitive stem cells to differentiate towards CFU-c. Secondly, CFU-c, which were in Go-phase are triggered into a proliferative state, resulting in an in- creased potency of colony formation in vitro. Finally, it is quite possible that the CFU-c population was expanding by self-replication. 148

Fig. 4.6: Cy tological aspects of human haemopoietic cells, harvested from conventional diffusion chambers after 13 days of culture.

4.6.a) large lymphoid cells, macrophages and myeloid cells with abundant vacuolization (May- Grunwald Giemsa) 4.6b) cluster of myeloid cells, surrounded by small and large lymphocytes (May-Griinwald- Giemsa) 4.6c) immature myeloid cells, with a strong affinity for Sudan-black-B 4.6.d) multinucleated megakaryocytes (May-Grunwald-Giemsa)

Therefore, diffusion chamber culture does not only allow committed progenitor cells to differentiate, but also provides an environment for expansions of the CFU-c compartment itself. 149

Fig. 4.7: Change in concentration of CFU-c during diffusion chamber culture of mononuclear peripheral blood cells from 6 normal persons.

Retransplantation 4 CFu-c/2x10 cells

600-

400-

2 4 6 8 10 12 Days after implantation

Each point is the mean of 4-6 diffusion chambers. Vertical bars indicate ± s.e.m. Ordinate: CFU-c/2 x 104 cells Abscissa: days of diffusion chamber culture before the harvested cells are transferred to methyl- cellulose culture for measurement of CFU-c concentration.

4.3.4 The proliferation and differentiation of haemopoietic progenitor cells in plasma clot diffusion chambers. "Histological" aspects To obtain more information about the expected colony formation, peri- pheral blood MNC suspensions were cultured in plasma clot diffusion chambers. 150

To avoid the abundant overgrowth of lymphoid blast cells and macro- phages, more "purified" progenitor cell suspensions were made (Fig. 4.2) and subsequently cultured in plasma clot diffusion chambers. After 7,9, 11 and 13 days of culture the chambers were processed for analysis according to the schedule in Fig. 4.1. Erythropoiesis was observed on day 7 and the following days. During the first few days, only small groups of predominantly immature ery- throid elements were seen. On day 11 and 13 most erythroid cell aggre- gates (CA) consisted of mature elements, recognizable by their abundant cytoplasm and condensated nuclei. On day 13 only a few proerythro- blastic CA were observed. Maturation to erythrocytes was convincingly observed in several cases. The human erythrocytes were distinct from "leaking" mouse erythrocytes by their abnormal cytoplasmic configu- ration. In the PAS-stain they were often positive and showed other signs of dyserythropoiesis. Myeloid elements were only observed from day 9 onwards, when the first immature granulopoietic CA were found. The majority of mature myeloid cells were found on day 11 and 13. Isolated granulocytes were often found in the vicinity of other CA, indicating active migration (Fig. 4.8a and 4.8b). On day 11 and 13, a compact CA of granulocytic cells, mostly com- prising less than 10 cells, consisted of myeloblasts and myelocytes with scattered promyelocytes. Basophilic granulocytes were rare. Eosinophilic granulocytes were often seen in mature stages and in those cases in compact clusters. Migration of eosinophilic granulocytes was hardly ever observed. Cytological details of all granulocytic lineages generally showed a normal differentiation and granulation pattern. Only in a few instances severe megaloblastic nuclear abnormalities manifested themselves. Maturation and differentiation within the CA was usually asynchronic, while differences in maturation existed among the different CA. Those CA usually consisted of only mature cells without younger elements. The monocytic elements were found at all times, but colonies of monocytes were very rare. Usually the monocytes were scattered through the plasma clot and showed abundant phagocytosis which caused PAS- positivity by phagocytosed plasma clot material. With their long cyto- plasmic protrusions they formed large star-like interruptions in the fibrin network. The nuclei of the monocytes had a very delicate chromatin pattern and a fine nuclear membrane. The nucleoli were seldom conspi- cuous; nuclei sometimes showed hyperlobulation, such as seen in mega- 151

Fig. 4.8a: Two adjacent cell aggregates, one consisting of mycloid cells, the other composed of small megakaryocytes mixed with some erythroids (small cells with dark condensed nuclei).

i

Fig. 4.8b: Another myeloid colony, with maturating cells in the centre and peripheral migrating mature neutrophils.

Day 13 of culture, methyl-methacrylate, Gallamine-Giemsa. Original magnification x600. 152 karyocytes. However, especially the distribution of PAS-positivity in the cytoplasm helped to distinguish macrophages from megakaryocytes. Megakaryocytes were already found on day 7. These cells were mostly found in groups of three to four per CA (Fig. 4.9). True megakaryoblastic elements were rare. At the stage of differentiation, at which we could distinguish the megakaryocytes, there were signs of abundant platelet formation. The cytoplasm showed a strong peripheral PAS-positivity with variable, but often abundant cytoplasmic protrusions and bleb formation. Isolated platelets, however, were hardly ever observed. A peculiar phenomenon was the presence of star-like ghosts, which in several instances ^uld be related to vanishing megakaryocytes, since their cor.uensing nuclei could be found within the centre of these stars. Peripheral PAS-positivity in the "dying" cells was hardly retained within their border but nevertheless, distinct from that seen in the macrophages with their cytoplasmic extensions. The nuclei of vital megakaryocytes showed a varying tendency to hyperlobulation, in particular megakaryo- cytes with clear signs of platelet formation. Fibroblasts with formation of collagen fibres were not observed. The groups of haematopoietic cells were usually limited to the area of the plasma clot itself. At the borders the background of fibrinous ma- terial sometimes vanished, thus leading to a seemingly liquid background in which scattered haemopoietic elements were found. Most cells, how- ever, were found within the plasma clot itself. In control sections made in close vicinity or through the millipore membranes, no preference for colony growth could be established. Combinations and fusions of CA were seen in instances where mature granulocytic cells spread out and intermingled with other types of CA. However, in some CA, fewer differentiated elements of various cell lines were recognized. The ery throid cell line, for instance, was most often found in combination with other cell lines, especially the megakaryocytic ones (Fig. 4.10). Combinations of the myelo-monocytic cell lines or neutrophilic-eosinophilic cell lines were not observed. In all other combi- nations, each cell line seemed to proliferate and differentiate in the same manner as in the mono-lineaged CA. The eosinophils within the mixed CA tended to remain clustered in compact groups, while the megaka- ryocytes tended to spread out in one or two directions, forming new clusters. Mixed CA, in which elements of different cell lines and in different maturation states were intermingled in a haphazard way, were not observed within the intact clot sections. 153

Fig. 4.9: Cell aggregate comprising megakaryocytes with platelet formation and expulsion.

Fig. 4.10: Same colony as in Fig. 4.9, but section obtained at a different level. Mixture of mega- karyocytes and ery throid cells.

AW

Day 13 of culture, methyl-methacrylate, GaUamine-Giemsa. Original magnification x400. 154

4.3.5 Quantitative aspects of cell aggregate (CA) formation in plasma clot diffusion chambers Since a CA is a cross section through a whole colony, the total number of CA does not represent the total number of whole colonies per dif- fusion chamber. Nevertheless, quantitative aspects of CA-formation could give an impression of progenitor cell proliferation in this system. Therefore, using 4-6 diffusion chambers per day of analysis, the total number of CA was scored from each of 4 normal donors on day 7, 9, 11 and 13 after implantation. Moreover, on day 13, 4-6 diffusion chambers from each of six additional donors were also analysed to obtain more in- formation concerning CA-formation at the moment of maximal cell pro- liferation. The total number of all kinds of CA per chamber was increasing with time (Fig. 4.11): 1.6 ± 1.3 on day 7; 2.3 ± 1.5 on day 9; 28.7 ± 9.4 on day 11, and 187.1 ± 30.5 on day 13. A "control" of unseparated MNC (suspension A) from the same 4 donors cultured in plasma clot diffusion chambers and analysed on day 13, showed a significantly lower (p < 0.05) total number of CA per chamber (23.8 ± 14.1; 8 plasma clot diffusion chambers). The increase in time of the total number of CA could represent either an increase in total number of colonies and /or an increase in the total number of cells per colony. With the scoring method used in these experiments, it was not possible to differentiate between these two possibilities. However, since the "purified" cell suspension B contains more CFU-c in vitro than suspen- sion A (see Table 4.1), and since this phenomenon is also analogized by a higher total number of CA found on day 13 (187.1 ± 30.5) compared to suspension A cultured in plasma clot diffusion chambers (23.8 ± 14.1), it may be assumed that the increased total number of CA in time was not only caused by an increasing number of cells per colony but also by an increasing number of colonies. As stated in section 4.3.4, mixed CA were also observed in the plasma clot diffusion chambers. Expressed as a percentage of the mean number of all CA per chamber on day 13 of culture, 5% mixed myeloid and megakaryocytic, 2.5% myeloid and erythroid, 0.5% myeloid, erythroid and megakaryocytic, and 0.5% megakaryocytic and erythroid CA were found. As CA were counted in cross-sections, it was important to obtain in- formation about the distribution of different kinds of CA throughout the analysed midportion of the chambers and whether this distribution is homogeneous. It is conceivable that CA are not equally distributed, but 155

Fig. 4.11: Change in total number of all kinds of cell aggregates (CA) with time after implanta- tion of 5 x 10s mononuclear blood cells (depleted of monocytes and T-lymphocytes = suspension B) from 4 healthy donors.

total number of all C.A.

300 -

200-

100- 80 - 60 -

40 -

20 -

10 - 8 -

6 -

I. -

2 •

1 I 13

Days of culture

Chambers were implanted into 650 Rad irradiated mice, and re-implanted after 7 days into newly irradiated mice. The CA-counts of 4-6 chambers per donor at each time of harvesting were col- lected and the mean values are given. Vertical bars indicate ± s.e.m. are preferentially localized in circumscript areas of the chambers. There- fore, 7 at random chosen plasma clot diffusion chambers from different individuals harvested on day 13, were further analysed. For myeloid and megakaryocytic CA the mean ± s.e.m. per section was determined by analysing all 50 sections per chamber. As can be seen from Table 4.II, the mean number of myeloid CA per section was 2.48 ± 0.39 and for megakaryocytic CA a mean number of 156

Table 4.H: Number of rnyeloid and megakaryocytic cell aggregates (CA) per section (50 sections from each of 7 plasma clot diffusion chambers were counted, scored on day 13)

chamber mean number Spearman's Spearman's test: number of CA/section skewness rank correla- significant diffe- tion (r) rence from zero

1 3.06 + 0.28 -0.51 p<0.05 2 3.90 + 0.48 -0.43 p<0.05 3 3.90 + 0.87 -0.77 p<0.05 myeloid-CA 4 1.90 + 0.65 -0.56 p<0.05 5 1.86 + 1.50 -0.24 NSD 6 1.36 + 0.62 -0.02 NSD 7 1.38 + 0.54 -0.10 NSD

mean (±s.e.m.) 2.48 + 0.70 of 7 chambers (±0.39)

1 1.30 + 1.28 -0.52 p<0.05 2 0.86 + 1.14 -0.65 p<0.05 3 0.70 + 1.57 -0.39 p<0.05 megakaryocytic-CA 4 0.54 + 1.60 -0.11 NSD 5 3.04 + 1.18 -0.57 p<0.05 6 1.42 + 0.45 -0.34 p<0.05 7 2.00 + 0.78 -0.24 NSD

mean (±s.e.m.) 1.40 + 1.14 of 7 chambers (±0.30)

For normal distributions: - 0.5 < skewness <+0.5. The homogeneity of the CA distribution in the 50 sections per chamber was tested with the Spearman's rank correlation test (p < 0.05 indicates a correlation and therefore an inhomogeneous distribution). NSD = non significant difference.

1.40 ± 0.30 was found, which difference is not significant. Formyeloid CA the distribution showed a mean skewness to the right of +0.7, while for megakaryocytic CA a mean skewness to the right of+1.14 was found (p < 0.05). This implies that lower CA-counts per section were more frequently obtained for megakaryocytic CA than for myeloid CA. With Spearman's rank correlation test, the homogeneity of the CA dis- tribution from section 1 to 50 for myeloid and megakaryocytic CA in the same 7 chambers was investigated. From Table 4.II it can be seen that several chambers are non-homogeneous with regard to CA distribu- tion, which is caused by negative or very low CA-counts in a substantial number of consecutive sections. This means, that quantitative or even 157 semi-quantitative estimation of colony formation using histological sections can easily lead to errors if they are taken from limited parts of the diffusion chamber content. Since the scoring of 50 sections per chamber was a very laborious pro- cedure, the total numbers of all kinds of CA per chamber were re-counted in a decreasing number of sections obtained from the same seven plasma clot diffusion chambers.

Fig. 4.12: Relationship between the number of counted sections per diffusion chamber and the scored number of myeloid and megakaryocytic cell aggregates (CA).

of myeloid. CA

200

0 10 20 30 40 50 60 0 10 20 30 40 ^0 60 #i of sections counted # of sections counted

The counts are given as absolute values from 7 plasma clot diffusion chambers, scored on day 13 of the culture. The sections were counted in a 1:1 (50 sections); 1:2 (25 sections); 1:3 (16 sections); and 1:4 (12 sections) "sample dilution" manner, whereas every first section was always included. When the numbers of counted sections were plotted against the total numbers of scored CA, straight lines were obtained for both megakaryocytic and myeloid CA (Fig. 4.12), if the points were fitted with the least square method. All the calculated intercepts were tested against the null-hypothesis: intercept = 0, using the 95% confidence limits of each intercept. The majority (except two) of the intercepts were not significantly different, so the null-hypothesis was not rejected. 158

These results demonstrate that, for the interval studied, a limited num- ber of sections could be scored to obtain the same quantitative informa- tion, provided that these sections were taken at regular intervals through- out the whole chamber to avoid errors caused by inhomogeneous dis- tributions.

4.3.6 Three-dimensional reconstruction of whole colonies in plasma clot diffusion chambers; emphasis on the structure of mixed colonies In the two-dimensional sections through the proliferating colonies various shapes of the CA were seen. A small CA tended to be spherical, while in mature and larger CA signs of migration seemed to exist. In the combination of two or more lineages within one CA, the two-dimensional sections did not inform us, whether these pictures resulted from a haphazard sectioning of fused, mixed or touching intrinsic spherical structures. The methacrylate sections are well suited for three- dimensional studies, by the lack of both shrinkage and distortion during histological processing. In series consisting of 80-120 consecutive sections through different chambers, we randomly selected small and large CA in the midportion of the chambers, which by analysis of serial sections could be studied in total. In a selection of 8 different CA consisting of different lineages and combinations, the cells per section were counted and scored individually to see if they had already been recognized in previous sections (Table 4.IV).

Table 4.FV: Several quantitative characteristics of the three-dimensional analysis of eight different colonies, scored on day 13 of culture in plasma clot diffusion chambers

total number Maximal number number of serial- of of all scored 5 JU sections, type of colony cells/colony cells/ needed to pass the single section whole colony col. #\.,eosinophilic 6 5 7(=35jU) col. #2, neutrophilic 52 28 11(=55JU) col. #3, pro-erythroblastic 10 9 7 (=35/1) col. #4, mixed megakaryocy'tic- megak.: 47 i 27 eosinophilic eos. :11J48 #15 (-75/1) col. #5, mixed megakaryocytic- megak.: 56 i _ 28 erythropoietic erythr.: 16 J [J]13 (=65 ju) col. #6, neutrophilic 11 6 8 (=40jU) col. #7, megakaryocytic 38 21 1O(=5OU) col. # 8, megakaryocytic 5 4 7(=35JU) 159

In the serial reconstructions of small colonies, the impression obtained from the two-dimensional sections, that small colonies tend to be speri- cal, was confirmed. This goes for colonies consisting of immature cells of all lineages, while differentiated neutrophilic cells and megakaryocytes showed definite signs of migration into the surrounding plasma clot. Eosinophils stay together in spherical groups, even when the CA con- sists of only polymorphonucleated forms. The structure of erythropoie- tic nests was more complicated, mainly due to the fact that during dif- ferentiation the nuclei disappear and mature erythrocytes are difficult to retrieve with certainty within the plasma clot in all instances. In the large and composite colonies this phenomenon can be best illustrated by the colony shown in Fig. 4.13. In the serial reconstruction, it was found that the eosinophils tended to cluster, but revealed slight migration into a direction which coincides with one of the main directions of megakaryocytic migration and pro- liferation. The megakaryocytic colonies showed protrusions in mainly three directions with irregular borders and isolated cells at some distance. This same tendency of outspreading megakaryocytic proliferation is also seen in Fig. 4.14. Here scattered nests of mature erythroid elements, still containing dark clumps of chromatin, are found scattered among the mi- grating megakaryocytic cells. No distinct centre of erythroid prolifera- tion and maturation was found. From these two examples it is clear, that mixed colonies do not consist of randomly or homogeneously mixed differentiating cells from various lineages, but that each lineage tends to grow in the same manner as in isolated colonies. Besides that, the growth pattern of non-migrating cells in CA was clearly influenced by the migrating activity of mature myeloid cells and megakaryocytes. However, the influence of migrating neutrophils on eosinophilic colony formation cannot be elucidated here, since we did not find combined neutrophil-eosinophil colonies. 160

f/i/4 r> /»>•» /••/*

Fig. 4.13: Three-dimensional impression of a mixed megakaryocytic-eosinophilic colony scored on day 13 of the culture. Both the serial section construction and the artist impression are presented. I Dark spots represent cross-sections through eosinophils. Magnification xl030. 161

/*.•

Fig. 4.14: Three-dimensional impression of a mixed megakaryocytic-erythropoietic colony scored on day 13 of the culture. Both the serial section construction and the artist impression are presented. Dark spots represent cross-sections through erythropoietic cells. Magnification xl030. 162

4.3.7 Cell aggregate (CA) formation in plasma clot diffusion chambers by human haemopoietic progenitor cells in irradiated and phenyl- hydrazine pretreated mice When human haemopoietic progenitor cells were cultured in diffusion chambers, erythroid cells were rarely observed. Therefore it was of interest to investigate if phenylhydrazine (PHZ) pretreatment (and TBI) of the host mice could induce an increased erythropoiesis in the diffusion chambers. PHZ induces a severe haemo- lysis and subsequently a stimulation of erythropoiesis in the host mice by a reactive increase in erythropoietin levels (28).

Table 4.HI: Numbers of myeloid and megakaryocytic cell aggregates (CA) per section (50 sections from each of 5 plasma clot diffusion chambers were counted, scored on day 13). Chamber bearing mice were prctreated with TBI and phenylhydrazine

chamber mean number of skewness number CA/section

I 4.90 + 0.82 2 1.38 + 1.60 myeloid-CA 3 1.42 + 0.45 4 7.34 + 0.03 5 0.96 + 0.52

mean (±s.e.m.) of 5 chambers 3.2±1.12 + 0.60

1 3.52 + 1.12 2 1.00 + 0.76 megakaryocytic-CA 3 0.34 + 2.10 4 2.36 + 1.69 5 2.35 + 1.03

mean (is.e.m.) 1.91+0.5 + 1.34 of 5 chambers

From the same four donors from whom CA-time profiles were obtained (see section 4.3.5), plasma clot diffusion chamber analysis was per- formed (10 chambers in total) after PHZ treatment of the host mice. Histological analysis on day 13 of culture showed no increase in the total number of erythroid CA, nor in the total number of cells per erythroid CA. Furthermore, no difference was found in the total number of all kinds of CA per chamber (165.7 ± 50.1) compared to pre-irradiated mice without PHZ treatment (187.1 ± 30.5). 163

CA-distribution profiles throughout the midportion of 5 out of 10 at random chosen chambers were determined and the results are presented in Table 4.III. The mean (± s.e.m.) number of myeloid CA per section (3.2 ±1.1) was not significantly different from the mean number of megakaryocytic CA (1.9 ± 0.5). Neither did these values differ from the results obtained with pre-irradiation only, as is described in section 4.3.5. From these experiments it can be concluded that an erythropoietic challenge with PHZ of the irradiated diffusion chamber bearing hosts does not result in an increase of erythroid cell proliferation or differen- tiation, nor in any significant alterations in the number of myeloid and/or megakaryocytic CA.

4.4 CONCLUSIONS AND DISCUSSION

Conclusions

1) Haemopoietic progenitor cells, which circulate in the human peri- pheral blood, can be induced to differentiate into mature erythro- cytes, granulocytes as well as thrombocytes, by culture in conven- tional diffusion chambers. 2) The number of progenitor cells, which in vitro produce myeloid colonies (CFU-c), increases if the peripheral blood cells are pre- cultured in diffusion chambers. 3) "Histological" analysis of colony formation in plasma clot diffusion chambers revealed neutrophilic colonies with normal asynchronous cell differentiation and signs of active cell migration. Sometimes megaloblastic changes were evident. The megakaryocytic cell aggre- gates revealed signs of platelet formation at the cell borders, while the scanty erythroid cell aggregates were able to form mature ery- throcytes. 4) Eosinophils and neutrophils were always found in separate colonies, supporting the hypothesis that both lineages have separate progenitors. Furthermore, the mature eosinophilic cell aggregates were very com- pact and did not show any sign of cell migration. Monocytic cell aggregates were very rare, while macrophages were frequently found scattered throughout the plasma clot. Cell aggregates consisting of lymphoid cells were never observed. Aggregates consisting of a mix- ture of various cell lineages were found in small amounts; especially 164

erythroid cells showed a tendency to grow in the vicinity of mega- karyocytic cells. 5) Cell aggregates are not homogeneously distributed throughout the plasma clot. This means that quantitative or even semi-quantitative estimation of colony formation, using histological sections, can easily lead to errors if these sections are taken from limited parts of the dif- fusion chamber content. 6) Pretreatment of the irradiated host mice with phenylhydrazine did not increase the number of erythroid cell aggregates, nor the total number of cells per erythroid cell aggregate. This is in contrast with observations done in mice. 7) Three-dimensional analysis of various colonies revealed that small colonies were perfect spheres, while large colonies revealed bizarre structures with signs of active cell migration and satellite formation. Analysis of "mixed" colonies suggested that the majority of these colonies did not consist of homogeneously mixed differentiating cells from various lineages, but were rather the result of cell migration from closely developing single cell aggregates. Especially the growth pat- terns were clearly influenced by cell-migrating activity of mature myeloid cells and megakaryocytes.

Discussion

In the literature of the past few years, modifications of the diffusion chamber technique, which facilitates the study of colony formation by precursor cells in semisolid agar (26, 27) or in fibrin clots (36, 37) inside the chambers, have been described. The types of colonies most frequently observed in fibrin clot cultures consist of neutrophilic, eosinophilic, megakaryocytic, or macrophage-like cells. An increased number of neutrophilic colonies is observed when the host mice are irradiated prior to chamber implantation (37). In contrast, irradiation of the host has not been found to stimulate the formation of eosinophilic colonies (37). Colony-forming units in diffusion chambers (CFU-d) are operationally defined as cells that form colonies in fibrin clot cultures in irradiated mice. From the studies done by Hoelzer et al. (34), Barr et al. (2) and Boecker et al. (8) it was suggested that committed or even pluripotent haemopoietic stem cells, circulating in human peripheral blood, pro- liferate and differentiate in diffusion chambers. 165

However, in contrast to bone marrow (37, 38, 54), analyses of colony- formation from human peripheral progenitor cells in plasma clot dif- fusion chambers have not yet been done. Such analysis is relevant in relation to the observations done by Gidali et al. (25) and Micklem et al. (47), that the pluripotent stem cells found in circulating blood of mice generally have less reproductive ability to sustain haemopoiesis than the CFU-s found in bone marrow. Furthermore, the proliferative and differentiating capacity of haemo- poeitic progenitor cells present in human peripheral blood must be studied more thoroughly before they can be used for transplant purposes. In the techniques described for bone marrow derived colony analysis, the cell aggregates are studied either after chamber centrifugation (36) or by direct staining techniques on manipulated plasma clots (54). In both techniques it is conceivable that, because of interference with the clot structure, alteration in the colony structure and morphological aspects can influence the interpretations. Also the light microscopical and ultra-structural techniques described by Ben-Ishay (3, 4) have the dis- advantage that clot manipulation is part of the procedure. By using the methyl-methacrylate embedding technique, the whole chamber (including the plastic ring) can be processed for analysis whereby semi-thin sections of high quality with a uniform thickness and without shrinkage can be obtained. The growth profiles of mononuclear cells in conventional diffusion chambers and subsequent cytological analysis as described in section 4.3.2 confirm the data of Boecker et al. (8) and Hoelzer et al. (34). The use of a mononuclear cells suspension for implantation also has the ad- vantage that the interpretation is not complicated by the possibility that these cells are survivors from implanted differentiated cells, as is the case for bone marrow (15). Under the experimental conditions of pre-irradiation, times of re- implantation and times of harvesting used, a growth pattern was obser- ved with an initial decrease of 50%. Thereafter proliferation of lymphoid blast cells, granulopoietic cells and macrophages led to a maximum cell count on day 13. This growth pattern is similar to that for the cul- ture of human bone marrow cells in diffusion chambers observed by B^yum et al. (11), Boecker et al. (8) and Squires (59). Mature megakaryocytes and erythrocytes were seen, and since only lymphocytes and monocytes were implanted, the appearance of these cells is good evidence that even if the human pluripotent stem cell does not proliferate in diffusion chambers, then at least all three committed precursor cells do so. 166

Although the capacity of the human granulopoietic progenitor cell (CFU-c) to proliferate and differentiate in vitro has been well defined, its capacity for self renewal and the extent to which its numbers are repli- cated by inflow from more primitive stem cell compartments, has not yet been extensively investigated. The present study extends the observation done by Hoelzer et al. (35) that the absolute numbers of CFU-c can increase substantially by short term pre-culturing in diffusion chambers. This phenomenon of increase in absolute numbers of CFU-c was : Iready known from pre-culturing experiments in liquid medium (44, 50). There would seem to be three, not necessarily mutually exclusive, explanations for this phenomenon. 1. more primitive progenitor cells are circulating, which differentiate towards the CFU-c, owing to the culture environment; 2. CFU-c present in the blood are triggered into self- replication; 3. CFU-c in Go-phase of the mitotic cycle are activated in the diffusion chamber system. The present data do not allow us to distinguish between these possibili- ties. The rapidity of the increase in CFU-c in diffusion chambers favours the possibility of self-renewal, since recruted more primitive progenitor cells would have to differentiate and proliferate immediately to account for the observed increase. Such an expected delay has been observed in murine bone marrow cultured in diffusion chambers in which an increase in CFU-s is not seen until after one or two days (61). Testa and Lajtha (63) suggested that CFU-c have a capacity for self- renewal limited to approximately five divisions, at least in the regenera- ting mouse bone marrow system, and Sutherland et al. (62) postulated from their short-term liquid culture studies on murine bone marrow that self-renewal alone could not account for the enormous increase they observed in CFU-c within two days. There is sufficient evidence from animal experiments, that indeed a more primitive progenitor cell is triggered to proliferate and differentiate towards more mature progenitor cells such as the CFU-c. Sutherland et al. (62) demonstrated by means of velocity sedimen- tation that this more primitive progenitor cell resembles the CFU-s. The most convincing evidence was presented by Kurrle et al. (42) in rats, where the CFU-c pool in the bone marrow was depleted by hydroxyurea. This "depleted" bone marrow was then cultured in diffusion chambers and a substantial increase in CFU-c was observed, suggesting an influx from earlier progenitor cells. The same observations were also done in mice (55). That hydroxyurea does not affect the CFU-s itself was demon- 167

strated by Haas (29), who demonstrated that hydroxyurea-pretreated bone marrow is able to repopulate lethally irradiated rats. In the human situation, Jacobson et al. (39,40,41) demonstrated that this more primitive granulopoietic precursor cell is in fact the same pre- cursor cell that gives rise to colonies in the in vivo diffusion chamber sys- tem. This was demonstrated by differences in velocity sedimentation and the percentage of cells in S-phase between the CFU-d and CFU-c. Additional evidence that this more primitive progenitor cell differs from the CFU-c has been presented by O'Hara (33), who demonstrated the absence of anti-i on this primitive precursor cell, which is in contrast to its presence on the CFU-c. This increase in committed progenitor cell by preculturing in diffusion chambers was also demonstrated for the BFU-e, but not for the CFU-e (28) and for eosinophilic progenitor cells, which substantiate the fact that the eosinophilic precursor cell is not identical with the granulopoietic precursor cell. With the methyl-methacrylate embedding technique it was demon- strated that human committed progenitor cells can produce discrete colonies within the plasma clot diffusion chambers. Colonies from dif- ferent cell lines were observed in the same chambers and in varying total number of cells per colony. Very small and large neutrophilic colonies were found at the same time of harvest. Per colony, the cells were only partly synchronous in their stage of differentiation. Earlier or later differentiation patterns could be observed in other colonies. A striking observation was that even in the plasma clot matrix, poly- morphonucleated cells showed signs of cell migration, as was already ob- served in vitro cultures by Chang (14) and Metcalf (46). Most myeloid cell aggregates were either of neutrophilic or eosino- philic origin, while mixed neutrophilic-eosinophilic aggregates were not observed. This is in accordance with the findings with human bone marrow in fibrin clot diffusion chambers as described by Jacobson et al. (39). Although these authors described some mixed neutrophilic-eosino- philic colonies, in our opinion, these are artefacts due to manipulation of the plasma clot by centrifugation. Eosinophilic colonies in plasma clot diffusion chambers were very compact in structure and did not show any sign of cell migration. This is analogous to the in vitro data presented by Dao et al. (20, 21) and Dresch (22). Basophilic cells were rarely observed and were not found as cell aggregates, which is in accordance with in vitro culture findings done by Dao et al. (21) and Parmley (52). 168

Sometimes the neutrophilic cell aggregates were mixed with erythroid or megakaryocytic cells, which confirms the observation of Preisler et al. (54) in human bone marrow colonies in plasma clot diffusion chambers. However, these authors found very small neutrophilic colonies (usually < 10 cells) on day 10 of culture, which contrasts with the sometimes very large neutrophilic colonies observed in our system. A reason for this difference could be the low irradiation dose (400 Rad) used for the pretreatment of the diffusion chamber bearing mice. This presumption is confirmed by the fact that Gordon (27) also found very large colonies in agar clot diffusion chambers, when the mice had been pretreated with 900 Rad. Lymphoid colonies were not observed, which is also the case in long-term in vitro liquid cultures, using the Dexter system (64). Macrophage colonies were not observed either, but these cells were found scattered throughout the sections, presumably also by active cell migration. Erythropoietic single colonies were very rare, but most of the ery- throid cells were found intermingled with other cell lines. The reason for this is not clear, but it could be that myeloid and/or megakaryocytic cells provide a special environment for erythropoietic cells to differentiate in this system. Furthermore it was interesting to observe that mature erythrocytes were found at the borders of the erythroid cell aggregates. Erythroblastic islands, consisting of a histiocyte surrounded by maturing erythroid cells as can be found in bone marrow sections (7), were very seldom observed in the histological sections through the clots. The central histiocyte in such an erythroblastic island is supposed to provide stimulating factors for the developing erythroid cells. These erythroblastic islands were observed by Ben-Ishay (3) when rat bone marrow was cultured in diffusion chambers, but they were not observed in mouse bone marrow cultures in plasma clot diffusion chambers as described by Gerard et al. (24). In this connection it should be noted that, in contrast to peripheral blood, bone marrow cultures in diffusion chambers give rise to fibroblast and histiocytic colony formation. No clear cell migration out of the erythroid aggregates was observed in the plasma clot diffusion chambers, which is in contrast to the cell migrations found in in vitro BFU-e cultures (17). It is known that erythropoietic cells in general have a very low growth potential in diffusion chambers, except when rabbit progenitor cells are used (68). By manipulation of the diffusion chamber bearing host, such as hypoxia (12) or inducement of haemolytic anaemia by phenylhydra- 169

zine (60), the number and size of erythroid colonies in the diffusion chambers can be increased. Phenylhydrazine induces a haemolytic anae- mia in the experimental animal and as a consequence an increase in the number of CFU-s (53) and the committed erythroid progenitor cells CFU-e and BFU-e (30, 31). On the other hand, there is a decrease in the number of myeloid progenitor cells, probably on account of stem cell competition (28, 56, 58). The alterations in various peripheral blood counts induced by phenyl- hydrazine pretreatment of our NMRI-mice are in accordance with observations of Richard et al. (58), Hara et al. (30, 31) and Steinberg et al. (60). Our observation that erythroid colony formation could not be increased by phenylhydrazine pretreatment could be explained by the fact that, in contrast to the above-mentioned studies, the diffusion chamber bearing mice were also irradiated. However, the re-implantation of the chambers on day 7 may have abolished the erythroid proliferation and differentiation. This is substantiated by the fact that Walther et al. (67) demonstrated that if chamber recipient mice are subjected to a com- bination of 750 Rad irradiation and long-term non-interrupted hypoxia without chamber re-implantation, erythroid cell proliferation and differentiation can be enhanced. Finally, it should be mentioned that, unlike adult human bone marrow, foetal human liver stem cells give rise to spontaneous erythroid colonies in diffusion chambers in vivo (49). Megakaryocytic colonies of various volumes (ranging from 5-65 cells) were also observed when human peripheral progenitor cells were cultured in plasma clot diffusion chambers. Interesting was the fact that all these megakaryocytes showed evidence of platelet production. These mega- karyocytic cell aggregates were mostly very large. This is in contrast with the relatively low number of cells per megakaryocytic colony found by Jacobson et al. (38, 39), when human bone marrow cells were cultured in fibrin clot diffusion chambers. Vainchenker et al. (64, 65), using a plasma clot culture technique in vitro, described a megakaryocyte colony formation technique for human bone marrow precursors. The majority of the megakaryocyte colonies (75%) were pure, while the others were mixed with erythroblasts. This is in accordance with our finding that erythroid cells can be found inter- mingled with megakaryocytic cell aggregates. Ultrastructural studies have shown that the maturation of the megakaryocytes is essentially normal and that platelet shedding occurs. In contrast to our system, erythropoietin must be added to the culture plates to obtain megakaryocytic colony formation. That shedding of 170

platelets can take place was observed earlier by Levine (43), using guinea-pig bone marrow cells in liquid cultures. Also McLeod (45) and Metcalf (46) demonstrated this phenomenon by in vitro cultures of bone marrow cells. The three-dimensional analysis we performed on several whole colonies demonstrated that small colonies were perfect spheres, but larger colonies could have very capricious forms. Especially neutrophilic and megakaryo- cytic colonies showed large protrusions of cell groups and evidence of cell migration. In accordance with the in vitro studies the cells became more differentiated at the border of the colonies, but in contrast to the in vitro cultured colonies, a complete differentiation to many mature granulocytes was observed. This can be explained by the free diffusion of inhibiting substances produced by mature myeloid cells out of the cham- ber and colony stimulating factors into the chamber. Our three-dimensional studies revealed that it is conceivable that during the chamber centrifugation technique of Jacobson (36) parts of the megakaryocytic colony can be separated and subsequently counted as isolated colonies. This could result in an over-estimate of the total number of megakaryocytic colonies present in plasma clot diffusion chambers. Furthermore, in the case of the described mixed megakaryocytic- erythropoietic colony, the erythropoietic cells were hidden in the centre of the megakaryocytic cell cluster and can easily be missed when direct staining techniques for the plasma clot are used. In general, we had the impression that mixed colonies were formed by two or more committed progenitor cells, which proliferate and differen- tiate in very close vicinity. On the other hand, it cannot be entirely ex- cluded that pluripotent haemopoietic stem cells proliferate in the plasma clot diffusion chamber, as was also demonstrated in vitro by Fauser et al. (23). These authors proved the clonal origin of these mixed colonies, present in human bone marrow and peripheral blood, by mixing male and female cells and subsequently demonstrated single X or Y chromo- somes in the mixed colonies. It has been suggested by Barr et al. (2) that the diffusion chamber cultures may support the proliferation of human multipotent stem cells. However, this suggestion is not yet supported by results obtained by clonal techniques for human haemopoietic cells in the diffusion chamber technique. Clonal techniques in vitro are probably more suitable to elucidate this problem (32). The quantitative analysis of cell growth in plasma clot diffusion 171

chambers using histologic techniques proved to be very cumbersome, not in the least by the fact that the cell aggregates were non-homogeneously distributed. However, since various cell lines are proliferating and dif- ferentiating simultaneously, and the histological sections provide an optimal insight in the cellular events, this technique can contribute to a better understanding of colony formation and in various kinds of haema- tologic diseases.

4.5 REFERENCES

1. Algire GH, Weaver JM, Prehn RT: Growth of cells in vivo in diffusion chambers. I. Survival of homografts in immunized mice. J. Natl. Cancer Inst., 15: 439,19S4. 2. Barr RD, Whang-Peng J and Perry S: Hemopoietic stem cells in human peripheral blood. Science, 190: 284,1975. 3. Ben-Ishay Z: Ultrastructural analysis of erythroid colonies in diffusion chambers in irra- diated rats. Scand. J. Haemat., 14: 369,1975. 4. Ben-Ishay Z and Sharon S: Macrophages and/or fibroblasts in hematopoietic diffusion chamber cultures. Israel. J. Med. ScL, 13: 385,1977. 5. Benestad HB: Formation of granulocytes and macrophages in diffusion chamber cultures of mouse blood leukocytes. Scand. J. Haemat., 7: 279,1970. 6. Berman I and Kaplan HS: The cultivation of mouse bone marrow in vitro. Blood, 14: 1040, 1959. 7. Bessis M and Breton-Gorius J: Iron metabolism in the bone marrow as seen by electron microscopy. A critical review. Blood, 19: 635,1962. 8. Boecker WR, B0yum A, Carsten AJ and Cronkite EP: Human bone marrow and blood stem cell kinetics. Blood, 38: 819,1971. 9. B^yum A: Isolation of mononuclear cells and granulocytes from human blood. Scand. J. Clin. Lab. Invest., 21, suppl. 97: 77,1968. 10. B^yum A and Borgstr^m R: The concentration of granulocytic stem cells in mouse bone marrow, determined with thediffusion chamber technique. Scand. J. Haemat., 7: 294,1970. 11. B^yum A, Boecker W, Carsten AL and Cronkite EP: Proliferation of human bone marrow cells in diffusion chambers implanted into normal or irradiated mice. Blood, 40: 163,1972. 12. B^yum A, Carsten AL, Laerum OD, and Cronkite EP: Kinetics of cell proliferation of murine bone marrow cells cultured ir> diffusion chambers: effect of hypoxia, bleeding, erythropoietin injection, polycythaemia any Eradiation of the host. Blood, 40: 174,1972. 13. Breivik H and Benestad HB: Regulation of granulocyte and macrophage formation in diffu- sion chamber cultures of mouse haematopoietic cells. Exp. Cell. Res., 70: 340,1972. 14. Chang YT: Cinemicrographic studies on granulopoiesis. J. Natl. Cancer Inst., 59: 1479, 1977. 15. Chikkappa G, Carsten AL, Chanane AD and Cronkite EP: Culture of normal human blood cells in diffusion chamber systems. I. Granulocyte survival and proliferation. Exp. Hemat., 6: 28,1978. 16. Differentiation of normal and neoplastic hematopoietic cells. Clarkson B, Marks PA and Till JE (Eds.) Cold Spring Harbor Conference on cell proliferation, Vol. 5, book A and B. Cold Spring Harbor Laboratory, 1978. 17. Cormack D: Time-lapse characterizations of erythrocytic colony forming cells in plasma cultures. Exp. Hemat., 4: 319,1976. 172

18. Cronkite EP, Boecker W, Carsten AL.Chikkappa G, Joel D, Laissue J and Ohl S: The use of diffusion chamber cultures in the study of normal and leukcmic cell proliferation in man. Hemopoiesis in culture. William A. Robinson Ed. National Cancer Inst. Bethesda, Maryland, 185, 1974. 19. Dacie JV and Lewis SM: Practical Haematology, 5th edn. Churchill Livingstone, Edinburgh, 1975. 20. Dao C, Metcalf D, Zittoun R and Bilski-Pasquier G: Normal human bone marrow cultures in vitro: cellular composition and maturation of the granulocytic colonies. Brit. J. Haemat., 37: 127,1977. 21. Dao C, Metcalf D and Bilski-Pasquier G: Eosinophil and neutrophil colony forming cells in culture. Blood, 50: 833,1977. 22. Dresch C, Johnson GR and Metcalf D: Eosinophil colony formation in scmisolid cultures of human bone marrow cells. Blood, 49: 835,1977. 23. Fauser AA and Messner HA: Granuloerythropoietic colonies in human bone marrow, peri- pheral blood, and cord blood. Blood, 52: 1243,1978. 24. Gerard E, Carsten AL and Cronkitc EP: The prolifcrative potential of plasma clot erythroid colony forming cells in diffusion chambers. Blood cells, 4: 105, 1978. 25. Gidali J, Feher J and Antal S: Some properties of the circulating hemopoictic stem cells. Blood, 43: 573,1974. 26. Gordon MY and Blackett NM: Stimulation of granulocytic colony formation in agar diffu- sion chambers implanted in cyclophosphamide pretreated mice. Br. J. Cancer, 32: 51, 1975. 27. Gordon MY, Blackett NM and Douglas IDC: Colony formation by human haemopoietic precursor cells cultured in semi-solid agar in diffusion chambers. Brit. J. Haemat., 31: 103, 1975. 28. Greenberg HM and Robinson SH: Studies of erythroid progenitor cells in vivo and in vitro. Blood cells, 4: 129, 1978. 29. Haas RJ, Flad HD, Fliedner TM et al.: Correlation between cytokinctically resting lympho- cytes and bone marrow restoration: experiments using a discontinuous albumin gradient. Blood, 42: 209,1973. 30. Hara H and Ogawa M: Erythropoietic precursors in mice with phenylhydrazine-induued anemia. Am. J.of Hemat., 1: 453,1976. 31. Hara H and Ogawa M: Erythropoietic precursors in mice under erythropoietic stimulation and suppression. Exp. Hemat., 5: 141,1977. 32. Hara H and Ogawa M: Murine hemopoietic colonies in culture containing normoblasts, macrophages, and megakaryocytes. Am. J. of Hemat., 4: 23, 1978. 33. O'Hara CJ, Shumek KH and Price GB: The effect of anti-i on early myeloid progenitor cells in human bone marrow. In: Baum SJ and Ledney GD (eds.) Exp. Hemat. Today. New York, Springer Verlag, 51,1979. 34. Hoelzer D, Kurrlc E, Ertl U and Milewski A: Growth of human leukaemic peripheral blood cells in diffusion chambers. Europ. J. Cancer, 10: 579, 1974. 35. Hoelzer D, Harris EB, Slade M and Kurrle E: Growth of in vitro colony-forming cells from normal human peripheral blood leukocytes cultured in diffusion chambers. J. Cell. Physiol., 89: 89,1976. 36. Jacobsen N: Chamber centrifugation: A harvesting technique for estimation of the growth of human haemopoietic cells in diffusion chambers. Brit. J. Haemat. 29: 171,1975. 37. Jacobsen N and Fauerholdt L: Human granulocytopoietic colonies in diffusion chambers in mice: growth of colonies and the effect of host irradiation. Scand. J. Haemat., 16: 101, 1976. 38. Jacobsen N and Fauerholdt L: Quantitative aspects of an in vivo diffusion chamber assay for normal human hemopoietic colony forming units. Exp. Hemat., 5: 171,1977. 39. Jacobsen N: Proliferation of diffusion chamber colony-forming units (CFU-d) in cultures of normal human bone marrow in diffusion chambers in mice. Blood, 49: 415,1977. 173

40. Jacobsen N, Broxmeyer HE, Grossbard E and Moore MAS: Diversity of human granulo- poietic precursor cells: separation of cells that form colonies in diffusion chambers (CFU-d) from populations of colony forming cells in vitro (CFU-c) by velocity sedimentation. Blood, 52: 221,1978. 41. Jacobsen N, Broxmeycr HE and Moore MAS: Physical separation and kinetics of colony- forming cells in diffusion chambers in vivo (CFU-d) and colony-forming cells in agar in vitro (CFU-c). In: Baum SJ and Ledney GD (eds.) Exp. Hemat. Today, 243,1978. 42. Kurrle E, Hoelzer D and Harris EB: Growth and differentiation of rat bone marrow stem cells in diffusion chamber culture. Exp. Hemat., 5: 241,1977. 43. Levine RF: Culture in vitro of isolated guinea pig megakaryocytcs: recovery, survival, mor- phologic changes, and maturation. Blood, 50: 713,1977. 44. McCulloch EA and Till JE: Effects of short term culture on populations of hemopoietic progenitor cells from mouse marrow. Cell. Tiss. Kinet., 4: 11,1971. 45. McLeod DL, Shreeve MM and Axelrad AA: Induction of megakaryocyte colonies with platelet formation in vitro. Nature, 261: 492,1976. 46. Metcalf D: Hemopoietic colonies - in vitro cloning of normal and leukemic cells. In: Recent results in Cancer Research, Springer Vcrlag, Berlin, Heidelberg, New York, 1977. 47. Micklem HS, Anderson N and Ross E: Limited potential of circulating haemopoietic stem cells. Nature, 256: 41,1975. 48. Nettesheim P, Makinodan T and Chadwick CJ: Improved diffusion chamber cultures for cytokinetic analysis of antibody response. Immunol., 11: 427, 1966. 49. Niskanen E, Gale RP, Golde DW ct al.: Culture of mycloid and crythroid colonies from human fetal liver in vivo and in vitro. Exp. Hemat., 7, suppl. 6: 63,1979. 50. Niho Y, Tii" JE and McCulloch EA: Granulopoietic progenitors in suspension culture: a comparison of stimulatory cells and conditioned media. Blood, 45: 811,1975. 51. Ornstein L, Hudson A and Saunders A: Improving manual differential white cell counts with cytochemistry. Blood cells, 2: 557,1976. 52. Parmley RT, Ogawa M, Spicer SS and Wright NJ: Ultrastructure and cytochemistry of bone marrow granulocytes in culture. Exp. Hemat., 4: 75,1976. 53. Ploemacher RE, Soest van PL and Vos O: Kinetics of erythropoiesis in the liver induced in adult mice by phenylhydrazine. Scand. J. Haemat., 19: 424, 1977. 54. Preisler HD, Fliedner von U, and Shoham D: Growth of human marrow in plasma clot diffusion chambers. Scand. J. Haemat., 21: 313,1978. 55. Quesenberry P, Niskanen E, Syman M et al.: Growth of stem cell concentrates in diffusion chambers. Cell. Tissue Kinet., 7: 337,1974. 56. Quesenberry P, Levin J, Zuckerman K et al.: Stem cell migration induced by erythropoietin or haemolytic anaemia: the effects of actinomycin and endotoxin contamination of erythro- poietin preparations. Brit. J. Haemat., 41: 253,1979. 57. Rencricca N, Rizzoli V, Howard D, and Stohlman F Jr.: Stem cell migration and prolifera- tion during severe anemia. Blood, 36: 764,1970. 58. Rickard KA, Rencricca NJ, Shadduck RK, Monette FC et al.: Myeloid stem cell kinetics during erythropoietic stress. Brit. J. Haemat., 21: 537,1971. 59. Squires DJP: The growth of human bone marrow in diffusion chambers. Brit. J. Haemat., 29: 89,1975. 60. Steinberg HN, Handler ES and Handler EE: Assessment of erythrocytic and granulocytic colony formation in an in vivo plasma clot diffusion chamber culture system. Blood, 47: 1041,1976. 61. Shulman LN and Robinson SH: Maintenance of hemopoietic stem cells in diffusion chamber cultures. J. Lab. Clin. Med., 90: 581,1977. 62. Sutherland DJA, Till JE and McCulloch EA: Short term cultures of mouse cells separated by velocity sedimentation. Cell. Tiss. Kinet., 4: 479,1971. 174

63. Testa NG and Lajtha LG: Comparison of the kinetics of colony forming units in spleen (CFU-s) and culture (CFU-c). Brit. J. Haemat., 24: 367,1973. 64. Vainchenker W, Bouguett J, Guichard J and Breton-Gorius J: Megakaryocyte colony forma- tion from human bone marrow precursors. Blood, 54: 940,1979. 65. Vainchenker W, Guichard J and Breton-Gorius J: Growth of human megakaryocyte colonies in culture from fetal, neonatal and adult peripheral blood cells. Ultrastructural analysis. Blood cells, 5: 25,1979. 66. Velde te J, Burkhardt R, Kleiverda K, Leenheers-Binnendijk L and Sommerfield W: Methyl- methacryiate as an embedding medium in histopathology. Histopathology, I: 319, 1977. 67. Walther F and Schopow K: Human erythropoiesis in diffusion chambers. Scand. J. Haemat., 23:367,1979. 68. Willemzc R: Regulation of granulopoiesis studied by bone marrow culture in diffusion chambers implanted in rabbits. Thesis, Leiden 1979. 69. Wright EG, Lord BI, Dexter TM and Lajtha LG: Mechanisms of haemopoietic stem cell proliferation control. Blood cells, 5: 247,1979. 175

Chapter 5

HAEMOPOIETIC PROGENITOR CELLS IN HUMAN PERIPHERAL BLOOD IN MYELOPROLIFERATIVE DISORDERS

5.1 INTRODUCTION

The concept of myeloproliferative disorders was introduced by Dameshek (5) to group haematological conditions under the common denominator of morphologic characteristics, without taking into account etiologic factors. It has been suggested that these disorders may be caused by ab- normalities in the bone marrow micro-environment, an intrinsic stem cell defect, a disturbance in the haemopoietic regulator systems, or the presence of a neoplastic cell clone (7, 19). In addition, the mechanism of the release of mature and immature cells from the marrow into the blood stream is ill-understood, and might be disturbed in these condi- tions. Traditional diagnostic criteria are often insufficient for precise charac- terization of these disorders. New techniques based upon cell function and kinetic analysis of cell growth patterns may supplement traditional morphological and clinical criteria. One such approach to cell growth patterns in vitro is the agar-colony system, which allows enumeration of the number of functional myeloid progenitor cells (CFU-c). There have been several reports on in vitro colony formation in pa- tients with chronic myeloid leukaemia (11, 18, 19, 20, 26), and only a limited number of reports on patients with other types of myelo-pro- liferative disorders (14, 19, 22). Besides studies concerning the number of haemopoietic progenitor cells in bone marrow, a lot of attention is given to circulating erythroid (17) and myeloid (3, 12, 23, 27, 28) progenitor cells in diseases such as chronic myeloid leukaemia, myelofibrosis and polycythaemia vera. Most of these studies deal with the determination of absolute numbers of circulating progenitor cells which are cultured in vitro. Circulating progenitor cells have also been cultured in diffusion 176

chamber systems, but only from patients with myelofibrosis (4, 25). These studies suggest, that the blood of patients with myelofibrosis con- tains an increased number of granulocytic, erythroid and megakaryocytic progenitors. But, the pattern of their outgrowth within the diffusion chambers has never been subject of study. There is little information available about the morphology of colonies growing in vitro or in diffusion chambers in myeloproliferative disorders. Since the aspects of proliferation and differentiation of normal human haemopoietic progenitor cells in plasma clot diffusion chambers are well known (see chapter 4), it will be possible to determine whether circulating progenitor cells from patients with myeloproliferative disorders reveal normal growth patterns or not. It is assumed that progenitor cells in diffusion chambers are growing without the influence of the bone marrow micro-environment. Abnor- malities in proliferation and differentiation would under these circum- stances indicate an intrinsic defect in the progenitor cell itself.

5.2 MATERIALS AND METHODS

Eight patients with various types of myeloproliferative disorders and two patients with aplastic anaemia were studied. For the plasma clot diffusion chamber cultures, 100 ml peripheral blood in preservative-free heparin was collected from each patient and processed to obtain a CFU-c enriched cell suspension, as described in section 2.3.4. These cell suspensions were cultured in vitro, according to the technique described in section 2.2.16, to determine absolute numbers of CFU-c. Simultaneously, 1 x 10s cells/diffusion chamber were processed for culture and subsequent histological analysis as described in chapter 4. Due to the low number of cells harvested, only 2-4 diffusion chambers per patient could be put to culture. From 9 patients bone marrow histology was investigated by pro- cessing a Jamshidi bone marrow biopsy, according to the methacrylate embedding technique described by Te Velde et al. (29). The bone mar- row sections were reviewed independently by two pathologists. The histological parameters concerned cellularity, presence and/or atypia of each cell line and stromal features such as fibrosis and abnormalities of the micro-circulation. The morphological aspects of the cell aggregates in the plasma clot 177

diffusion chambers (harvested on day +13) were compared with the histological aspects of the bone marrow of each patient.

5.3 RESULTS

Relevant clinical and culture data are given in Tables 5.1 and 5.II.

Patient 1: Female, 30 years of age, suffering from Ph®-chronic myeloid leukaemia for 10 years. Until 8 years ago treated with busulphan and recently with thiotepa because of thrombocytosis. In the bone marrow histology erythropoiesis was diminished. Granulopoiesis with marked eosinophilia was intermingled with large numbers of predominantly small and imma- ture megakaryocytes. The marrow circulation seemed fairly normal, in the sinus lumina only scattered immature granulocytic and eosinophilic elements were observed. Microscopy of sections through the diffusion chambers revealed no viable cell aggregates; only some star-like cell ghosts without nuclei were found.

Patient 2: Female, 56 years old, suffering from primary thrombocytosis. Treatment consisted of one injection with radioactive phosphorus (= p32) and inter- mittent chemotherapy with thiotepa. In the bone marrow histology the number of mature megakaryocytes was markedly increased, associated with a slight increase of reticulin fibres. At the time of investigation, sinusectasy with intravasation of erythroblasts and immature granulo- cytes was only focally observed. In the diffusion chambers only two cell aggregates were found, al- though star-like cell ghosts were present.

Patient 3: Female, 52 years old, suffering from polycythaemia vera for 10 years, which recently transformed to thrombocytosis. No bone marrow his- tology available, but cytological examination showed an increase of all cell lines. In the diffusion chambers only a few aggregates were found, among which two eosinophilic aggregates. Their granulation was definitely 178

abnormal, showing fewer but large granula. Star-like cell ghosts were not conspicuous but present.

Patient 4: Female, 53 years old, suffering from polycythaemia vera for 4 years, controlled by chemotherapy and p32. In the bone marrow all cell lines were increased with additionally eosinophilia and only focal increase of reticulin fibres. The sinuses were partially dilated with immature granulo- cytes and a few megakaryocytes in their lumina. Cell growth in the diffusion chambers mainly consisted of granulocytic aggregates, which were small but had normal morphology. Megakaryo- cytic aggregates consisted of only a few megakaryocytes. Star-like cell ghosts were present in all sections.

Patient 5: Female, 51 years old, suffering from polycythaemia vera for 10 years, treated with chemotherapy because of thrombocytosis. Splenomegaly was present. In the bone marrow histology, diffuse reticulin fibrosis with focal condensation to collagen fibrosis was observed. The sinuses were wide with in their lumina predominantly young erythropoietic cells and several megakaryocytes. Although granulopoiesis was very abundant in the marrow, only focally immature granulocytes within the vascular sys- tem were observed. Yet in the diffusion chambers many and huge myeloid aggregates were seen without cytological abnormalities (Fig. 5.1). Megakaryopoiesis and ery thropoiesis were both abundantly present, forming large colonies (Fig. 5.2). The megakaryocytes tended to be rather small with one round nucleus, while at the cell border small and large PAS-positive thrombo- cytes were present. Star-like cell ghosts could be found, but were rare. Mature erythropoietic cells showed severe nuclear abnormalities, but no PAS-positive cytoplasmic granules. In the bone marrow histology PAS-positivity in the erythropoiesis was conspicuous, but stainable marrow iron was absent.

Patient 6: Male, 40 years old, for one year known to be suffering from polycythae- mia vera, with splenomegaly and moderate marrow fibrosis. Treatment had consisted of phlebotomy only. Marrow histology showed a pre- dominance of erythropoiesis and megakaryopoiesis, with rather atypical 179

Fig. 5.1: (Pat. No. 5) Granulocytic cell aggregate, intermingled with abnormal erythroid elements. Gallamine-Giemsa (x400).

Fig. 5.2: (Pat. No. 5) Megakaryocytic aggregates with (dark) platelet formation. In the vicinity PAS-negative cell fragments (platelets) of vaiying sizes. PAS (x400). 180

megakaryocytes. Granulopoiesis was only slightly increased with abnor- mal localization of the young elements within the central marrow areas and in the sinuses. The number of eosinophils was decreased. The sinus- oidal system was wide and in its lumina megakaryocytes and immature erythro- and granulopoietic cells were observed. Cell growth in the diffusion chambers was minimal; the few cell aggregates present however showed no abnormalities. Isolated eosino- phils were found. Star-like cell ghosts were abundant.

Patient 7: Male, 69 years old, only recently discovered to be suffering from myelo- fibrosis with splenomegaly and in the bone marrow moderate fibrosis and an increased megakaryo- and granulopoiesis. All cell lines exhibited moderate cellular atypia. The sinusoidal system was wide, with in its lumina abundant immature erythropoiesis, mature megakaryocytes and scattered immature myelopoiesis. In the diffusion chambers, massive outgrowth of huge aggregates of all cell lines, with only a few eosinophils was observed. Star-like cell ghosts were present, but rare.

Patient S.- Male, 51 years old, initially diagnosed as polycythaemia vera, but with increasing spleen size and progressive myelofibrosis. At the time of inves- tigation, the marrow was still hypercellular with conspicuous eosinophi- lia. The sinus sytem was moderately and predominantly focally dilated, with especially in these areas intrasinusoidal megakaryocytes, immature neutrophils and eosinophils. ' - wth in the diffusion chambers mainly consisted of megakaryocytes, in which the nuclei were often condensed. The cytoplasm was disinte- grated into small and large cell fragments, forming clusters of atypical thrombocytes (Fig. 5.3). At the same time numerous star-like ghosts were found (Fig. 5.4). Granulopoietic clusters mainly consisted of small immature cells, which were mostly in the same stage of differentiation.

Patient 9: Female, 54 years old, suffering from aplastic anaemia with absence of haemopoiesis in the bone marrow histology and absence of growth in the diffusion chambers. 181

Fig. 5.3: (Pat. No. 8) Small clusters of megakaryocytes dissolving into platelets of varying sizes, and with nuclear condensation. Gallamine-Giemsa (x400).

Fig. 5.4: (Pat. No. 8) Star-like cell ghost, probably representing the remnants of dissoluted megakaryocytes. In contrast, megakaryocytic cells in the same section are demonstrated. Gallamine-Giemsa (x400). 182

Patient 10: Male, 19 years old, suffering from aplastic. anaemia with absence of haemopoiesis in the bone marrow and no growth in the diffusion chambers.

Summarizing the features of the cell lines growing in diffusion cham- bers, it can be concluded that the spectrum of nuclear and cellular abnormalities resembles the spectrum of abnormalities in the bone marrow. Erythropoiesis in the normal control* diffusion chambers often showed slight nuclear abnormalities in the mature forms. In one normal control we observed PAS-positive granules in the cytoplasm of maturating cells. Yet in the patients, several of whom showed PAS-positivity in the bone marrow (although partly associated with iron-negative polycythaemia vera), no PAS-positivity was observed in the diffusion chambers. The difference between these patients and the normal controls es- pecially consisted in the number of cells in erythroid aggregates. In some patients, large aggregates of pro-erythroblasts and large fields of matura- ting erythroids were found, while in the normal controls (on day +13) erythropoiesis mainly consisted of small nests of maturating cells. Granulopoiesis in the neutrophilic lineage differed from normal con- trols mainly in the amount of cells per aggregate. In a few instances the aggregates consisted of cells in the same stage of maturation, which phenomenon was less evident in the smaller aggregates observed in nor- mal controls. Migration of mature neutrophils was observed in material from both patients and normal controls. Megakaryopoiesis in the patient material showed the most conspi- cuous abnormalities. The cytological details differed from patient to patient, but mature megakaryocytes sometimes consisted of small cells with one round nucleus and production of rather uniform thrombocytes at the cell borders. In others, bizarre megakaryocytes were disintegrating into thrombo- cytes of various sizes and degree of PAS-positivity. In several patients, the diffusion chamber sections showed an abundancy of dead cells resulting in star-like cell ghosts. Within these star-like structures neither nuclei nor cytoplasmic details could be detected. These cell ghosts were found scattered through the sections, and were hafdly ever clustered in aggregates. Although their distribution resembled the aspect of migrating * Normal control = progenitor cells obtained from normal individuals and cultured in diffusion chambers as described in chapter 4. 183 macrophages, patient no. 8 showed the simultaneous appearance of cell ghosts, dissolving megakaryocytes and bizarre and large megakaryocytes with intact nuclei and cytoplasm. Abundancy of cell ghosts was found in patients no. 6 and 8, while in patients no. 2 and 4 their number was lower. In the other patients some cell ghosts could be found, while these were never observed in aplastic anaemia. Eosinophils in the normal controls were mostly found in compact aggregates. In the patients they were most often found as scattered, iso- lated elements within the fibrin clot. In one patient the granulation was clearly abnormal in distribution, form and stainability, which confirmed the observations that eosinophils in bone marrow can be abnormal in myeloproliferative disorders.

Between the two methods of culturing peripherally circulating pre- cursor cells, namely the in vitro CFU-c and the diffusion chamber tech- nique, no correlation was found for patiens with myeloproliferative disorders. Cells from some patients did grow in diffusion chambers, while no CFU-c growth in vitro was observed. In the bone marrow histology of patients with myeloproliferative dis- orders, cellular hyperplasia was generally observed. Yet, no correlation existed between the amount of proliferation within the marrow paren- chyma and the outgrowth of precursor cells from the peripheral blood in diffusion chambers. Furthermore, a clear correlation between outgrowth in diffusion chambers and the clinical diagnosis, the application of chemotherapy, and blood cell counts could not be established in this limited number of patients. 184

5.4 CONCLUSIONS AND DISCUSSION

Conclusions

1) With the diffusion chamber system haemopoietic progenitor cells can be detected in the peripheral blood of patients with myeloprolifera- tive disorders, even in the absence of "in vitro" CFU-c growth. 2) The morphology and distribution of cell aggregates in diffusion chambers in myeloproliferative disorders is abnormal, as compared to normal controls described in chapter 4, indicating an intrinsic pro- genitor cell defect. 3) Star-like cell ghosts observed on day +13 of diffusion chamber culture are probably disintegrating megakaryocytes. They might reflect the presence of abnormal megakaryocytic precursors, differing in rate of proliferation and differentiation as compared to normal megakaryo- cytic precursors. 4) In aplastic anaemia with absent haemopoiesis in the bone marrow, circulating haemopoietic precursor cells can neither be detected with the CFU-c nor the diffusion chamber technique.

Discussion

The myeloproliferative disorders — polycy thaemia vera, idiopathic throm- bocythaemia, myelofibrosis and chronic myeloid leukaemia — may be regarded as diseases resulting from an irreversible change in the prolifera- tive behaviour of the haemopoietic stem cell in both medullary and potential extramedullary sites (7). The use of genetic markers, such as glucose-6-phosphate dehydro- genase (G-6-PD) isoenzyme analysis has shown that blood cells in poly- cythaemia vera and myelofibrosis, like those in chronic myeloid leukae- mia, are derived from a single clone of cells (6). Originally it was thought that the marrow fibrosis seen in some cases of this type was also part of the disordered haemopoietic cell growth. However, studies with the same marker reveal that cultured fibroblasts, taken from the marrow of patients with myelofibrosis and chronic myeloid leukaemia, are not derived from the neoplastic clone; thus it seems that myelofibrosis in these syndromes is a reaction by normal stromal cells to abnormal haemopoietic cells directly or indirectly by changes induced in their environment (16). 185

Due to the development of various in vitro progenitor cell culture systems, more information is now available concerning the patho-physio- logy of these disorders. However, most of these earlier studies deal with the quantitative aspects of progenitor cells, measured with various in vitro techniques and not with the morphological aspects of colony for- mation. For instance, it was reported by Rickard et al. (27) that untreated patients with chronic myeloid leukaemia generally had an increased CFU-c concentration in the bone marrow. This parameter was often re- duced in patients who had commenced chemotherapy. In the peripheral blood, CFU-c concentrations were greatly increased, up to 1,000-60,000 times the normal value (18), although these values were normal when the patients were in clinical remission (27). Also a good correlation existed between the total number of white blood cells and the counted number of CFU-c (2, 18, 21). The CFU-c in bone marrow and peripheral blood of these patients had the same percentage of cells in S-phase as normal controls (2), while mature granulocytes obtained from the peripheral blood did not inhibit CFU-c proliferation in vitro (2), suggesting an abnormal regulation mechanism which allows a tremendous expansion of the myeloid cell production. Furthermore, an increased number of light buoyant, density CFU-c was frequently observed, with a preferential accumulation of these cells in the peripheral blood (2). There is suggestive evidence, that the majority of these circulating progenitor cells are released by the spleen and not by the marrow (1,2, 23). However, another possibility is that it results from seeding of stem cells, that are released prematurely from the disorganized marrow, into the spleen. In polycythaemia vera, there is an expanded erythroid progenitor cell compartment in the bone marrow and peripheral blood, which maintains a normal responsiveness to erythropoietin modulation (9, 10, i /, 30). A tentative synthesis of present evidence suggests, that the pathogenesis of polycythaemia vera relates to evolution of an abnormal pluripotent stem cell with subsequent expansion of committed progenitor cell pools, occurring most prominently in the erythroid line. In patients with myelofibrosis, the number of CFU-c in the bone marrow is reduced (27), while the blood was shown to contain increased numbers of granulocytic precursor cells (3, 12, 28); in the same order of magnitude as found in patients with chronic myeloid leukaemia (11). In myelofibrosis a correlation between the total number of white blood 186

cells in the peripheral blood and thenumber of CFU-c was also observed (12). As in chronic myeloid leukaemia, in this disorder high numbers of CFU-c were detected in the splenic venous system (8). Although these quantitative aspects give some insight in the patho- physiology of these disorders, little attention is given to the morpho- logical aspects of progenitor cell colony formation in vitro or in diffusion chamber systems. Abnormalities in this resepct would then be an indica- tion for intrinsic defects in the cell function. This finding would give additional support for a primary cellular aberration instead of defects in the bone marrow environment or humoral regulation mechanisms. It was found by Grande et al. (13) that cultures from untreated patients with chronic myeloid leukaemia contained colonies which exhibited morphologically normal in vitro differentiation, except some eosinophilic colonies which could show morphological and cytochemical abnormalities. In contrast to the in vivo situation, the myeloid cells in these colonies can accumulate normal levels of leucocyte alkaline phosphatase (15), suggesting the existence of extracellular factors which inhibit the function of the white blood cells in these patients. However, these results can also be regarded as artefacts induced by the culture environment. Results of earlier conventional diffusion chamber culture studies of blood cells from patients with myelofibrosis suggest not only the presence of increased numbers of granulocytic, but also erythrocytic and mega- karyocytic precursors (4, 25). The proliferation and maturation, based on cytological aspects of the cell lines, was reported to be normal. However, the contribution of our study was a clear demonstration of abnormal cell proliferation and differentiation, especially in patients with myelofibrosis. In particular, the abnormalities in the megakaryocytic cell line were striking. It was remarkable that in one patient with poly- cythaemia vera, huge and abnormal erythroid colonies were observed, while it is known that erythroid progenitor cells have a very low growth capacity in diffusion chambers. The fact that we observed cytological abnormalities in the various cell lines, in contrast to other investigators, can be explained by the fact that methyl-methacrylate embedded plasm clot diffusion chambers allow a better histologic evaluation than the conventional diffusion chambers, where cytological smears are processed. Our findings, although the num- ber of patients is limited, are very suggestive for intrinsic cell defects in myeloproliferative disorders in all cell-lines, such as abnormal proliferative and differentiating capacities in the erythroid cell-line in polycythaemia vera. 187

In some patients no CFU-c activity in vitro could be detected, while in the diffusion chambers cell growth was observed. This phenomenon was observed by Niskanen et al. (24) in normal individuals, when peripheral progenitor cells were cultured both in vitro and in diffusion chamber systems. Their study suggests that human peripheral blood contains two types of progenitor cells that differentiate along the granulocytic path- way. They are separable by their ability to form colonies in vitro (CFU-c) and in diffusion chambers. Kinetic studies suggest that the colonies in the diffusion chambers are derived from an intermediate between the still hypothetical human pluripotent stem cell and the CFU-c (24). Clearly, the exact localization of the defect in replication and differentiation is still to be studied, together with an analysis of the regulatory system. But OUT findings suggest that diffusion chamber cultures can supply additional information concerning the presence and differentiating behaviour of haemopoietic precursor cells in various myeloproliferative disorders.

5.5 REFERENCES

1. Barrett AJ, Longhurst PA and Humble JG: Changes in peripheral blood CFU-c during splenic irradiation in a patient with CGL. Exp. Hematol., 4: 151,1976. 2. Berthier R, Douady F and Hollard D: Cellular factors regulating granulopoiesis in myeloid leukaemia. Blood cells, 3: 461,1977. 3. Chervenick PA: Increase in circulating stem cells in patients with myelofibrosis. Bood, 41: 67,1973. 4. Chikkapa G, Carsten AL, Charana AD, et al.: Increased granulocytic, erythrocytic, and megakaryocytic progenitors in myelofibrosis with myeloid metaplasia. Am. J. Hematol., 4: 121,1978. 5. Dameshek W: Some speculations on the myeloproliferative syndromes. Blood, 6: 372,1951. 6. Fialkow PJ, Denman AM, Singer J, Jacobson RJ and Lowenthal MN: Human myelopro- liferative disorders: clonical origin in pluripotent stem cells. In: Differentiation of normal and neoplastic hematopoietic cells, Book A. Ed.: Clarkson B, Marks PA and Till JE. Cold Spring Harbor Conferences on cell proliferation, volume 5,131,1978. 7. Gilbert HS: A reappraisal of the "myeloproliferative disease" Concept. J. Mt Sinai Hosp., 37: 426,1970. 8. Goldberg J, Tice D, Nelson D, Landaw SA and Gottlieb AJ: The spleen as a site of colony forming cell production in primary myelofibrosis (Abstract). Blood 50, suppl. 1: 148,1977. 9. Golde DW and Cline MJ: Pathogenesis of polycythemia vera - new concepts. Am. J. Hema- tol., 1: 351,1976. 10. Golde D, Bersch N and Cline MJ: Polycythemia vera: hormonal modulation of erythro- poiesis in vitro. Blood, 49: 399,1977. 11. Goldman JM, Th'ng KH and Lowenthal RM: In vitro colony forming cells and colony stimulating factor in chronic granulocytic leukaemia. Br. J. Cancer, 30:1,1974. 12. Goldman JM, Seitanides B, Ruutu T and Th'ng KH: Granulocyte committed progenitor cells in the blood of patients with myelosclerosis. Scand. J. Haemat, 21: 318,1978. 188

13. Grande M and Liso V: Cy to chemical study of the cell colonies from agar culture of normal bone marrow and of peripheral blood from patients with chronic myeloid leukaemia. Nouv. Rev. Franc. d'He'matol., 18: 563,1977. 14. Greenberg P, Mara B, Bax I, Brossel R and Schrier S: The myeloproliferative disorders - correlation between clinical evolution and alterations of granulopoiesis. Am. J. Med., 61: 878,1976. 15. Hellmann A and Goldman JM: Alkaline phosphatase activity of chronic granulocytic leu- kaemic neutrophils in agar culture. Scand. J. Haemat., 24: 237, 1980. 16. Jacobson RJ, Salo A and Fialkow PJ: Agnogcnic myeloid metaplasia clonal proliferation of hematopoietic stem cells with secundary myelofibrosis. Blood, 51: 189,1978. 17. Lacombe C, Casadevall N and Varet B: Polycythemia vera: in vitro studies of circulating erythroid progenitors. Brit. J. Haematol., 44: 189,1980. 18. Moberg C, Olofsson T and Olsson I: Granulopoicsis in chronic myeloid leukaemia. I. In vitro cloning of blood and bone marrow cells in agar culture. Scand. J. Haemat., 12: 381,1974. 19. Moore MAS, Williams W and Mctcalf D: In vitro colony formation by normal and leukaemic human haematopoietic cells: characterisation of the colony-forming cells. J. Natl. Cancer lnst.,50: 603,1973. 20. Moore MAS: In vitro studies in the myeloid leukaemias. Cleton MFJ, Crowther D and Malpas JS (Eds.) Advances in acute leukaemia, page 161, ASP-biological and medical press. Amsterdam, 1974. 21. Moore MAS: Agar culture studies in CML and blastic transformation. Ser. Haematol., 8: 11,1975. 22. Moore MAS: Marrow cultures in polycythemia vera and myelofibrosis. Cultures de cellules he'matopoie'tiques in vitro. Les seminairesde PInserm, 10-12 Mars 1975,42,167. 23. Muller-B<5rat CN, Wantzin GL, Philip P et al.: Agar culture studies of bone marrow, blood, spleen and liver in chronic myeloid leukemia. Leukemia Research, Vol. 1, No. 2/3, 123, 1977. 24. Niskanen E, Olofsson T and Cline MJ: Hemopoietic precursor cells in human peripheral blood. Am. J. Hematol., 7: 201,1979. 25. Ohl S, Carsten AL, Chanana AD et al.: Increased erythrocytic and neutrophilic progenitors in myelofibrosis with myeloid metaplasia. Eur. J. Cancer, 12: 131, 1976. 26. C.ofsson T and Olsson I: Granulopoiesis in chronic myeloid leukaemia. II. Serial cloning of blood and bone marrow in agar culture. Blood, 48: 351,1976. 27. Rickard KA, Brown RD, Wilkinson T and Kronenberg H: The colony forming cell in the myeloproliferative disorders and aplastic anaemia. Scand. J. Haemat., 22: 121,1979. 28. Schmitt M and Mayer S: La culture "in vitro" des cellules-souches du sang circulant au cours des myeloscleroses primitives. Nouv. Rev. Franc. d'Hematol., 18: 573,1977. 29. Velde J te, Burkhardt R, Kleiverda K, Leenheers-Binnendijk L and Sommerfield W: Methyl- methacrylate as an embedding medium in histopathology. Histopathology, I: 319, 1977. 30. Zanjani ED, Lutton JD, Hoffman R and Wasserman LR: Erythroid colony formation by polycythemia vera bone marrow in vitro - dependence on erythropoietin. J. Gin. Invest., 59:841,1977. Table S.I: Summary of the clinical aspects and diffusion chamber data of 8 patients with myeloproliferative disorders and 2 patients with aplastic anaemia.

Peripheral blood counts Diffusion chamber (DC) cultures - Quantitative evaluation -

Pat Sex/ Clinical Current or Hb Retic. WBC Platelets Spleno CFU-c*/ Total* Total# Total# Total* Total* 9 9 4 # age diagnosis pastchemo- (mmol/1) °/00 (xlO /l) (xlO /l) megaly 2xlO of DC ofmye- of mega- ofery- of mixed therapy cells scored loidCA karyo- throid CA scored *** scored cyticCA CA in all in all scored scored DC DC in all in all DC DC

1 9.30 Chronic +(Busulfan) 7.1 8 9.2 430 0 3 0 0 0 0 myeloid and leukaemia Thiotepa) 2 9,56 Primary +(32 p and 8.9 21 7.7 235 0 2 2 0 0 1 thrombo- Thiotepa) cytosis 3 9.52 Thrombo- +(32 p and 8.S 15 6.5 440 0.3+0-1 3 3 0 0 1 cytosis Thiotepa) (2 eos) OO 4 9.53 Polycythae- +(32 p and 10.2 11 9.3 325 1.6±0.4 3 12 5 0 0 mia vera Thiotepa) (3 eos) 5 9.51 Polycythae- +(Thiotepa) 7.7 21 14.0 540 + 0 4" 2.2±0.2 2.1±0.2 1.9+0.2 1.6+0.2 mia vera and phlebotomy) 6 d,40 Polycythae- -(phlebotomy) 9.0 32 6.8 280 + 0.3+0.1 3 3 3 0 0 mia vera 7 d,69 Myelo- -(phlebotomy) 8.9 23 8.0 245 ++ 53.6±2.1 3** 5.4±1.4 37.2±2.3 4.0+2.6 4.2±1.1 fibrosis 8 d,51 Myelo- -(phlebotomy) 10.0 12 10.6 250 ++ 0 2 6 9 2 2 fibrosis

9 9,54 Aplastic -(none) 4.0 1 0.4 4 0 2 0 0 0 0 anaemia

10 6,19 Aplastic -(none) 5.1 16 0.5 2.5 0 2 0 0 0 0 anaemia

* : normal value: 8.0/2xl04 cells ** : in these chambers abundant cell-aggregate formation was observed, therefore scored as mean ± s.e.m./per section *** : 12 sections per DC were avaluated Table 5.H: Summary of the bone marrow histology and diffusion chamber data of 8 patients with myeloproliferative disorders and 2 patients with aplastic anaemia

Bone-marrow histology # of cell Pat Sex/ Clinical Current or Spleno- Cellul- Fibro- Sinus- Lineages aggregates CFU-c/ Recognizable # age diagnosis past chemo- megaly arity sis cctasy present in: in diffusion 2x104 precursors in therapy paren- sinus** chambers (sec ct-Us peripheral chyma table 5.1) blood smears

1 9,30 chronic + (Busulfan) er 0 0 Some meta- and myeloid and gr ++ 0 myelocytes. No leukaemia Thiotepa) eo ++ 0 erythroblasts meg ++ 0(shades ±) 2 9,56 primary +(32 p and er N 0 None thrombo- Thiotepa) gr N 2 cytosis eo 0 meg 0(shades +) 3 9,52 Thrombo- +(32 p and er +' 0 0.3±0.1 None cytosis Thiotepa) gr + 1 eo + 2 in eg + 0 (shades ±)

4 9,53 polycythaemia +(32 p and er 0 1.6+0.4 None vera Thiotepa) 9 eo 3 meg 5 (shades +) 5 9,51 polycythaemia +(Thiotepa + 1.9+0.2 Few erythro- vera and 2.2+0.2 blasts phlebotomy) eo 0 meg 2.1 ±0.2 (shades ±) 6 d,40 polycythaemia -(phlebotomy) + er 0 0.3±0.1 Few metamyelo- vera gr 3 cytes eo ± (scattered) meg 3 (sliades+++) 7 6,69 myelofibrosis -(phlebotomy) ++ er 4.0±2.6 53.6±2.1 Erythroblasts, gr 5.4±1.4 meta- and myelo- eo ± (scattered) cytes meg 37.2±2.3(shades±) 8 d,51 myelofibrosis -(phlebotomy) ++ er + 2 None gr + 6 eo ++ 0 meg + 9 (shades+++)

9 9,54 aplastic -(none) er 0 None gr 0 eo 0 meg 0 (shades -) 10 c5,19 aplastic -(none) 0 None anaemia 0 eo 0 meg 0 (shades -)

* No biopsy available, only cytology; ** Polymorphonuclear leucocytes and erythrocytes excluded; N = normal; er = erythroid;gr = granulocytic;eo = eosinophilic: meg = megakaryocytic. Shades = star-like cell-remnants. 192

SUMMARY

Chapter 1 is an introduction into the present views on circulating haemo- poietic progenitor cells in animals and men. Most implantation studies have been performed in dogs; in which it was established that a consistent haemopoietic engraftment can be achieved with mononuclear cells obtained from the peripheral blood. The number of mononuclear cells required for a haemopoietic engraft- ment varies between 0.32 and 1.63 x 109/cells per kg body weight, cor- responding with 0.5-1.5 x 104 CFU-c per kg body weight. The number of peripheral blood CFU-c collected with different leucapheresis techniques can be increased by chemotherapy, sequential pheresis and lymphocyte mobilizing agents such as Dextran sulphate, Pyran and Poly vinyl sulphuric acid (PVSA). It was also established that the rate of haemopoietic engraftment was directly correlated with the number of CFU-c infused and that cryopreservation of peripheral haemo- poietic progenitor cells for prolonged periods was feasible with only a limited loss in CFU-c number and/or function. Furthermore, it was found that the graft versus host reaction in dogs can be prevented by using cells which were separated on discontinuous albumin gradients. In humans, it was established that haemopoietic progenitor cells, as monitored by the in vitro CFU-c technique, are present in the peri- pheral blood in a concentration of approximately 60 CFU-c per 1 x 106 mononuclear cells, if a colony is defined as equal to or more than 20 cells. Using continuous flow cell separation between 6.3 and 18.7 x 109 mononuclear cells (mean: 12.4 x 109) can be collected, corresponding with 1.5-15.8 x 105 CFU-c (mean 8.3 x 105) during a single 4-hour pheresis procedure. Furthermore, it was established that sequential pheresis will increase the CFU-c yield with a mean of 134.8% (range: 36.8 to 232.8%). If the cell dose required for haemopoietic engraftment in dogs (± 1.6 x 104 CFU-c per kg body weight) is applied to men, then approximately 1.0 x 106 CFU-c in total are required for a 70 kg adult. This can be achieved with about 1 to 3 continuous flow cell separation procedures, without previous chemotherapy or the use of lymphocyte mobilizing agents. In this first chapter a review is presented about the different cell sepa- 193

ration procedures that have been performed to concentrate haemopoietic progenitor cells from human peripheral blood. In most other studies, multi-step cell separation techniques with density gradients were used. It was established that the CFU-c were present in the "null cell" lymphocyte population and that functional separation between haemo- poietic progenitor cells and immunocompetent cells could not be achieved by density separation alone. Therefore, specific adaptations of the tech- nique are necessary for the removal of monocytes and lymphocytes. It was also demonstrated that the erythroid precursor cells are present in human peripheral blood. Although no assay is available for the human pluripotent haemopoietic stem cell, the quantitative and qualitative aspects of the CFU-c reflect their presence, as was established in various animal studies. It is to be expected that progenitor cells can be easily harvested in sufficient numbers to be used for transplantation purposes, when better methods for haemopoietic progenitor cell mobilization in the peripheral blood, leucapheresis techniques and improved cell separation techniques have been developed.

Chapter 2 deals with the cell separation technique, that was developed to obtain a purified CFU-c suspension by a limited number of cell sepa- ration steps. Initially it was established that in leucocyte suspensions the CFU-c content per 5 x 10s cells was 10.4 ± 1.9, which correlated with a mean absolute CFU-c concentration of 20,800 per 1 x 109 leucocytes. After removal of the granulocytes by Ficoll-Isopaque density centrifugation, 27.1 ± 10.3 CFU-c per 5 x 10s mononuclear cells were counted. By the use of linear density gradient centrifugation it was demonstrated that the CFU-c were localized in the light density region, with a peak at 1.065 g/ml. In the same region the B-lymphocytes and monocytes were localized, while the T-lymphocytes revealed a peak in the higher density region. It was found that the CFU-c did not adhere to nylon wool columns, neither did they bear Fc-receptors for IgG as was studied with various EA-rosette techniques. A two-step cell separation technique was developed to concentrate and purify the CFU-c. In the first step monocytes were eliminated by pre-incubation with carbonyl-iron and centrifugation in a density 1.077 g/ml Ficoll-Isopaque gradient. To remove the T-, B-lymphocytes and Fc- receptor bearing lymphocytes, a triple rosette combination was developed 194

by which these three cell types were simultaneously eliminated during a second Ficoll-Isopaque (density 1.072 g/ml) centrifugation step. This resulted in an increase in CFU-c ratio from 1:121,951 (buffy coat sus- pensions) to 1:2.515; implying a purification factor of approximately 50-fold. However, although the CFU-c purification could be quickly achieved, the ultimate cell yield was only 0.26% and the CFU-c loss was substantial. However, compared to other existing methods, this approach is easy to perform and not time-consuming.

In chapter 3 experiments are described which were performed to define more precisely various in vitro characteristics of the myeloid haemopoie- tic precursor cell (CFU-c). The CFU-c enriched null cell suspensions obtained by the two-step density separation technique showed a very low immunological respon- siveness, as demonstrated by PHA-stimulation. In the mixed lymphocyte culture (MLC-test) it was observed that these null-cell suspensions had a normal capacity to respond, but a very high stimulating capacity. The latter phenomenon was not abolished by co-cultivation with isolated T-lymphocytes. Therefore, it is very unlikely that this increased stimula- ting capacity could be caused by suppressor cell elimination during the cell separation procedure. With various cytochemical staining techniques the CFU-c enriched cell suspensions did not reveal a specific pattern. Compared to human bone marrow, in which 15-20% of the CFU-c are in S-phase of DNA-synthesis as was demonstrated with the radioactive thymidine suicide technique, the peripheral circulating CFU-c revealed a lower percentage of cells in S-phase: ± 5%. Irradiation studies revealed that for both human bone marrow and peripheral blood the CFU-c were less radioresistent than clusters (colonies > 3 < 20 cells). For peripheral blood CFU-c a mean (± s.e.m.) Do-value of 65.7 ± 4.8 Rad was found, while clusters showed a Do-value of 87.4 ± 5.5 Rad. The irradiation dose rate did not alter the Do-value or extra- polation numbers. It was demonstrated that the CFU-c present in human peripheral blood showed a higher radiosensitivity than CFU-c in human bone marrow. Elimination of monocytes (and granulocytes) from the test suspensions induced an alteration in radiosensitivity parameters. For human bone marrow this resulted in disappearance of the shoulder in the survival curves and consequently a lower extrapolation number. Moreover, the 195

Do-values increased, which indicates a lower radiosensitivity. For peri- pheral blood progenitor cells the removal of monocytes and granulocytes from the test suspensions induced an increase in radiosensitivity in the low dose irradiation region. From these experiments it was concluded that the radiosensitivity of haemopoietic progenitor cells is influenced by the presence or absence of other haemopoietic cells. Finally, volume profile analysis of different mononuclear cell suspen- sions revealed that the CFU-c enriched null cells have a higher mean volume than unseparated mononuclear cell suspensions. However, there was a substantial overlap in volume distribution with other lymphocyte subpopulations.

In chapter 4 the proliferation and differentiation of haemopoietic pre- cursor cells in diffusion chambers is described. This system provides a more physiological environment than the in vitro CFU-c cultures. It was demonstrated that haemopoietic progenitor cells, present in human peripheral blood, can be induced to differentiate into mature erythrocytes, granulocytes as well as thrombocytes, by culturing them in conventional diffusion chambers. Furthermore, more immature precursor cells are thought to differen- tiate towards the CFU-c compartment, as was demonstrated by the fact that the number of CFU-c counted in vitro increased, if the peripheral blood cells were precultured in diffusion chambers. Histological analysis of colony formation in plasma clot diffusion chambers revealed neutrophilic colonies with asynchronous cell differen- tiation and signs of active migration of mature neutrophils. The megakaryocytic cell aggregates revealed signs of platelet forma- tion at the cell borders, while the erythroid cell aggregates, although not frequently observed, were able to form mature erythrocytes. Pretreat- ment of the irradiated host mice with phenylhydrazine did not increase the number of erythroid cell aggregates, nor the total number of cells per aggregate. Eosinophils and neutrophils were always found in separate colonies, supporting the thesis that both lineages have separate committed pro- genitor cells. Furthermore, the mature eosinophilic cell aggregates were very compact and did not show any sign of cell migration. Monocytic cell aggregates were very rare, while macrophages were frequently found scattered throughout the plasma clot. Cell aggregates consisting of lymphoid ceils were never observed. Aggregates consisting of a mixture of various cell lineages were found in small numbers. 196

Three-dimensional analysis of various colonies revealed that small colonies were perfect spheres, while large colonies showed bizarre struc- tures with signs of active cell migration and satellite formation. Analysis of mixed colonies suggested that the majority of those colonies did not consist of homogeneously mixed differentiating cells of various lineages, but were rather the result of cell migration from closely developing single cell aggregates. Especially erythroid cells showed a tendency to grow in the vicinity of megakaryocytic cells. These studies demonstrated that progenitor cells of erythro-, leuco- and megakaryopoiesis are present in human peripheral blood and can be induced to form colonies in the plasma clot diffusion chambers. Histo- logical analysis of these chambers provides a means to study the prolifer- ative and differentiating capacity of these progenitor cells.

In chapter 5 various techniques were applied to study the proliferative and differentiating capacity oi haemopoietic progenitor cells present in the peripheral blood of patients with myeloproliferative disorders. Most earlier studies were dealing with quantitative aspects of CFU-c in peripheral blood, while little attention was paid to colony morphology. It was demonstrated that the morphology and distribution of cell aggre- gates in plasma clot diffusion chambers in myeloproliferative disorders was abnormal with regard to cell numbers and cytological asepcts. These findings indicate that, whatever the role of changes in the micro-environment in the bone marrow might be, an intrinsic progenitor cell defect is present in myeloproliferative disorders. 197

SAMENVATTING

In hoofdstuk 1 wordt een overzicht gegeven van de huidige kennis om- trent hemopoiëtische voorlopercellen die voorkomen in het perifere bloed van mens en dier. Bij honden is aangetoond, dat transplantatie van hemopoiëtische cellen kan worden bereikt door gebruik te maken van mononucleaire cellen uit het perifere bloed. Het aantal cellen dat hiervoor nodig is varieert tussen 0,32 en 1,63 x 109 per kg lichaamsgewicht, hetgeen overeenkomt met 0,5 tot 1,5 x 104 CFU-c's per kg lichaamsgewicht. Het aantal CFU-c's dat kan worden verzameld uit het perifere bloed, door middel van verschillende leucapherese-technieken, kan men doen toenemen door voorafgaande chemotherapie, opeenvolgende leucaphe- rese-procedures en "lymfocyten-mobiliserende" stoffen zoals Dextran sulfaat, Pyran en polyvinyl sulphuric acid (PVSA). Ook werd aangetoond, dat de snelheid van het hematologisch herstel direct gecorreleerd is aan het aantal toegediende CFU-c's en dat boven- dien cryopreservatie van deze hemopoiëtische voorlopercellen voor zeer lange tijd mogelijk is, zonder groot verlies in aantal of functie. Boven- dien bleek het mogelijk de graft-versus-host reactie in honden te voor- komen door hemopoiëtische voorlopercellen te gebruiken, die waren ge- scheiden van immunocompetente cellen door middel van discontinue albumine gradiënten. Het aantal CFU-c's in het perifere bloed van de mens bedraagt 60 per 1 x 106 mononucleaire cellen, indien een kolonie gedefinieerd is als > 20 cellen. Door middel van een eenmalige 4 uur durende leucapherese- procedure kunnen tussen de 6,3 en 18,7 x 109 mononucleaire cellen met een gemiddelde van 12,4 x 109) verzameld worden, hetgeen overeen- komt met 1,5-15,8 x 10s CFU-c's (een gemiddelde van 8,3 x 105). Bovendien werd gevonden dat achtereenvolgende leucapherese-procedures het aantal CFU-c's in het perifere bloed doen stijgen met een gemiddelde van 134,8% (spreiding: 36,8 tot 232,8%). Indien de celdosis die bij honden nodig is om een hemopoiëtisch transplantaat te bewerkstelligen (± 1,6 x 104 CFU-c's per kg lichaamsge- wicht) zou gelden voor de mens, dan is voor een volwassene van 70 kg ongeveer 1,0 x 106 CFU-c's in totaal nodig. Dit kan worden verkregen door ongeveer 1 tot 3 leucapherese-procedures met het "continuous 198

flow" cel-scheidingsapparaat, zonder voorafgaande chemotherapie of toedieningen van "lymfocyten mobiliserende" stoffen. In dit hoofdstuk wordt verder een overzicht gegeven van de verschil- lende celscheidingstechnieken die zijn toegepast om humane hemo- poiëtische voorlopercellen uit het perifere bloed te isoleren. Gevonden werd dat de CFU-c's aanwezig zijn in de zgn. "nul-cel" lymfocyten- populatie en dat een functionele scheiding tussen hemopoiëtische voor- lopercellen en immunocompetente cellen door middel van dichtheids- gradiënt-centrifugatie alleen niet kan worden bereikt. Daarom zijn specifieke aanpassingen nodig om monocyten en lymfocyten te ver- wijderen. Eveneens werd aangetoond dat ook voorlopercellen van de erythroiUe reeks in het perifere bloed van de mens voorkomen. Alhoewel er geen techniek voorhanden is om de humane pluripotente hemopoiëtische stamcel aan te tonen, geven de kwantitatieve en kwali- tatieve aspecten van de CFU-c's toch indirect hun aanwezigheid aan, zoals in verschillende diermodellen is aangetoond. Het valt te verwachten dat hemopoiëtische voorlopercellen voor transplantatiedoeleinden op eenvoudiger wijze en in voldoende mate kunnen worden geoogst, indien er betere technieken voor mobilisatie van hemopoiëtische voorlopercellen in het perifere bloed, leucapherese-procedures en betere celscheidings- technieken ontwikkeld worden.

In hoofdstuk 2 wordt ingegaan op de celscheidingstechniek die is ont- wikkeld om in een beperkt aantal stappen een met CFU-c verrijkte cel- suspensie te verkrijgen. In eerste instantie werd aangetoond dat in onbewerkte leukocyten- suspensies het aantal CFU-c's 10,4 ± 1,9 per 5 x 10s cellen bedraagt, hetgeen overeenkomt met een absolute CFU-c-concentratie van 20.800 per 1 x 109 leukocyten. Na verwijdering van granulocyten door middel van Ficoll-Isopaque centrifugatie werden 26,1 ± 10,3 CFU-c's per 5 x 10s mononucleaire cellen geteld. Door middel van lineaire dichtheidsgradiënt-centrifugatie werd aan- getoond dat de CFU-c's zich bevinden in het lichte dichtheidsgebied met een piek bij 1.065 g/ml. In hetzelfde gebied bevinden zich de B-lymfo- cyten en monocyten, terwijl de T-lymfocyten een hogere dichtheidspiek vertonen. Voorts werd aangetoond dat de CFU-c's zich niet binden aan nylon- wol kolommen en ook geen receptoren dragen voor het Fc-gedeelte van het IgG-molecuul, zoals werd aangetoond met verscheidene EA-rosette- technieken. 199

Een twee-staps celscheidingstechniek werd ontwikkeld om CFU-c's te zuiveren en te concentreren. Tijdens de eerste stap werden monocyten verwijderd door centrifugatie op een Ficoll-Isopaque dichtheidsgradiënt van 1.077 g/ml, na pre-incubatie met carbonylijzer. Teneinde de T-, B-lymfocyten en lymfocyten met Fc-receptoren te verwijderen, werd een combinatie gemaakt van 3 typen rosettes, die gelijktijdig tijdens de tweede centrifugatie op een Ficoll-Isopaque gradiënt (met een dichtheid van 1.072 g/ml) konden worden verwijderd. Dit resulteerde uiteindelijk in een stijging van de CFU-c ratio van 1:121.951 (de uitgangsleukocyten suspensie) tot 1:2.515, hetgeen overeenkomt met een zuivering van ongeveer 50x. Alhoewel de CFU-c verrijking zeer snel en eenvoudig kon worden bereikt, was de uiteindelijke celopbrengst slechts 0,26% en het verlies aan CFU-c's dus aanzienlijk. In vergelijking met andere bestaande methoden is deze procedure echter gemakkelijk en tijdbesparend.

In hoofdstuk 3 worden een aantal experimenten beschreven die zijn verricht om de myelofde hemopoiëtische voorlopercel (CFU-c) beter te karakteriseren. De met CFU-c verrijkte nul-cel-suspensies, die zijn verkregen d.m.v. de twee-staps dichtheidsgradiënt-centrifugatie, vertoonden na PHA-simulatie een zeer gering reactievermogen. In de gemengde lymfocytenkweek werd een normaal reactievermogen gevonden, maar daarentegen een zeer hoog stimulatievermogen. Dit hoge stimulatievermogen werd niet geremd door co-cultivatie met geisoleerde T-lymfocyten. Het is dan ook onwaarschijn- lijk dat dit verschijnsel berust op verwijdering van suppressor cellen tijdens de celseparatie-procedure. Ondanks de toepassing van verschillende cytochemische kleurings- methodieken vertoonde de nul-cel lymfocytenpopulatie geen specifiek patroon. Door middel van radioactief thymidine kon worden aangetoond, dat van de CFU-c's in humaan perifeer bloed zich 5% in de S-fase van DNA- synthese bevindt, terwijl dit voor CFU-c's in humaan beenmerg 20% is. Bestralingsexperimenten toonden aan dat de CFU-c's gevoeliger zijn voor bestraling dan clusters (cel-aantal > 3 < 20). Dit geldt zowel voor de CFU-c's en clusters in humaan beenmerg als voorperifeer bloed. Voor de CFU-c's in perifeer bloed werd een gemiddelde (± s.e.m.) Do-waarde van 65,7 ± 4,8 Rad gevonden, terwijl dit voor clusters 87,4 ± 5,5 Rad bedroeg. De snelheid van bestraling had echter geen invloed op de Do-waarde of extrapolatiegetallen. 200

Ds CFU-c's die aanwezig zijn in het perifere bloed toonden een grotere gevoeligheid voor bestraling dan die in humaan beenmerg. Verwijdering van monocyten (en granulocyten) uit de testsuspensies resulteerde in een verandering van de bestralingsgevoeligheid. Met name voor humaan beenmerg leidde dit tot verdwijning van het schoudergebied uit de overlevingscurve en derhalve een lager extrapolatiegetal. Bovendien nam de Do-waarde toe, hetgeen duidt op een lagere bestralingsgevoeligheid. De bestralingsgevoeligheid, in het lage doseringsgebied, van CFU-c's in het perifere bloed, nam toe door verwijdering van monocyten en granulo- cyten uit de testsuspensies. Uit deze experimenten werd geconcludeerd dat de bestralingsgevoelig- heid van hemopoiëtische voorlopercellen beihvloed wordt door de aanwezigheid van andere bloedcellen. Ten slotte werd d.m.v. volume-profielanalyse aangetoond, dat de met CFU-c's verrijkte nul-cel suspensies een hoger gemiddeld volume hadden dan de ongescheiden mononucleaire cellen. Er was echter een vrij grote overlapping in de volumeprofielen van de verschillende lymfocyten- subpopulaties.

In hoofdstuk 4 wordt de analyse van proliferatie- en de differentiatie- capaciteit van hemopoiëtische voorlopercellen in humaan perifeer bloed in het diffusiekamersysteem beschreven. Cultivatie van hemopoiëtische voorlopercellen in conventionele diffusiekamers leidde tot differentiatie naar rijpe erythrocyten, granulo- cyten en thrombocyten. Verder werd uit deze experimenten de veronder- stelling getrokken, dat nog vroegere voorlopercellen gaan differentiëren in de richting van het CFU-c compartiment, doordat een hoger aantal CFU-c's "in vitro" werd geteld indien de testsuspensie een aantal dagen werd voorgekweekt in diffusiekamers. Door middel van histologisch onderzoek van plasma-clot diffusiekamers werd aangetoond, dat er neutrofiele kolonies aanwezig zijn die een asyn- chrone celdifferentiatie vertonen, alsmede tekenen van actieve migratie van de rijpe granulocyten. In de megakaryocytaire cel-aggregaten werden aanwijzingen gevonden voor produktie van thrombocyten, terwijl de spaarzame aanwezige erythroide cel-aggregaten in staat waren erythrocyten te produceren. Voorbehandeling van de bestraalde gastheer-muizen met fenylhydrazine leidde niet tot een toename van erythrofde cel-aggregaten, noch tot een toename van het aantal cellen per aggregaat. Eosinofiele leukocyten en granulocyten werden steeds in afzonderlijke 201

kolonies gevonden, hetgeen wijst op een aparte voorlopercel van de eosi- nofiele granulocyt. De rijpe eosinofiele cel-aggregaten waren altijd zeer compact van structuur en vertoonden geen tekenen van celmigratie. Monocytofde cel-aggregaten waren spaarzaam, terwijl macrofagen juist veelvuldig en verspreid door de plasma-clot werden aangetroffen. Lym- foi'de cel-aggregaten werden niet gevonden. Wel werden, in kleine hoeveel- heden, gemengde cel-aggregaten gezien. Drie-dimensionale analyse van verscheidene kolonies bracht aan het licht dat kleine kolonies een zuiver ronde vorm hadden, terwijl grotere kolonies zeer grillig van structuur konden zijn met tekenen van actieve celmigratie en satellietvorming. Analoog onderzoek van de gemengde kolonies maakte het waarschijnlijk, dat in de meeste gevallen de menging een gevolg was van celmigratie vanuit enkelvoudige celaggregaten, die toevallig vlak tegen elkaar aan lagen. Met name toonden de erythroide cellen de neiging tussen megakaryocytaire cellen te groeien. De in dit hoofdstuk beschreven onderzoekingen tonen aan, dat in humaan perifeer bloed voorlopercellen aanwezig zijn van de erythro-, leuko- en megakaryopoiese, die in het plasma-clot diffusiekamersysteem onderzocht kunnen worden op hun proliferatieve en differentiatie eigenschappen.

In hoofdstuk 5 worden de eerder beschreven technieken toegepast om de proliferatie- en differentiatiecapaciteit van perifere hemopoiëtische voorlopercellen te bestuderen bij patiënten met myeloproliferatieve aandoeningen. Gevonden werd, dat het aantal, de morfologie en de cellulariteit van de verschillende kolonies afwijkend waren. Deze waarnemingen duiden erop, dat bij myeloproliferatieve aandoeningen, afgezien van afwijkingen in de beenmerg-matrix, intrinsieke defecten in de hemopoiëtische voor- lopercellen blijken te bestaan. I 202

ABBREVIATIONS AND DEFINITIONS

A antibody ACD acid citrate dextrose AET 2 amino-ethyl-isothioazonium bromide hydrobromide BFU-e burst-forming unit - erythroid KSA bovine serum albumin B-lymphocyte bursa-dependent lymphocyte CA cell aggregates CFC continuous flow centrifugation CFU-c colony-forming unit — culture CFU-s colony-formint unit - spleen CFU-e colony-forming unit - erythroid CPM counts per minute CSA colony-stimulating activity CSF colony-stimulating factor DLA dog leucocyte antigens DMSO dimethylsulphoxide ds density DS dextran sulphate E erythrocyte EA erythrocyte-antibody complex EA-RFC EA-rosette forming cell EAC erythrocyte-antibody-complement complex EAC-RFC EAC-rosette forming cell EBV Epstein Barr virus E-RFC E-rosette forming cell FCS foetal calf serum FITC fluorescein isothiocyanate GahuFab goat anti-human Fab GVHD graft versus host disease 3H-TdR methyl-3 H-thymidine HSC haemopoietic pluripotent stem cells HBSS Hanks' balanced salt solution HES hydroxyethyl starch HuRBC human red blood cells Ig Imrnunoglobulin IgG-Fc Fc-portion of the IgG-molecule IgM-Fc Fc-portion of the IgM-molecule 203

IU international units kg b.w. kilogram body weight LTT lymphocyte transformation test M molar MLC mixed lymphocyte culture MNC mononuclear cells MRBC mouse red biood cells MW molecular weight N number NSD non significant difference ORBC ox red blood cells PAS periodic acid Schiff PBS phosphate buffered saline PHA phytohaemagglutinin PHZ phenylhydrazine PVSA poly vinylsulphuric acid RBC red blood cells RI refractive index SCC semicontinuous flovv centrifugation SIg surface immunoglobulins SRBC sheep red blood cells TBI total body irradiation T-lymphocyte thymus-dependent lymphocyte TRIC tetramethyl-rhodamineisothiocyanate W/V weight/volume ze zymosan complement complex

EA.-RFC Fc-receptor bearing cells, forming rosUtes with anti- D-IgG-coated HuRBC EA2-RFC Fc-receptor bearing cells, forming rosettes with rabbit anti-sheep IgG-coated SRBC EA3-RFC Fc-receptor bearing cells, forming rosettes with rabbit anti-ox IgG-coated ORBC M-RFC B-cell subpopulation, forming spontaneous rosettes with mouse erythrocytes 204

CFU-c : colony-forming unit — culture The morphologically as yet unidentified precursor of granuloid and/or macrophage colonies in bone marrow or peripheral blood cultures, grown on agar or methylcellulose in the presence of colony-stimulating factor (CSF).

CFU-s : colony-forming unit - spleen The morphologically as yet unidentified precursor of spleen colonies in the mouse. CFU-s give rise to erythroid, granuloid, megakaryocytic or mixed colonies. Because of their pluripotency, CFU-s are considered to represent haemo- poietic pluripotent stem cells (HSC).

HSC : haemopoietic pluripotent stem cell The haemopoietic precursor cell capable of self-production or commit- ment to erythroid, granuloid, or megakaryocytic differentiation. In the mouse, the CFU-s represents the HSC.

Spleen colony assay Assay for CFU-s by enumeration of grossly visible colonies in of lethally irradiated mice, 7 to 10 days after intravenous injection of sus- pensions of bone marrow, spleen or blood cells.

CFU-e : colony-forming unit — erythroid The morphologically as yet unidentified precursor of erythroid colonies in the bone marrow culture in methylcellulose or in plasma clot, grown in the presence of erythropoietin. Colonies consist of 6 to 64 haemoglobin positive cells, which may include reticulocytes. When enumerated, only colonies with at least eight cells are considered. CFU-e is believed to be a cell derived from BFU-e.

BFU-e : burst-forming unit - erythroid The morphologically as yet unidentified precursor of a burst. The BFU-e is believed to be a "committed" cell derived from HSC. 205

Burst An aggregate of three or more erythroid colonies in bone marrow cultures grown on methylcellulose or plasma clot in the presence of high con- centration of erythropoietin (large single colonies containing more than 1,000 erythroid cells are also called bursts).

Do forD31) value One of the numerical values used to characterize the apparently expo- nential decline of biological activity as a function of radiation dose. D3 7 refers to the dose required to reduce the surviving fraction to e"1 or 0.37 on the exponential portion of the curve. f-factor (= seeding efficiency) The measured proportion of infused stem cells seeding into the spleen, as a part of the CFU-s assay. 206

CURRICULUM VITAE

De schrijver van dit proefschrift werd op 20 oktober 1944 te 's-Graven- hage geboren. Zijn middelbare schoolopleiding (HBS-b) genoot hij in Rijswijk van 1958 tot 1963. Vervolgens studeerde hij geneeskunde aan de Rijksuniversiteit te Leiden, waar hij het doctoraalexamen aflegde op 7 juni 1968. Na zijn artsexamen (9 oktober 1970) volgde hij zijn opleiding tot internist op de afdeling Inwendige Geneeskunde van het Militair Hospi- taal "Dr. A. Mathijsen" te Utrecht (hoofd: Dr. C.J. van Belle) van 1971 tot 1972, en de afdeling Inwendige Geneeskunde van het Academisch Ziekenhuis te Leiden (hoofd: Prof. Dr. J. de Graeff) van 1972 tot 1976. Het "vrije jaar" van de opleiding werd doorgebracht op de afdeling Haematologie (hoofd: Dr. B. Speek). Op 1 april 1976 volgde inschrijving in het specialistenregister als internist. Sinds april 1976 is de schrijver in dienst van het J.A. Cohen Instituut voor Radiopathologie en Stralenbescherming ten behoeve van het Isolatie Paviljoen en het Laboratorium voor Experimentele Haematologie.