FINNISH RED CROSS BLOOD SERVICE AND FACULTY OF BIOCIENCES, DEPARTMENT OF BIOLOGICAL AND ENVIRONMENTAL SCIENCES, DIVISION OF GENETICS, UNIVERSITY OF HELSINKI, FINLAND

TRANSCRIPTOME AND GLYCOME PROFILING OF HUMAN HEMATOPOIETIC CD133+ AND CD34+ CELLS

Heidi Anderson

ACADEMIC DISSERTATION

To be publicly discussed, with permission of the Faculty of Science of the University of Helsinki in the Nevanlinna Auditorium of the Finnish Red Cross Blood Service Kivihaantie 7, Helsinki, on February 15th, at 12 noon.

Helsinki 2008 ACADEMIC DISSERTATIONS FROM THE FINNISH RED CROSS BLOOD SERVICE NUMBER 52

SUPERVISORS

Docent Taina Jaatinen, PhD Finnish Red Cross Blood Service Helsinki, Finland

Docent Jukka Partanen PhD Finnish Red Cross Blood Service Helsinki, Finland

REVIEWERS

Docent Heli Skottman, PhD University of Tampere Regea Institute for Regenerative Medicine Tampere, Finland

Docent Timo Tuuri, PhD Infertility Clinic The Family Federation of Finland Helsinki, Finland

OPPONENT

Professor, Director Riitta Lahesmaa, MD, PhD Turku Centre for Biotechnology University of Turku, Turku, Finland

ISBN 978-952-5457-16-2 (print) ISBN 978-952-5457-17-9 (pdf) ISSN 1236-0341 http://ethesis.helsinki.fi Helsinki 2008 Yliopistopaino To my family CONTENTS

PUBLICATIONS ...... 6

ABBREVIATIONS...... 7

ABSTRACT ...... 8

REVIEW OF THE LITERATURE ...... 10 Hematopoietic stem cells ...... 10 Phenotypic characterization ...... 11 Functional characterization ...... 14

Cord blood-derived hematopoietic stem cells ...... 15 Collection and banking ...... 15 Cord blood transplantation ...... 16 Ex vivo expansion ...... 21

Transcriptome profi ling ...... 22 Methods for transcriptome profi ling ...... 22 expression profi ling of murine hematopoietic stem cells ...... 26 profi les of human hematopoietic stem cells ...... 28

Glycosylation ...... 29 N-glycans and N-glycan synthesis ...... 29 Glycosylation in hematopoiesis ...... 33 Effect of glycosylation on homing...... 33

AIMS OF THE STUDY ...... 35

MATERIALS AND METHODS ...... 36 Ethics ...... 36 Cells ...... 36 Methods ...... 37 Gene expression reanalysis ...... 39 Annotation ...... 40 RESULTS AND DISCUSSION ...... 41

CD133+ and CD34+ cells have similar gene expression (studies I and II) ...... 41

Novel molecular markers for HSCs could not be found (study II) .... 42

Differential gene expression between CD133+ and CD34+ cells suggests that CD133+ cells are more primitive (studies I and II) .... 43

CD133+ and CD34+ cells demonstrate similar total colony forming potential (studies I and II) ...... 44

Reanalysis of the original gene expression data focuses on the similarities between CD133+ and CD34+ cells ...... 45

CD133+ and CD34+ cells have divergent roles in hematopoietic regeneration (studies I and II) ...... 49

N-glycosylation-related gene expression is similar in CD133+ and CD34+ cells and correlates with the N-glycome profi le (study III) ... 52

Characteristic mannosylation pattern of CD133+ and CD34+ cells (study III) ...... 52

Differential expression of N-acetylglucosaminyl-transferase and the enrichment of biantennary complex type N-glycans in CD133+ cells (study III) ...... 54

Despite the differential expression of genes coding for β1,4-galacto- syltransferases there are no differences in the β1,4-galactosylation of CD133+ and CD133- cells (study III) ...... 55

CD133+ and CD34+ cell specifi c biosynthesis of N-glycan terminal epitopes (study III) ...... 55

CONCLUSIONS ...... 58

ACKNOWLEDGEMENTS ...... 60

REFERENCES ...... 62 6 PUBLICATIONS

PUBLICATIONS

This thesis is based on the following original publications, which have been reproduced with the permission of the copyright holders.

I. Jaatinen T, Hemmoranta H, Hautaniemi S, Niemi J, Nicorici D, Laine J, Yli-Harja O, Partanen J. Global gene expression profi le of human cord blood-derived CD133+ cells. Stem Cells 2006;24:631-641.

II. Hemmoranta H, Hautaniemi S, Niemi J, Nicorici D, Laine J, Yli- Harja O, Partanen J, Jaatinen T. Transcriptional profi ling refl ects shared and unique characters for CD34+ and CD133+ cells. Stem Cells Dev 2006;15:839-851.

III. Hemmoranta H, Satomaa T, Blomqvist M, Heiskanen A, Aitio O, Saarinen J, Natunen J, Partanen J, Laine J, Jaatinen T. N-glycan structures and associated gene expression refl ect the characteristic N-glycosylation pattern of human hematopoietic stem and progenitor cells. Exp Hematol 2007;35:1279-1292.

In addition, some unpublished data are presented. ABBREVIATIONS 7

ABBREVIATIONS

BFU burst-forming erythoid BM bone marrow CB cord blood CD cluster of differentiation cDNA complementary deoxyribonucleic acid CFU colony-forming unit DNA deoxyribonucleic acid E erythroid GEMM granulocyte-erythroid- macrophage-megakaryocyte GM granulocyte-macrophage HSC hematopoietic stem cell Lin lineage differentiation N-glycan asparagine-linked glycan NMR nuclear magnetic resonance mHSC murine hematopoietic stem cell mRNA messenger ribonucleic acid PB peripheral blood RNA ribonucleic acid SAGE serial analysis of gene expression qRT-PCR quantitative real-time reverse transcriptase- polymerase chain reaction 8 ABSTRACT

ABSTRACT

Human blood contains a large variety of differentiated cells with a limited half-life. New blood cells are continuously provided by self-renewing multipotent hematopoietic stem cells (HSC). The capacity of HSCs to regenerate the hematopoietic system is utilized in the treatment of patients with hematological malignancies. HSCs can be obtained from bone marrow (BM) or peripheral blood (PB) after cytokine-induced HSC mobilization. In addition, blood from both placenta and umbilical cord is also rich in HSCs. Cord blood (CB) can be collected after delivery, cryopreserved for the possible future need of an HSC graft for patients with no suitable BM donor available.

HSCs can be enriched using an antibody-based recognition of CD34 on the cell surface. Another glycoprotein, CD133, has also been used for identifi cation of primitive cells. The CD133+ cell population has been shown to be more homogeneous and contain a higher number of potential HSCs than CD34+ cells. CD133 and CD34 molecules are often coexpressed, yet the CD133+ and CD34+ cells may have partly different roles in hematopoiesis.

Each cell has a glycome typical for that cell type. The glycome of HSCs is poorly known. Glycans at the outermost surface of the cell transmit various signals between the matrix environment and the surrounding cells. A CD34+ cell-specifi c glycoform of CD44, cell adhesion molecule, has been shown to have a possible function in HSC homing.

The aim of present studies has been to characterize the global gene expression of CB CD133+ and CD34+ cells. Furthermore, the identically constituted gene expression profi les of CD133+ and CD34+ cells were compared to unravel shared and unique gene expression in CD133+ and CD34+ cells. A prioritization algorithm was used to rank the genes best separating CD133+ and CD34+ cells from their counterparts, CD133- and CD34- cells. Thus, these genes were suggested to play a part in the regulation of primitive hematopoietic cells. CD133+ and CD34+ cells solely expressed several genes encoding transmembrane proteins that could putatively serve to identify HSCs. Moreover, cell type-specifi c gene expression was demonstrated in both CD133+ and CD34+ cells. These differentially expressed genes were associated with divergent biological processes suggesting differences in the transcriptional regulation of ABSTRACT 9

CD133+ and CD34+ cells. Thus, they may represent differences in the self-renewal, differentiation or homing properties. Moreover, CD133+ cells expressed a higher number of genes associated with HSC maintenance and pluripotency indicating the more primitive nature of CD133+ cells. Furthermore, the gene expression data bank of CD133+ and CD34+ cells may be used to study the pathogenesis of hematological disorders and the development of malignancies.

Another aim of these studies was to identify the characteristic glycosylation pattern of primitive hematopoietic cells and the genes producing the key glycan entities. High mannose type N-glycans were the most prevalent glycan type in CD133+ cells. CD133+ cells had more biantennary complex type N-glycans than CD133- cells, whereas multiantennary complex type N-glycans were fewer in CD133+ cells than in CD133- cells. Moreover, the CD133+ cell N-glycans showed enrichment of terminal α2,3-sialylation. Combined gene expression and glycan profi ling allowed the identifi cation of the key genes regulating the N- glycosylation characteristic for CD133+ cells. The critical genes were similarly expressed in CD34+ cells, suggesting a similar glycosylation pattern in CD133+ and CD34+ cells. Knowledge of HSC glycobiology can be used to design therapeutic cells with improved cell proliferation or homing properties. Improved HSC homing would diminish the number of HSCs needed for the therapy of patients, and allow for greater use of CB grafts. 10 REVIEW OF THE LITERATURE

REVIEW OF THE LITERATURE

Hematopoietic stem cells

HSCs are small cell populations in adult BM. They have enormous capacity to self-renew and to generate progenitor cells that can differentiate into mature blood cells1. The life span of most white blood cells is from a few hours to a couple of days. The most common cell type in blood, red blood cells, has an average life expectancy of 120 days. Adult humans have tens of billions of red blood cells. Thus, an average of two million new red blood cells are required every second. HSCs produce new cells to balance the loss of cells each day throughout life.

The mammalian hematopoietic tissue derives from the mesoderm layer of the embryo. The HSCs fi rst germinate in the dorsal aorta2. They are also produced in the major embryonic vessels, umbilical and vitelline arteries2. From the aorta, HSCs migrate to yolk sac, and thereafter to liver and BM. In addition, the placenta supports a large stem cell pool during development and is suggested to serve as source of pre-HSCs/ HSCs3. The number of HSCs in the placenta and cord veins drops during the trimester of the gestation. Yet, blood from the umbilical cord and placenta of full-term infant provides a rich source of HSCs.

BM is the primary location for HSCs in the adult. In addition, small amounts of HSCs are in both circulation and in the spleen. Most of the HSCs reside in their BM niche in a quiescent, noncycling state. However, to sustain hematopoiesis and to maintain the HSC pool, cell divisions are required. HSCs have been thought to divide both symmetrically and asymmetrically4,5. In symmetric cell division, two identical HSCs are formed. Asymmetric cell division leads to the formation of two different daughter cells, one that is similar to the mother cell and contains long- term repopulation capacity, and a daughter cell that has short-term repopulation capacity. Long-term HSCs have life-long hematopoietic reconstruction capacity. Short-term HSCs have reduced capacity to sustain hematopoiesis, but they give rise to lineage committed progenitors. According to estimates, only 1/10 000 cells in BM has long- term repopulation capacity6. Once HSC has been committed to lineage differentiation, it no longer has HSC capacity and properties.

The tremendous progeny of HSCs is able to repopulate the marrow. HSC transplantation for marrow repopulation after high doses of chemotherapy REVIEW OF THE LITERATURE 11 or total body irradiation is an established procedure for the treatment of leukemia. HSC transplantations have also been used in therapy for other hematological disorders, immunodefi ciency syndromes, inborn errors of metabolism and autoimmune diseases7-9. It has been suggested that HSCs have the capacity to differentiate into cells of nonhematopoietic tissue as well10-12. Current studies are targeted to fi nd out if HSCs can be utilized in the treatment of such diseases as diabetes, Parkinson’s disease and spinal cord injury.

Phenotypic characterization

HSCs are morphologically similar to leukocytes. They are found among small, rounded cells that have little cytoplasm13. HSCs are mainly noncycling. They stain poorly with vital dyes such as rhodamine123, which locates the mitochondria of living cell, or Hoechst stains 33342/33258, which recognizes DNA of live or fi xed cells. Poorly stainable noncycling cells are called Rho/Hoechst 33342low, in contrast to Rho/Hoechst 33342high which indicate the actively cycling cell population. Thus, Rholow cells contain a higher number of HSCs than Rhohigh cells14. Moreover, HSCs have high aldehyde dehydrogenase activity that can be used to enrich primitive cell populations15.

Leukocytes carry various cluster of differentiation (CD) molecules on their cell surface. These have been used as markers to characterize cell type, maturity and activity of the cell. There is no single marker to identify HSCs, but the presence of the CD34 molecule marks a highly enriched HSC population. As a result, CD34-based isolation of HSCs is most widely used. CD34 antigen is also carried by hematopoietic progenitor cells and cells in the vascular endothelium. Determination of the cell type depends on which other CD molecules are present with CD34. HSCs are found among a population of CD34+ cells that do not express CD38 or other lineage differentiation (Lin) markers such as CD2, CD3, CD14, CD16, CD19 CD24, CD56, CD66b and Glycophorin A (Figure 1). 12 REVIEW OF THE LITERATURE

Figure 1. Schematic representation of the hematopoietic lineage differentiation markers during hematopoiesis. Modifi ed after Kyoto Encyclopedia of Genes and Genomes Pathway Database16. REVIEW OF THE LITERATURE 13

However, other markers such as CD90 (Thy-1) and CD117 (c-Kit) are carried on HSCs. The expression of CD135 (FLT3) is typical for multi- potent progenitors17.

Hematopoiesis has been studied mainly in the mouse model. Murine HSCs (mHSC) have phenotypic characteristics divergent of human HSCs. They are enriched by selecting Lin- cells that carry Sca-1 and c-Kit molecules on their cell surface18. Unlike in humans, mHSCs are CD34 negative18,19. CD90 is expressed only at low level in mHSCs20. The signaling lymphocyte activation molecule family receptors, the presence of CD150 molecule and the lack of CD48 molecule, can also be used to highly purify mHSCs21.

A small population of CD34-CD38- cells carrying the CD133 molecule on their surface seems to be able to give rise to CD34+ cells in humans22. However, in human HSCs, long-term marrow repopulating capacity is obtained only after the expression of CD3423. On average, 80% of the CD34+ cells carry CD133 on their surface. CD133+ cells are almost all CD34 positive as well24. Adult stem cells of non-hematopoietic origin have been found to express CD13325-27. Its expression is also typical in the embryoid bodies derived from embryonic stem cells28. The CD133 molecule is present in circulating cells with endothelial differentiation capacity, suggesting that the differentiation capacity of CD133+ cells might be greater than in CD34+ cells25.

HSC enrichment based on antibody-recognition of cell surface molecules is not an ideal method. Antibody binding on HSCs decreases HSC en- graftment potential, which is an outcome that should be avoided29. HSC markers have been present on functionally limited cells30. HSCs can have altered cell surface marker expression in in vitro conditions31. Fur- thermore, both functionally competent HSCs and cells with less capacity may have the same phenotype. Many ongoing investigations are aimed toward HSC enrichment by methods that take use of such methods as density gradient difference between cell types or the divergent selectin binding affi nity of HSCs32-35. These methods may exceed up to a three- fold enrichment of HSCs when compared to a mononuclear cell popula- tion. However, no methods have been more effi cient in HSC identifi ca- tion than antibody-based strategies. In the immunomagnetic selection of CD34+ or CD133+ cells purities of over 90% can be obtained36. Evalu- ation of HSC potential requires the study of their functional properties. 14 REVIEW OF THE LITERATURE

Functional characterization

Functional assays are useful in the measurement of the potential of the cell. However, they are complicated to use for cell selection purposes. Better understanding of HSC biology may help to develop novel methods for fast read-out of HSC potential.

The best model used to measure long-term marrow repopulating capacity is provided by severe combined immunodefi ciency mice repopulating assay. Of the severe combined immunodefi ciency mice transplanted human CD34+CD38- cells, one out of 617 cells is able to engraft37. The relatively low capacity of the CD34+CD38- cell population to repopulate mouse marrow is affected by the xenoenvironment. Engraftment capacity has increased with direct intra-BM transplantation, where one out of 44 CD34+CD38- cells has been able to populate the marrow38,39. Human CD34+ cells negative for CD133 have not been able to provide engraftment in mice model40. Moreover, the number of vacant HSC niches in the recipient’s BM regulate the engraftment of transplanted HSCs41.

In vitro assays, such as long-term culture initiating cells or cobble stone area forming cells, have also been used to estimate the number of long-term HSCs. In these methods, stromal cells are used to support HSC development and their colony forming capacity is tested after the long-term culture. However, differential kinetics between cells in in vivo and in vitro assays indicate that long-term culture conditions may not necessarily be able to represent the in vivo long-term repopulation capacity of HSCs40.

Colony forming unit assay (CFU) exploits a cell’s ability to produce progeny in vitro. The cells possessing multipotent progeny, the highest differentiation potential, form granulocyte erythrocyte macrophage megakaryocyte colonies. CD133+CD34+ cells’ progeny are mainly granulocyte macrophage and granulocyte erythrocyte macrophage megakaryocyte colonies40. Thus, CD133+ cells are suggested to be more naive than CD133-CD34+ cells, whose progeny also produce some unipotent burst-forming erythrocyte and erythrocyte colonies40. REVIEW OF THE LITERATURE 15

Cord blood-derived hematopoietic stem cells

CB CD34+ cells have greater proliferation potential and they produce signifi cantly more progeny than BM CD34+ cells10,42,43. Thus, the greater number of more immature HSCs may be found in CB than in BM ori- gin44. Moreover, CB HSCs have been shown to be able to differentiate into endothelial and cardiomyocyte-like cells45. Unlike BM CD34+ cells, the CB CD34+ cell population contains very few B cell lineage commit- ted CD34+CD19+ progenitors (Figure 1). In addition, late-erythroid CD34+CD71highCD64-, late-myeloid CD34+CD33+CD15+ and granulo- monocytic CD34+CD71+CD64+ progenitors are rarer in CB than BM46. On the other hand, early-myeloid CD34+CD33+CD15- and T cell lineage committed CD34+CD7+CD3- cells are more abundant in CB than in BM.

In cell culture, CB-derived CD34+ cells signifi cantly increase their prolif- eration when exposed to external cytokines and growth factors and are easier to culture in vitro than BM CD34+ cells47,48. The environment of CB HSCs is different from that of BM HSCs. BM HSCs are attached to the osteoblastic cells via the engagement of very late antigen 4 and vascular cell adhesion molecule 149,50. The osteoblastic cells secrete growth fac- tors and cytokines such as interleukin-6, stem cell factor and chemokine (C-X-C motif) ligand 12, and provide ligands such as fi bronectin for the adhesion of HSC51. CB HSCs are, in contrast, devoid of osteoblastic cells. While the placenta is a supportive niche for HSCs in the fetal environ- ment during midgestation3, the placenta and CB of a full-term infant is likely to contain low amounts of growth factors and cytokines. Moreover, CB HSCs may be naturally prepared for oxidative stress and survive bet- ter in the 20% oxygen of most cell culture. The oxygen concentration in the CB environment is higher than in BM endosteum, the niche for BM HSCs51. The greater success in culturing CB HSCs may also be aug- mented by the fact that CB HSCs have markedly longer telomeres than in BM or PB HSCs’52. This suggests that CB HSCs are able to go through more cell duplications than BM HSCs.

Collection and banking

After the fi rst successful CB transplantation in 198853, CB has been stored and cryopreserved for potential future use. CB banks have been created to increase the availability of CB units. Today, there are tens of public CB banks worldwide storing more than 200,000 CB units54,55. In addition, an unknown number of CB units have been stored privately. 16 REVIEW OF THE LITERATURE

The CB units stored in public banks are available to practically everyone. The International NETCORD Foundation provides information regarding the CB banks jointly in Europe, the United States, Japan and Australia. An online virtual offi ce for real-time searching allows an even faster sur- vey of available CB grafts and makes their use more effi cient. This espe- cially enhances the search of grafts for patients with a minority genetic background that have only a few suitable stem cell donors available55. The accredited CB banks of the Foundation for the Accreditation of Cel- lular Therapy, such as the Finnish Red Cross Blood Service Cord Blood Bank, ensure that minimum requirements for CB quality, handling and storage are met56. As a result, accreditation criteria maintain the quality of the banked CB.

CB is a byproduct of delivery, it is easy to obtain and ex utero collection carries no risk to the donor57. CB collection is similar to PB collection, i.e. the cord vein is punctured with a needle and the collection bag is fi lled by gravity. Suitable CB donors have no known genetic or transmitted disease, have been informed of the situation and have provided written consent for the utilization of the donated CB. Further, CB is tested for human immunodefi ciency virus, hepatitis C virus, hepatitis B virus, human T cell leukemia virus and syphilis. In addition, donor mothers have been analyzed for the presence of parvo and cytomegalovirus.

Yet it is not known whether CB cells, cryopreserved for long periods of time, are successful in transplantation. The results on mouse marrow populating assay and CFU suggest a rather long half-life for stored CB units. Thawed CB HSCs have been able to repopulate the marrow in a mouse model after 15 years of storage in liquid nitrogen58. The colony forming potential of HSCs has been shown to be 97% of that of fresh HSCs after 12 years of storage in liquid nitrogen59.

Cord blood transplantation

Because fast treatment of the patient may be crucial, CB is a noteworthy alternative to transplantation when no suitable related BM donor is readily available. CB as readily available and cryopreserved in CB banks is faster and easier to obtain than unrelated BM donor graft. To obtain BM graft, the donor candidates in the BM donor registry have to fi rst be located. Their HLA-typing results are confi rmed, they are informed of the collection process and their consent is requested. Moreover, donors need to pass a complete physical examination and and need to take REVIEW OF THE LITERATURE 17 additional blood tests to become a suitable BM donor. After all this, the date for transplantation can be scheduled. An average of 13.5 or 49 days are required to obtain CB or BM graft, respectively60.

CB transplantation has already been performed on approximately 4000 patients. CB has been used to treat a variety of diseases7-9,55,61. A full list of diseases treated by CB transplantation is shown in Table 1. Novel ap- plications aim to improve the engraftment of CB grafts. Methods such as HSC expansion, supplement of CD34+ cells from PB, mesenchymal stem cells and platelet microparticles are under investigation62-67. Gene trans- fer into CB HSCs has been used to improve HSC capacity and their use in non-hematopoietic injured tissues. Initial transduced CD34+ cells have been successfully used in the treatment of children with severe com- bined immune defi ciency caused by adenosine deaminase-defi ciency68. These patients have been given autologous CB CD34+ cells transduced with an adenosine deaminase gene.

Human leukocyte antigen-matched adult donors are not available for all the patients. A patient receiving an unrelated CB graft has an equal survival prognosis than patients getting a BM graft from an unrelated donor69. However, patients receiving a human leukocyte antigen-mis- matched CB graft from an unrelated donor show less severe graft versus host disease than patients treated with an human leukocyte antigen- mismatched BM or PB graft70-75. It has been suggested that greater tolerance of human leukocyte antigen-disparity in CB transplantation is due to the immature lymphocytes of newborn76. Also, CB dendritic cells are more immature and have diminished functionality when compared to PB73. Dendritic cells present antigens to T and B cells and are respon- sible for initiating and shaping the adaptive immune system.

The main inconvenience of CB transplantation is the limited number of cells available in a single CB unit. In CB transplantation, 2x107 cells per kilogram is typically considered adequate for transplantation77. The number of cells per kilogram is 10 times more in BM transplantation. Lower cell dose causes less graft versus host disease, whereas higher cell dose has been associated with faster myeloid and platelet engraft- ment71.

CB grafts have been mostly used in the treatment of pediatric patients. However, reduced intensity transplantation or double CB grafts have been incorporated to overcome the shortage of cells for the therapy of adult patients78-80. Smaller cell doses are suitable for reduced intensity 18 REVIEW OF THE LITERATURE transplantation which may be applied when patients are not strong enough to go through standard pre-transplant treatment to destroy disease cells81. In the reduced intensity transplantation, the donor immune cells are used to fi ght the disease. Moreover, the double CB grafts, when compared to the single CB grafts, have shown better engraftment and improved graft versus leukemia response in patients82,83.

Despite the lower risk of graft versus host disease with a CB graft, a similar graft versus leukemia response has been seen after CB and BM transplantations. Immune reconstruction, recovery of early T cells and B cells after CB transplantation, is similar to that of BM85,86. However, some clinics have observed an increase in treatment-related mortality with CB grafts. This has been suggested to be due to slower engraftment of the graft, but could also be due to greater human leukocyte antigen- disparity or lower cell dose of CB grafts used for the treatment. Slower engraftment increases the risk of bacterial infections, which seem to be slightly more common after CB transplantation87. However, infection- related mortality is not increased after CB transplantation87. REVIEW OF THE LITERATURE 19

Table 1. List of diseases and conditions treated with CB transplantation. Adapted from Cord Blood Center84. 20 REVIEW OF THE LITERATURE REVIEW OF THE LITERATURE 21

Ex vivo expansion

The ex vivo expansion systems have succeeded in augmenting HSC proliferation and increasing the number of progenitor cells with the abil- ity to generate T lymphocytes and Natural Killer cells88. The challenge remaining is the expansion of long-term HSCs. CD34+CD38- cells have been expanded in culture, but the marrow repopulating capacity has not been increased88. Moreover, expanded and cycling cells have dem- onstrated increased susceptibility to apoptosis when compared to quies- cent HSCs89. This could cause the early death of transplanted HSCs and impaired initial engraftment.

In the ex vivo expansion, the CB-derived cells are cultured in a media supplemented with various growth factors and cytokines such as interleukin-1β, -3, -6, -11, stem cell factor, fms-tyrosine kinase 3 ligand, trombopoietin, granulocyte-macrophage colony stimulating factor, erythropoietin and chemokine ligand 3. Alternatively, HSC growth has been supported by co-culture with stromal cells or mesenchymal stem cells. A variation of this method uses conditioned media containing secreted factors from stroma or mesenchymal stem cells.

The engraftment potential of HSCs is limited mostly to cells in the 90 91 G0 state . HSCs induced to proliferate do not reenter the G0 state . These are the challenges that need to be overcome in order to expand HSCs. Fortunately, the quiescent state of the cell is not as critical to the engraftment potential of CB HSCs as it is to PB HSCs91.

A better understanding of molecular mechanisms controlling self- renewal would enhance the ability to expand CB cells. The protein level of transcription factor Homeobox B4 (HOXB4) has been shown to positively correlate with better in vitro proliferation92. HOXB4 transduced HSCs have been shown to maintain mHSC capacity93. Unfortunately, the same outcome has not been achieved in humans93. The potential risk with transduction-based method and cell culture is that they change HSC gene expression and the expression of cell surface molecules94. This may have an effect on HSC properties such as homing and therefore on the engraftment of the transplanted cells. 22 REVIEW OF THE LITERATURE

Transcriptome profi ling

In recent years, transcriptome profi ling has been widely used to understand the genetic regulation of a particular cell type. Transcriptome encompass the global gene expression in a cell at a specifi c time. It can proffer valuable information on the signifi cant biological processes behind the maintenance of the functionality of the cell. Transcriptome profi ling is used to reveal the key genes involved in HSC self-renewal versus differentiation decisions.

The technology for the study of transcriptome is not dependent on any prior knowledge of the genes expressed in the cells of the study. However, challenges remain in regards to the administration and in- terpretation of the enormous data provided by transcriptome profi ling. Transcriptomics provides fundaments for more defi nitively designed studies and guidance to select the genes for functional studies. It should be kept in mind that the small change in the expression level of a gene may relate to a great difference in the total amount of the corresponding protein present in the cell. The presence of a molecule in a cell depends on the translation features of the mRNA sequence and the amount of antisense sequences degrading the corresponding sense sequences95,96. Results from the transcriptome and proteome analysis of a similar cell type can not usually be fully matched partly due to the heterogeneity or instability of the proteins97. Technical limitations may also explain some of the divergences found in trancriptome and proteome level98.

Methods for transcriptome profi ling

Gene expression profi ling of 45 Arabidopsis thaliana genes was reported in 1995 as the fi rst microarray-based experiment99. Today there are many different kinds of microarray chips used, some of which are the laboratories own cDNA arrays and others which are commercially pro- duced microarrays. The basic idea behind these is the same (Figure 2). Preanalysis and normalization of the data prior to the analysis of the results is one noticeable part of the data interpretation.

Microarray analysis is still the most widely employed technology for transcriptome profi ling, but serial analysis of gene expression (SAGE) (Figure 3) and subtractive hybridization (Figure 4) have also been com- monly used. Microarray is designed for the study of differential gene REVIEW OF THE LITERATURE 23

Figure 2. Schematic representation of microarray technology. Total RNA from the cell is isolated and converted into cDNA. cDNA is labeled and hybridized to an array containing oligonucleotide probes. After hybridization, the level of fl uorescence of the attached sequences on the chip is measured with a laser scanner, and microarray analysis methods are used to calculate the abundance of each represented RNA. Adapted from Affymetrix GeneChip Expression Analysis Technical Manual100.

expression. The subtractive hybridization reveals solely the differen- tially expressed genes. On the other hand, SAGE technology is effi cient in detection of the copy number of the transcripts expressed. 24 REVIEW OF THE LITERATURE

Figure 3. The overview of SAGE technology. RNA from the cells is converted into cDNA molecules in the presence of oligo(dT) magnetic beads. Immobilized cDNA is cleaved with an anchoring (usually NlaIII) and tagged (usually with BsmFI) to form SAGE tags. Tags are combined to di-tags, amplifi ed and linked to concatamers of several SAGE tags. Concatamers are cloned in bacteria and then sequenced to reveal the identity and abundance of unique sequences. Modifi ed after (Broxmeyer HE 2004) with data from Serial Analysis of Gene Expression101,102.

Several other methodologies such as differential display analysis, representational difference analysis, massive parallel signature sequencing and analysis of expressed sequence tags can also be utilized. The advantages and disadvantages of the three most commonly used REVIEW OF THE LITERATURE 25

Figure 4. The general concept of subtractive hybridization. RNA or cDNA from the cells of interest is hybridized to single-stranded cDNA-containing paramagnetic beads from control cells. Hybrids and excess driver cDNA are removed using a magnet. The remaining fragments present the RNA expressed specifi cally in the cells of interest. Modifi ed from (Lönneborg et al. 1995)103.

technologies; microarray analysis, SAGE and subtractive hybridization are summarized in Table 2. The high costs of using several high throughput methods often limit parallel use of these technologies by a single laboratory. Instead, quantitative real-time reverse transcriptase- 26 REVIEW OF THE LITERATURE polymerase chain reaction (qRT-PCR) is often used to verify global gene expression profi ling results. Usually this means that expressions of a selected set of genes are reanalyzed.

Table 2. Comparison of three different methods for gene expression analysis.

It has been shown that results from qRT-PCR, microarray and SAGE are usually correlated, especially when the gene is expressed on the high or medium level104. As a sequence-based technology, the sensitivity of qRT- PCR and SAGE analysis is higher than the sensitivity of hybridization- based microarray analysis. Both qRT-PCR and SAGE are able to detect gene expression at a very low level. The level of differential gene expression is often detected lower in microarray analysis than in qRT- PCR105.

Gene expression profi ling of murine hematopoietic stem cells

Previously, particular with the earliest studies, the focus was on discovering transcriptional regulation of mHSCs. Despite the differences in the genes related to hematopoiesis in mice and humans, murine gene expression profi ling has proven to be valuable in understanding HSC biology. Genetic programming in stem cells has been underlined in the comparison of the transcriptional expression in HSCs, progenitors and mature hematopoietic cells. REVIEW OF THE LITERATURE 27

The fi rst large scale analysis of HSCs identifi ed over 2000 genes differentially expressed in fetal mHSCs (AA4.1+Sca+c-Kit+Lin-/low) and mature leukocytes (AA4.1-)106. The primitive cells showed enriched expression of genes associated with cell signalling, RNA synthesis, metabolism, protein synthesis, cell division, cell structure, and cell defence. The majority (53%) of the expressed transcripts were not previously known. Gene expression associated with cell signalling has been particularly prevalent in long-term HSC populations, whereas an increase in cell cycle initiation, DNA repair and protein synthesis has been shown to be characteristic of short-term HSCs. Genes typical to lineage committed progenitors are expressed in low levels in short-term mHSCs107.

In the study of gene expression in HSCs and various lineage committed progenitors, 43% of the differentially expressed genes were shown to be specifi c to the HSC population108. Gene expression profi les of fetal and adult mHSCs have been shown to have 70% overlap109. Fetal HSCs express a few embryonic stem cell-related genes such as Dnmt3b and microH2A1.2 that have not been found to be prevalent in adult BM HSCs. It has also been shown that fetal HSCs are in an active state of cell cycle more often than adult HSCs110,111.

HSCs and neural stem cells have shown common gene expression not present in mature hematopoietic and neuronal cells108. However, highly HSC enriched cells have shown more similar gene expression patterns with mature hematopoietic cells than with stem cells from neural or embryonic origin112. Different stem cell types possess more similarities in the representative biological processes than on the level of individual genes113. Genes associated with chromatin remodeling have been upregulated in various stem cell populations. It has been suggested that this process regulate the self-renewal and differentiation potential of stem cells114. Of the genes common to all stem cell populations, only 4 transcripts uridine phosphorylase, suppressor of Lec15 and two expressed sequence tags have been identifi ed. This suggests that only a few genes have restricted expression in stem cells112. 28 REVIEW OF THE LITERATURE

Gene expression profi les of human hematopoietic stem cells

The differential gene expression between BM, PB and CB-derived CD34+ cells may be related to the possible differing roles of CD34+ cells in different environments10,109,115. The HSC mobilization actuate the gene expression resulting in partly divergent gene expression in BM or granulocyte colony-stimulating factor mobilized CD34+ cells from humans116. Cell cycle promoting genes are more common in BM CD34+ cells than mobilized PB CD34+ cells. The number of genes encoding transcription factors or gene products associated with apoptosis is abundant in mobilized cells when compared to BM CD34+ cells116. The gene expression in CB and BM CD34+ cells is more similar than gene expression in mobilized PB CD34+ cells10,115. BM CD34+ cells have shown enriched expression of genes associated with support of myeloid lineage development and inhibition of apoptosis. Cell adhesion and mobilization- associated genes such as CXCR4, which is important in the homing and engraftment of CD34+ cells, are underexpressed in CB CD34+ cells when compared to BM CD34+ cells.

Typically, a high amount of expressed genes is common to HSCs. This may be due to their relatively open or transcription permissive chromatin structure117,118. CB CD34+ cells have the highest number of overexpressed genes when CB, BM and PB CD34+ cells are compared10,115. The number of overexpressed genes is higher in BM CD34+ cells than in PB CD34+ cells. The greater number of active genes suggests broader transcriptional options for HSCs. Many genes expressed by HSCs have been located in the pericentric heterochromatin, unlike the genes related to activation in mature lymphocytes located in centromeric heterochromatin118. This suggests at least a partly different pathway for the activation of genes in HSC and mature lymphocytes. Its possible role is to provide fast recruiting cells to generate the type of blood cells needed.

Low levels of embryonic stem cell marker gene Oct4 have been de- tected in CB CD133+ cells119. Other pluripotent markers such as Sox1, Sox2, FGF4 of Rex1 are also expressed in CB CD133+ cells119. These fi ndings suggest that CD133+ cells, especially from CB, may have the potential to differentiate into nonhematopoietic tissue types. Moreover, the slowly dividing CB CD34+CD38- cells have been shown to express several genes differently when compared to fast dividing CD34+CD38- cells120. These slowly dividing, more primitive, hematopoietic cells were shown to express CD133 abundantly. REVIEW OF THE LITERATURE 29

Glycosylation

Glycosylation is specifi c to each cell type. It involves common posttrans- lational modifi cations that take place on the nascent protein. There are two kinds of glycosylation; N-glycosylation and O-glycosylation. Nearly all proteins traveling through endoplasmic reticulum become N-glycosylated. Glycosylated proteins mainly function in the extracellular compartments and in the secretory pathway. Due to their location on the cell surface, they serve a role as providers of information transfer in cell recognition and in interactions with other cells or in the extracellular matrix.

Revealing glycosylation features characteristic of HSCs broadens the knowledge of HSC biology. Two molecules may have identical amino acid composition, but differ due to differential glycosylation. Diverse glyco- forms of Thy-1, a stem cell marker, have been found in the thymus and brain121. Moreover, individual glycosylation leads to the formation of blood groups such as ABO(H), Secretor, P and Lewis on erythrocytes122. Changes in glycosylation are involved in cell differentiation, embryogenesis and tissue morphogenesis.

N-glycans and N-glycan synthesis

N-glycans support protein folding, stability and oligomerization, and par- ticipate in traffi cking, recognition of receptor binding sites and protein localization. N-glycans are vital for organisms from yeast to human, and most of the known glycosylation disorders are caused by defective N- glycosylation123,124. The defects have been most dramatic in cells going through rapid alterations, such as leukocytes. For example, the inhibition of N-glycan backbone trimming mannosidases cause a blockage in B cell differentiation125 (Figure 5). In addition to N-glycan, other glycan types including O-glycans, glycosphingolipids, glycosaminoglycans and glyco- phospholipid anchors are common126. Due to the technical challenges of studying the structure of these other gycan types, they are less well known.

In the biosynthetic pathway of N-glycosylation, the preformed glycan is transferred to the amino acid asparagine (N) glycosylation site on the nascent protein in endoplasmic reticulum (Figure 6). N-glycans can be grouped into three classes; high mannose, hybrid and complex type structures, depending on the amount and type of glycan modifi cations they have gone through127 (Figure 6). 30 REVIEW OF THE LITERATURE

Figure 5. Schematic representation of N-glycans. N-glycans consist of distinct regions of N-glycan core, backbone and terminal epitopes that are synthesized by different and glycosidase families. Monosaccharide symbols: = N-acetyl-D- glucosamine; = D-mannose; = D-galactose; = L-fucose; = N-acetylneuraminic acid.

High mannose N-glycans represent the unprocessed glycan structures with a maximum of 9 D-mannoses attached to the N-glycan core. The hybrid type or intermediately processed glycans possess characteristics of both high mannose and complex type N-glycans. Complex type glycans are so-called fully processed N-glycans and occur as bi-, tri-, tetra- or pentaantennary structures.

The α-mannosidase 1 encoded by MAN1 genes and β-N-acetyl glucosaminyltransferase 1 encoded by MGAT1 promotes the conversion of high mannose N-glycans into more processed glycan types (Figure 6). MAN1 gene products remove the terminal α-mannose residues, while β- N-acetylglucosaminyltransferase 1 transfer an N-acetyl-D-glucosamine residue to form the fi rst branch towards processing more developed N-glycans. The synthesis of hybrid and complex type N-glycans is developmentally essential128.

MAN2 genes encode α-mannosidase 2 enzymes, which trim the hybrid type N-glycans prior to the action of MGAT2 coded β-N-acetylglucosami- nyltransferase 2. Further, β-N-acetylglucosaminyltransferase 2 forms the complex type N-glycans. Complex type N-glycans have also been found in MAN2-defi cient mice despite the absence of family 2 α-man- nosidases129. This suggests that there are other enzymes, possibly other α-mannosidases, involved in the demannosylation of hybrid type N-gly- cans. Accumulation of hybrid type N-glycans and the lack of complex N- REVIEW OF THE LITERATURE 31

Figure 6. N-glycan synthesis. The attachment of N-glycan precursor to the gly- cosylation site asparagine (N) of a nascent protein initiates the synthesis. Further, the three and one mannose residues are enzymatically removed from the N-glycan. The processing of N-glycan into the hybrid type starts by the removal of the mannose monosaccharides by family 1 mannosidases and adding one N-acetyl- D-glucosamine by β-N-acetylglucosaminyltransferase 1. The rest of the removable mannoses are docked by mannosidase 2 family members. Different glycosyltrans- ferases, modifying the glycan backbone and terminal epitopes, form the vast variety of N-glycans by differentially adding N-acetyl-D-glucosamine, D-galactose, L-fucose and N-acetylneuraminic acid residues. Monosaccharide symbols: = glucose; = N-acetyl-D-glucosamine; = D-mannose; = D-galactose; = L-fucose; = N-acetylneuraminic acid. Glycosidic linkages are indicated by lines connecting the monosaccharides.

glycans in organisms may affect cell signaling. A mutation in the human ortholog of the MGAT2 gene has been associated with a rare human disease known as congenital disorders of glycosylation type IIa130. This disease affects disfunction of multiple physiological systems.

The tri-mannosyl core on N-glycans of both hybrid and complex types can be modifi ed by MGAT3 encoded β-N-acetylglucosaminyltransferase 3, which adds the so called bisecting N-acetylglucosamine. Two other branching moieties, the tri- and tetra-antennary structures, are gener- 32 REVIEW OF THE LITERATURE ated by MGAT4a, MGAT4b and MGAT5 encoded β-N-acetylglucosami- nyltransferases. Insuffi cient expression of MGAT4a has been shown to cause failure in pancreatic beta cells leading to type 2 diabetes131. In some cases, β-N-acetylglucosaminyltransferase 5 forms another branch in the α1,3-linked core mannose. MGAT5 does not seem to be essential for survival132. On the other hand, enhanced MGAT5 expression has been demonstrated to support tumor growth and metastasis133.

The variability of N-glycans is increased with terminal epitopes modifi ed by β1,4-, β1,3-galactosyltransferases, α2,3-, α2,6-sialyltransferases and α1,2-, α3/4-, α1,6-fucosyltransferases (Figure 5). Mutations in the terminal epitopes are not usually crucial for survival, but they often affect cells in the hematopoietic system. Mice with mutations in the B4GALT1 gene have reduced neutrophil traffi cking in the infl amed tissues134. In humans, the mutation in B4GALT1 cause congenital disorder of glycosylation type IId disease with severe neuronal disease and blood clotting defects135,136. The role of the 4 other β1,4-galactosyltransferases in N-glycans synthesis is poorly known. However, it has been suggested that these β1,4-galactosyltransferases have overlapping functions.

Immunodefi ciency and attenuated B cell formation has been observed in ST6Gal1-knockout mice137. ST3Gal4 is involved in the biosynthesis of si- alyl-Lewisx and sialyl-Lewisa antigens, both known as selectin ligands138. ST3Gal4-defi ciency causes infl ammation response defi cit, formation of defi cient platelets and defects in blood coagulation139. The role of ST- 3Gal6, the fourth α2,3-sialyltransferase in the N-glycan synthesis is still obscure140.

Fucosyltransferases do not seem to be essential in mice141-144. However, FUT genes have an important role in hematopoiesis and the immune response. FUT4-null mice have partial neutrophil adhesion defi cit and selectin ligand defect similar to the FUT7-knockout144,145. The selectin ligand activity is completely lost in FUT4 and FUT7 knockout mice144, suggesting that the gene products are pivotal re- sponsible for the synthesis of active selectin ligands. In humans, the mutation in the FUT7 gene results in modest reductions of leukocyte selectin ligand activity146.

Considerable glycosylation differences are prevalent between organisms. All other mammals except the humans, apes and Old World monkeys, have functional α1,3-galactosyltransferase147. In humans, the Gal- α1,3-Gal glycoconjucates synthesized by α1,3-Galactosyltransferase REVIEW OF THE LITERATURE 33 are antigenic and induce serum anti-Gal antibodies148. Moreover, the defect in N-glycolylneuraminic acid synthesis is specifi c to humans149. The glycosylation differences between organisms need to be taken into account when defi ning the components of a cell culture media for stem cells to be transplanted into humans150,151.

Glycosylation in hematopoiesis

One of the most dramatic alterations of glycosylation occurs at the time of postnatal development, where fetal erythrocyte i type polylactosamine changes into branched I type polylactosamine152. Thus, glycans can be used as biomarkers for cellular maturity. Moreover, typical glyco- sylation features of hematopoietic lineages separate the different cell types. Lymphoid cells have a large amount of N-glycan α2,6-sialylation, whereas sialyl-LewisX antigens are enriched in myeloid cells153,154. In these cells, a localized increase in α2,6-sialylation of CD11b/CD18 molecules has been seen during myeloid cell maturation and liberation from BM155.

Glycosylation of HSCs is still poorly known. α2,6-sialylation is also dominant in CD34+ cell N-glycans156. α2,6-sialylation has been increased in granulocyte growth stimulating factor mobilized CD34+ cells when compared to naturally circulating CD34+ cells157. It has been suggested that sialylation events modify the adhesion between HSCs and osteoblasts158.

Effect of glycosylation on homing

Glycosylation plays an important role in the adhesion and homing of HSCs and leukocytes occurring in a selectin-dependent manner159. Leu- kocyte rolling on endothelial cells is supported by the interaction between the lymph node homing receptor L-selectin and its ligands. Leukocytes express the P-selectin glycoprotein ligand 1 which is able to bind to L-, P- and E-selectins160,161. The selectin binding of P-selectin glycoprotein ligand 1 is dependent on O-glycosylation162.

CB CD34+ cells have impaired α1,3-fucosylation not seen in BM HSCs163. CB CD34+ cells express a form of P-selectin glycoprotein ligand 1 that does not bind to P-selectin163. Enforced α1,3-fucosylation of CB CD34+ cells improved the binding to P-selectin possibly in a P-selectin glycopro- 34 REVIEW OF THE LITERATURE tein ligand 1 -dependent manner164. α1,3-fucosylation is important for the synthesis of the selectin ligand glycan structure sialyl-Lewisx144.

A selectin ligand, CD44, has a specifi c role in CD34+ cells binding to both L and E-selectin165,166. A CD34+ cell specifi c glycoform of CD44 contains N-glycosylation, which is crucial to its function165,167. CD44 has been suggested to play a major role in CD34+ cell adhesion and movement along endothelium and leukocytes168.

In human leukemia CD34+ KG1a cells, the CD34+ cell-specifi c CD44 has been shown to have a 5-fold higher affi nity to selectin binding than P-se- lectin glycoprotein ligand 1165,166,168. Both P-selectin glycoprotein ligand 1 and CD44 may participate in the homing of HSCs and the dysfunction- al P-selectin glycoprotein ligand 1 in CB-derived cells may conduce to reduced homing capacity of CB HSCs164,165,168. The enforced fucosylation of the glycans of CB CD34+ cells have shown contrasting results in their ability to improve their homing163,164.

The modifi cation of HSCs by glycan remodelling may provide new tools for HSC therapeutics. Engineering of cells through glycan modifi cations could be used to enhance HSC homing capacity. This would provide a greater use for CB HSCs, as a lower number of cells might be suffi cient for successful engraftment. AIMS OF THE STUDY 35

AIMS OF THE STUDY

The aims of this study were

1. To characterize the global gene expression and glycome of human hematopoietic CD133+ and CD34+ cell populations from CB (studies I-III) 2. To reveal the differential gene expression in CD133+ and CD34+ cells and to relate the fi ndings to the characteristic biological processes of these cells (study II) 3. To identify the genes responsible for cell type-specifi c N- glycosylation in CD133+ and CD34+ cells (study III) 4. To fi nd novel molecular markers specifi c to HSCs (study II) 36 MATERIALS AND METHODS

MATERIALS AND METHODS

Ethics

This study was approved by the ethical board of the Finnish Red Cross Blood Service and Helsinki University Hospital. The CB donors were informed and written consent was obtained. Only CB units unsuitable for therapeutic use were studied. A small total volume or the low number of total cells were the most common reasons for discharging CB units.

Cells

CB units were collected at the Helsinki Maternity Hospital and the Department of Obstetrics and Gynaecology at Helsinki University Hospital. The collections were performed during 2004-2007. In all the studies, fresh CB units were used.

In study I, CD133+ cells were selected from the 4 CB unit and CD133- cells were collected for control purposes. CD133+ data were compared to CD133- data from the same CB unit in the microarray analysis. The differential gene expression between CD133+ and CD133- cells was to be the same in all replicates. The fold-change cutoff value was set to 3. To verify the information obtained from the microarray data, selected genes were subjected to qRT-PCR analysis using pools of three samples in each. Analysis was performed on two biological replicates. In addition, CD133+ and CD133- cells were used to evaluate the purity of the cell fractions. The CFU assay was performed for CB CD133+, CD133- and mononuclear cells in duplicate.

In study II, CD34+ cells were selected from the 3 CB unit and CD34- cells were collected for control purposes. Similarly to study I, CD34+ data were compared to CD34- data from the same CB unit and the results verifi ed using qRT-PCR analysis. Moreover, the assessment of the purity of the cell fractions and the CFU assay were performed for CD34+ and CD34- cells as in study I. Further, similarly constructed CD133+ and CD34+ gene expression profi les were compared to reveal the specifi c expression patterns and differences of CD133+ and CD34+ cells. In addition, gene expression data of CD133+ and CD34+ cells were directly compared to the gene expression data of PB mononuclear cells. Thus, the PB mononuclear cell sample was provided as a common MATERIALS AND METHODS 37 benchmark to control the differential gene expression in CD133+ and CD34+ cells.

In addition, the gene expression profi le results suggested several putative membrane proteins that could serve as an HSC marker in the isolation of HSCs. For the analysis of the presence of novel HSC marker molecules, selectively monoclonal antibody-labeled CB mononuclear cells were the subject of the fl ow cytometric analysis.

In study III, CD133+ and CD133- cell N-glycans were isolated for mass spectrometric analysis. During the preparation process, ultrapure bovine serum albumin was used to avoid oligosaccharide contamination. To demonstrate the quantitative differences between N-glycosylation of CD133+ and CD133- cells, CD133+ and CD133- cell N-glycan profi les were compared to each other. In addition, CB CD133+ and CD8+ T cell N-glycan profi les were compared to each other. The CD8+ T cell N-glycan profi le served as an independent, immunoganetically selected mature hematopoietic cell control.

The mononuclear cells were analyzed in nuclear magnetic resonance (NMR) analysis to obtain detailed data about most abundant N-glycan structures. In addition, N-glycan fractions of CD133+ and CD133- cells were digested with specifi c exoglycosidase enzymes to characterize the terminal epitopes on CD133+ and CD133- cell N-glycans. Further, the glycan profi le and glycosylation-related gene expression were compared to unravel the transcriptional regulation of the glycosylation features characteristic of CD133+ cells. Also, the glycosylation-related gene expression of CD34+ cells was observed. The glycan determinants on the cell surface of mononuclear and CD34+ cells were demonstrated by a lectin binding assay. For this, Glycophorin A-depleted CB mononuclear cells from 3 CB units were used.

Methods

Methods used in the following studies (I-III) are described in detail in the original publications and are listed in Table 3. 38 MATERIALS AND METHODS

Table 3. The methods used in studies I-III36,100,169-180. MATERIALS AND METHODS 39

Gene expression reanalysis

The improved knowledge on gene and transcript sequences, as well as genome assembly, reveal a great number of previously defi ned probe sets in Affymetrix microarray GeneChips that are not applicable in the detection of the gene. The unreliable representative gene identifi er has been assigned for approximately 18% of the probe sets in the Affymetrix U133 Plus 2.0 chip181. Moreover, about 12% of the probe sets contain probes that may hybridize to transcripts of more than one gene or to a non-coding sequence, while some probes no longer match any sequence in the current databases. Thus, reanalysis of all the gene expression data was performed to access more accurate annotation of the differentially expressed genes.

Raw data was normalized using the GC Robust Multi-array Average background adjustment for intensities, quantile normalization, and median-polish summarization182. Biostatistic tools were used in R software. The probes of each Affymetrix probe set, directed toward identifi cation of a transcript, were redesigned to sets of probes using GeneChip library fi les (http://brainarray.mbni.med.umich.edu/ CustomCDF). The probe sets of these library fi les were chosen to match a more recently updated Unigene cluster representing certain genes (http://arrayanalysis.mbni.med.umich.edu/ps/)181. To obtain CD133+ and CD34+ cell gene expression profi les, CD133+/CD34+ cells were compared to CD133-/CD34- cells. From the CD133+ and CD34+ profi les, 500 genes with the most differential expression pattern were selected according to their median fold change. That is, the ratio of the median gene expression level in CD133+/CD133- cells or CD34+/CD34- cells was calculated and transformed to an absolute value of log2. Differentially expressed genes could be overexpressed or underexpressed in CD133+/ CD34+ cells when compared to CD133-/CD34- cells. The statistical signifi cance of the differential expression was performed using a T-test. The differentially expressed genes with a q-value>0.05 refl ected the probability of a false positive result and were discharged. To directly compare differences in CD133+ and CD34+ cells, a pairwise analysis utilizing GeneSifter software was employed183. The signifi cant change in the expression between CD133+ and CD34+ cells was validated using Student’s T-test and a p-value that was ≥0.05. 40 MATERIALS AND METHODS

Annotation

Due to the ongoing changes and revisions in the annotations of Affyme- trix probe sets, the differentially expressed genes in studies I and II were reannotated using the GeneAnnot Microarray Gene Annotation software (Weizmann Institute of Science, http://bioinfo2.weizmann.ac.il/genean- not/)184,185. The Database for Annotation, Visualization and Integrated Discovery tools were employed to obtain molecular functions, biological processes, and cellular components for the transcripts common in the CD133+ and CD34+ expression profi les in the original and reanalyzed data186. The molecular interactions for the protein products were as- sessed through GeneSifter software, which uses Kyoto Encyclopedia of Genes and Genomes Pathway database16. RESULTS AND DISCUSSION 41

RESULTS AND DISCUSSION

CD133+ and CD34+ cells have similar gene expression (studies I and II)

The comparison of CD133+ and CD133- data sets resulted in 690 tran- scripts that were differentially expressed at least three-fold in all the samples. Of these, 393 transcripts were upregulated and 297 were down- regulated in CD133+ cells. Similarly, CD34+ data sets were compared to CD34- data sets and 620 differentially expressed transcripts with at least a three-fold difference were found. In CD34+ cells, 169 transcripts were overexpressed and 451 transcripts underexpressed. The inte- grated sets of differential gene expression between CD133+ and CD133- cells, and CD34+ and CD34- cells, showed that 85% of the transcripts overexpressed in both CD133+ and CD34+ cells and 36% of the tran- scripts underexpressed in both CD133+ and CD34+ cells were the same. Transcripts found in both CD133+ and CD34+ cells showed the highest overexpression when compared to CD133-/CD34- cells. Both cell types expressed many genes previously identifi ed to be present in HSCs (such as KIT, LAPTM4B, MEIS1, RBPMS, HLF, BAALC and C17)109,115,120,187-190. Also, many genes supporting the undifferentiated state of HSC (such as GATA2 and N-MYC and HOXA9)106,191,192. These genes were expressed at a very similar expression level across the CD133+ and CD34+ cell replicates indicating an important role for these genes in the regulation of CD133+ and CD34+ cells. Both cell types expressed a high number of transcripts. In addition, these transcripts were arranged similarly in SOM analysis suggesting similar regulation for the common genes in CD133+ and CD34+ cells. In the CD133+ profi le, the number of over- expressed transcripts was higher than in the CD34+ profi le. This could explain the additional cluster of overexpressed transcripts illustrated in U-matrix of CD133+ data.

CD133+ and CD34+ cells expressed 85 transcripts that were not pres- ent in CD133- or CD34- cells. These transcripts were either downregu- lated or absent in PB mononuclear cells. The 85 common transcripts represented 80 genes. The encoded molecules are involved in cell pro- liferation, metabolism and localization. Of these, the expression of 44 genes in HSCs has been confi rmed in other studies109,115,120,188,193. The transmembrane protein encoding genes FLT3 (CD135), ALCAM (CD166) and DSG2 were expressed in CD133+ and CD34+ cells only. Moreover, expression of SV2A was found in CD133+ cells, but not in CD34+ cells. 42 RESULTS AND DISCUSSION

Novel molecular markers for HSCs could not be found (study II)

To determine whether transmembrane protein encoding genes expressed solely in CD133+ and CD34+ cells could provide novel molecular markers for HSCs, the presence of the protein products on the cell surface were observed. Total leukocyte population was labeled with specifi c antibodies binding to CD135, CD166, DSG2 or SV2A. In addition, anti-CD34 or anti-CD133 antibody labeling was used. Coexpression of CD135 and CD34/CD133 molecules was demonstrated (Figure 7). Moreover, CD166 was expressed on some of the CD133+/CD34+ cells (Figure 6). CD135 was also present on 11% of the leukocytes and CD166 was carried by 20% of the leukocytes. Thus, neither CD135 nor CD166 alone can be used for selective purifi cation of HSCs.

SV2A or DSG2 molecules could not be found on leukocytes, including CD133+/CD34+ cells. It is possible that the antibodies against SV2A and DSG2 molecules do not posses adequate specifi city. Therefore, no single molecular marker to identify HSCs could be found using microarray analysis. In addition, known embryonic stem cell markers SSEA3, SSEA4, TRA-1-60 and TRA-1-81 were searched for on the leukocytes (data not shown). No hematopoietic cell populations carrying these markers could be demonstrated.

Figure 7. Presence of CD135 and CD166 molecules on CD34+ and mononuclear cells. RESULTS AND DISCUSSION 43

Differential gene expression between CD133+ and CD34+ cells suggests that CD133+ cells are more primitive (studies I and II)

Despite the highly similar gene expression in CD133+ and CD34+ cells, unique gene expression patterns related to divergent biological processes were distinguished in CD133+ and CD34+ cells. The number of overexpressed transcripts in CD133+ cells, when compared to CD133- cells, was higher than in CD34+ cells when they were compared to CD34- cells.

In the study I, a set of 257 transcripts were identifi ed expressed in CD133+ cells, but not in CD133- cells. Of these, 125 genes were also expressed in CD34+ cells. However, the 172 transcripts overexpressed solely in CD133+ cells (when compared to CD133- cells) showed signifi cant association with regulation of chromatin structure, cell cycle and DNA metabolism. CD133+ cells expressed SMARCA1, SMARCA2 and SMARCA4. SMARCA genes are related to chromatin remodeling pathway194,195. Only SMARCA1 expression was detected in CD34+ cells. The high number of expressed transcripts is typical for HSCs that have been suggested to have permissive or more open chromatin structure.

Embryonic stem cell-related transcripts including DNMT3A, DNMT3B and DPPA4 were shown only in CD133+ cells. The expression of transcripts related to pluripotency suggests a more primitive nature for CD133+ cells. The embryonic-type gene expression may also be due to the CB origin of the cells. The expression of MPDZ, encoding a protein associated with tight junctions and maintenance of cell polarity, suggests that some of the CD133+ cells may be going through asymmetric cell division.

The products of the expressed transcripts in CD34+ cells were related to regulation of development, response to stimulus and DNA metabolism. These biological processes were also typical of CD133+ cells. On the other hand, CD34+ cells expressed transcripts encoding markers for lineage committed cells.

Several studies have shown transcriptional as well as functional differences between CD34+ cells from CB, BM and PB115,196-198. These studies have suggested a more primitive nature for CB CD34+ cells. Moreover, differential gene expression has been found between CB CD133+ and PB CD133+ cells193. This study demonstrated divergent gene expression between CB CD133+ and CD34+ cells. The gene expression profi les of 44 RESULTS AND DISCUSSION

CD133+ and CD34+ cells were identically constructed. The samples were prepared in the same laboratory, hybridized at the same time and analyzed using equal parameters. Therefore, many nonbiological error sources for differential gene expression between the two cell populations were avoided. The CD133+ and CD34+ gene expression profi les of the present study were also compared to CD133+ cell gene expression profi le in another study193. CD133+ gene expression profi les were more similar to each other than with CD34+ cell gene expression profi le of the present study. This confi rms that the different transcriptional expression in CD133+ and CD34+ cells is due to differences in these two cell populations.

The differentially expressed transcripts between CD133+ and CD34+ cells were related to divergent biological processes, suggesting different role for these cells in hematopoiesis. The divergences between CD133+ and CD34+ cell populations may be more obvious in those cells from BM or PB. It may also be that the divergences between CD133+ and CD34+ cells refl ect specifi c primitiveness of CB CD133+ cells.

CD133+ and CD34+ cells demonstrate similar total colony forming potential (studies I and II)

CFU assay was used to identify primitive hematopoietic cells from CD133+, CD34+ CD133-, CD34- and mononuclear cells. A similar number of total CFUs were formed by CD133+ and CD34+ cells. CD133+ cells formed almost only multipotent granulocyte-erythroid-macrophage- megakaryocyte (38%) and granulocyte-macrophage colonies (58%). Burst-forming colonies represented 4.2% of the progeny in CD133+ cells, whereas no erythroid colonies were observed. A vast majority of CD34+ cell progeny was also multipotential, 33% granulocyte- erythroid-macrophage-megakaryocyte colonies and 58% granulocyte- macrophage colonies. A little more of both the burst-forming (7.2%) and unipotent erythroid colonies (1.4%) were formed by CD34+ cells (Figure 8). Combining the CFU assay and gene expression profi ling results it seems likely that CD133+ cells are more primitive in their nature when compared to CD34+ cells. RESULTS AND DISCUSSION 45

Figure 8. Clonogenic progenitor capacity of CD34+ and CD133+ cells by CFU as- say. Abbreviations: BFU, burst-forming erythroid; CFU, colony-forming unit; GEMM, granulocyte-erythroid-macrophage-megakaryocyte; GM, granulogyce-macrophage.

Reanalysis of the original gene expression data focuses on the similarities between CD133+ and CD34+ cells

Due to the fast development of gene expression analysis and data normalization methods, the original data from microarray analysis (study I and II) were reanalyzed using a more modern method. In the reanalysis, the top 500 genes with differential expression in CD133+/ CD133- or CD34+/CD34- cells were chosen. After discharging putative false positive results, based on the q-value, 492 genes in the CD133+ gene expression profi le and 425 genes in the CD34+ gene expression profi le were considered valid (Figure 9). Of these, CD133+/CD133- and CD34+/CD34- cells had 283 differentially expressed genes in common. In comparison to CD133- and CD34- data, 118 genes were overexpressed and 165 genes were underexpressed in CD133+ and CD34+ cells.

Reanalysis of the microarray data verifi ed the overexpression of 67 genes in CD133+ and CD34+ cells when compared to CD133- and CD34- cells. These include KIT, FLT3, MEIS1, HOX9A, GATA2 and SOCS2 genes with a signifi cant role in HSC regulation. Similarly, 73 genes downregulated in CD133+ and CD34+ cells, when compared to CD133- and CD34- cells, were the same between the original and reanalyzed data. No genes that were defi ned as overexpressed in CD133+ and CD34+ cells by the original data analysis were shown to be underexpressed after reanalysis, or vice versa. However, the reanalysis of the data identifi ed 51 new genes with a higher expression level in CD133+ and CD34+ cells than in 46 RESULTS AND DISCUSSION

Figure 9. Schematic representation of the number of differentially expressed transcripts in CB CD133+ and CD34+ cells (when compared to CD133- and CD34- cells) and their intersections. A) In the original data, differentially expressed transcripts between CD133+/CD133- and CD34+/CD34- samples represented at least a three- fold change in all sample pairs (studies I and II). B) Reanalysis results represented the most differentially expressed genes between CD133+/CD133- and CD34+/CD34- samples based on the mean gene expression level of the replicates. No fold-change cutoff value was used.

CD133- and CD34- cells (data not shown). Moreover, 92 new genes were downregulated in CD133+ and CD34+ cells when compared to CD133- and CD34- cells. The role of these novel genes in the regulation of HSCs requires further investigation.

The association with a biological process was defi ned for the set of differentially expressed genes in the original data and the reanalyzed data. Only the genes similarly expressed in CD133+ and CD34+ cells were considered. Annotation was performed anew to both data sets (Figure 10). In both cases, the biological processes representative of CD133+ and CD34+ cells were clearly different from their negative counterparts. The associated biological processes enriched in CD133+ and CD34+ cells have been shown to be characteristics to HSC populations106,109,199. Many of the related biological processes were the same between the original data and the reanalyzed data, especially in the processes that represent CD133- and CD34- cells.

The causes for genes with higher expression level in CD133+ and CD34+ cells than in CD133- and CD34- cells related to catalytic activity (38 genes), cytokine activity (5 genes) and endopeptidase/protease inhibitor activity (4 genes). Cytokine activity-associated genes SOCS2, TNFSF4, TRIP6, PXDN and IPO11 were the same as identifi ed in the original data (studies I and II). The 4 protease inhibitor genes were SERPING1, SERPINE2, SPINK2 and TFPI. Similarly to other protease inhibitors200,201, RESULTS AND DISCUSSION 47

Figure 10. Biological processes represented by differentially expressed genes in CD133+ and CD34+ cells when compared to CD133- and CD34- cells. A) Original data (study II) and B) reanalyzed data. these genes may have a role in the modulation of cytokine expression. Their overexpression in HSCs has been previously shown112,115,188,193. Forty-nine of the genes overexpressed in CD133+ and CD34+ cells were associated with intracellular compartments, especially to cytoplasm (25 genes) (Table 4).

The genes downregulated in CD133+ and CD34+ cells when compared to CD133- and CD34- cells were associated with cell communication (32 genes), response to other organism (17 genes), apoptosis (16 genes) and cell activation (7 genes). Moreover, the encoded protein products were associated with protein binding (42 genes) and receptor activity (20 genes). The most strikingly emphasized cellular compartment for the encoded gene products was cell membrane (55 genes). CD133- and CD34- cell populations include lymphocytes, mononuclear phagocytes, dendritic cells and granulocytes that act in the immune system. It is crucial for these cells to have active signaling and communication with both other cells and the matrix. The immune response is dependent on detected changes in the environment and the activation of the cell is initiated by extrinsic factors. 48 RESULTS AND DISCUSSION

Table 4. The most common cellular compartments for products encoded by expressed genes in CD133+ and CD34+ cells. RESULTS AND DISCUSSION 49

The main functional categories related to the CD133+ and CD34+ gene expression profi les were the same in the original data and the reanalyzed data using the most recent annotation. There are several reasons to explain the differences between the original data and the reanalyzed data. These are: the difference between the redesigned probe sets and original Affymetrix probe sets, different preanalysis and normalization of the data, and the criteria of the differentially expressed genes. Redesigned probe sets or sequence matched probes have shown a 30-50% discrep- ancy of results in differential gene expression analysis when compared to analysis using the original Affymetrix probe sets181,202. It is important to notice that the differences between the original and reanalyzed data sets of differentially expressed genes in CD133+/CD133- and CD34+/ CD34- cells are not necessarily faulty results. Well-characterized genes expressed in CD133+/CD34+ cells are not all among the list of the most highly expressed genes in the reanalysis due to the selection criteria that was used. Thus, the differential gene expression that was shown in the original data, but not in reanalyzed data, is still in the main part usable. However, redesigned probes have the improved capacity to de- tect the expressed genes. Thus, the reanalysis of the gene expression data may help in identifi cation of new genes and provide novel data for further studies.

CD133+ and CD34+ cells have divergent roles in hematopoietic regeneration (studies I and II)

The divergent gene expression between CD133+ and CD34+ cells was revealed by direct pairwise analysis. The represented analysis was specially focused on genes associated with hematopoiesis, cell cycle and cell adhesion. CD133+ cells had a higher expression level of CD133, CD34, CD135 (FLT3) and CD117 (KIT) genes. The higher expression of CD34 in the CD133+ cell population may be explained by the higher abundance of CD34 on the cell surface of CD133+ cells. Nearly all CD133+ cells are CD34-positive, whereas the CD34+ cell population includes lineage committed progenitors that carry a lower number of CD34 molecules on their cell surface.

The hematopoietic lineage-specifi c markers were downregulated in CD133+ and CD34+ cells when compared to their negative counterparts. However, there was a difference in the level of underexpression between CD133+ and CD34+ cells (Table 5). CD133+ cells expressed IL3R (CD123), a marker typical for common myeloid progenitors, pronormoblasts and 50 RESULTS AND DISCUSSION megakaryoblasts. The other genes encoding megakaryoblast markers, such as IL-11R or CD126, were not expressed in CD133+ cells. On the other hand, one or more of the CD34+ sample replicates expressed several genes related to hematopoietic diffentiation. The complete list of genes related to hematopoiesis is shown in Table 5.

The lineage differentiation marker expression may be connected to the cells in the CD133+ and CD34+ subpopulations. Hence, these results suggest that the CD34+ cell population contains a higher proportion of pro T cells, other differentiating T cells, promonoblasts, monoblasts and megakaryocytes than the CD133+ cell population (Figure 1). On the other hand, it has been suggested that HSCs express lineage differentiation marker genes at a low level to enable the fast differentiation of HSCs when a differentiated cell type is needed in the blood system. It may also be that CD34+ cell population consists of a higher number of short- term HSCs with better readiness to form all types of blood cells.

CD34+ cells expressed a wider variety of genes encoding cell adhesion molecules, such as selectin E and selectin P. Selectins are important for cell traffi cking. CD34+ cells may thus have greater capacity to be mobilized. However, the engraftment potential of CB HSCs has been delayed when compared to HSCs from other sources203. The presence of very late antigen 4 has been proven crucial for the homing of HSCs204. The expression of gene coding for very late antigen 4 was higher in CD133+ cells.

The gene expression profi le of CD133+ cells revealed the expression of many genes related to the cell cycle (study I). CD133+ cells expressed 20 cell cycle genes on a higher level than CD34+ cells. These products of the cell cycle genes have inhibitor as well as promoter effects. The results suggest that both quiescent cells and actively cycling cells are found in the CD133+ cell population. Long-term HSCs are mostly considered to be noncycling, but that is not as critical a parameter in CB HSCs91. Hence, CB HSC in an active cell cycle state can have self-renewing capacity.

CD133+ and CD34+ cells had a high number of commonly expressed genes typical of HSCs. They also had many overlapping biological processes. Both CD133+ and CD34+ cells have been successfully used in the reconstruction of the blood system in irradiated patients’ marrow. Thus both cell populations contain cells with long-term repopulation and short-term repopulation potential. The direct comparison of CD133+ and CD34+ cells revealed very delicate differences between the cell types. Previous studies have shown that differences in the gene expression RESULTS AND DISCUSSION 51 profi le refl ect true changes between closely related cell populations205,206. Thus, the differences demonstrated in expression profi les of CD133+ and CD34+ cells suggest functional divergences between the cell types.

The gene expression profi les indicate that faster repopulation of the recipient could be obtained after CD34+ cell transplantation. Due to the more primitive nature of CD133+ cells, selected CD133+ cell population may not include enough cells readily available for development of all blood cell types or they may have poorer capacity to travel into the BM,

Table 5. Differential expression of hematopoietic cell marker genes in CD133+ and CD34+ cells. 52 RESULTS AND DISCUSSION or both. CD133+ cells, as a more primitive source of HSCs, could be better suited for HSC expansion and gene therapy. Due to the limited number of transplantations performed using selected CD133+ and CD34+ cells, it is not possible to say whether there is a difference in the repopulation after transplantation. It should be kept in mind that despite the divergent biological capacities, binding of an antibody to the CD133 or CD34 molecules may infl uence cell function.

N-glycosylation-related gene expression is similar in CD133+ and CD34+ cells and correlates with the N- glycome profi le (study III)

CD133+ and CD34+ cells had characteristic N-glycosylation-related gene expression when compared to CD133-/CD34- cells (study III). Moreover, mass spectrometric analysis of CD133+ and CD133- cells, combined with results from NMR and exoglycosidase digestion, revealed differences in the glycosylation of these cells. CD133+ cells contained signifi cantly more fully processed, especially biantennary type N-glycans than CD133- cells (Figure 11). Yet, fully processed multiantennary N-glycans were fewer in CD133+ cells than in CD133- cells. The most frequent glycosylation of CD133+ cells was unprocessed, high-mannose type N-glycans. High mannose type N-glycans were more abundant in CD133+ cells than in CD133- cells. Enrichment of high mannose type N-glycans in CD133+ cells was evident in the comparison of CD133+ cells to CD8+ cells which represent a mature leukocyte population (data not shown). However, more analyses are needed to confi rm that the differences in the relative abundance of neutral N-glycans are due to tissue-specifi c glycosylation changes.

Characteristic mannosylation pattern of CD133+ and CD34+ cells (study III)

The differential expression of one mannosidase and three glycosyltransferase encoding genes were related to the changes observed in the CD133+ and CD133- cell glycome. The differential gene expression was observed in both CD133+ and CD34+ cells as compared to their negative counterparts. CD133+ and CD34+ cells, but not CD133- and CD34- cells, lacked the expression of MAN1C1 encoding one of the key α1,2-mannosidases participating in N-glycan maturation. No difference was seen in the expression of other α1,2-mannosidase encoding genes RESULTS AND DISCUSSION 53

MAN1A1, MAN1A2 and MAN1B1, yet their expression was detected in CD133+ and CD34+ cells as well as CD133- and CD34- cells.

MAN1 enzymes remove α1,2-mannosidases from different positions in N-glycans, and their functions are highly overlapping (Figure 6). MAN1A, MAN1B and MAN1C are located in different genes on 6 and 1, and they have independent regulatory pathways. The unique specifi city of MAN1C1 encoded α1,2mannosidase 1C is that it prefers the removal of terminal α1,2-linked mannose on the α1,3 branch in 207 Man9GlcNAc2 and Man8GlcNAc2 isomer B . The α1,2-mannosidases 1A and 1B remove terminal α1,2-linked mannose from the α1,6 branch from Man8GlcNAc2 isomer B before the α1,3 branch is demannosylated. Hence, the absence of MAN1C1 expression in CD133+ and CD34+ cells may affect the enrichment of high mannose N-glycans. It is likely that most of the high mannose N-glycans seen in the CD133+ and CD133- cell glycan profi le were intracellular. N-glycan profi les include all N- glycans, not only those on the cell surface. However, labeling with mannose binding lectins Pisum sativum agglutinin and Hippeastrum hybrid agglutinin confi rmed the presence of mannose-rich N-glycans on the cell surface.

High mannose N-glycans promote proper protein folding208. The high mannose N-glycans transported to the cell surface are known to be demannosylated. A half-life of 6.7 hours for cell surface high mannose

Figure 11. Schematic representation of the differences in CD133+ and CD133- cell N- glycan profi les. In CD133+ cells, high mannose N-glycans and biantennary complex type N-glycans were the most enriched N-glycan structures. In contrast, the proportion of low mannose type N-glycans and multiantennary structures greater than biantennary com- plex N-glycans was increased in CD133- cells. Monosaccharide symbols: = N-acetyl-D- glucosamine; = D-mannose. Glycosidic linkages are indicated by lines connecting the monosaccharides. 54 RESULTS AND DISCUSSION

N-glycans has been observed in the hepatic cell line209. On the other hand, 4TO7 tumor cells are capable of spreading to the lungs and they have been shown to have more high mannose and N-glycans on their cell surface than nonmetastatic tumor cells210. Further, it has been shown that sialic acids have no effect on the migration and invasion properties of 4TO7 cells. Thus, high mannose N-glycans may have yet unknown functions.

The 5% increase of high mannose type N-glycans demonstrated in CD133+ cells when compared to CD133- cells may correspond to a variety of . There may be a greater number of recently transported proteins on the surface of CD133+ cells. Hence, there may simply be more time for trimming mannose residues to form the low mannose type N-glycans more abundant in CD133- cells. If there is a greater proportion of mannose-rich glycans on the surface of CD133+ cells, it could be due to the lower number of mannose trimming enzymes in these cells. The unique mannosylation of CD133+ cells may be useful for classifi cation of a primitive cell type. The lack of MAN1C1 expression may function as a marker for HSCs.

Differential expression of N-acetylglucosaminyl- transferase genes and the enrichment of biantennary complex type N-glycans in CD133+ cells (study III)

Differential expression of two genes encoding N-glycan backbone branching N-acetylglucosaminyltransferase enzymes was demonstrated between CD133+/CD133- and CD34+/CD34- cells. MGAT2 was overex- pressed 1.9-fold in CD133+ cells when compared to CD133- cells. MGAT2 overexpression was 1.7-fold in CD34+ cells when compared to CD34- cells. MGAT4A was underexpressed 2.8-fold in CD133+ cells and 2.0- fold in CD34+ cells when compared to their negative counterparts. The increase in the amount of biantennary complex type N-glycans demon- strated in CD133+ cells correlated with the higher expression of MGAT2. Furthermore, a lower number of triantennary and other multiantennary N-glycans were seen in CD133+ cells. MGAT4 gene encoding N-acetyl- glucosaminyltransferase 4 participates in the biosynthesis of trianten- nary and other multiantennary complex type N-glycans. Thus, underex- pression of MGAT4A may be related to the reduction of multiantennary N-glycans in CD133+ and CD34+ cells. Complex type N-glycans seem to be developmentally important as MGAT2-null mice suffer from he- matological and osteogenic abnormalities and die soon after birth. This RESULTS AND DISCUSSION 55 indicates that complex type N-glycans could have a function in the in- teraction between HSCs and BM osteoblasts. High binding of Phaseolus vulgaris-derived Phytohemagglutinin-L lectin, specifi c to large complex type N-glycans, demonstrated the presence of large complex type N- glycans on leukocytes including the CD34+ cells.

Despite the differential expression of genes coding for β1,4-galactosyltransferases there are no differences in the β1,4-galactosylation of CD133+ and CD133- cells (study III)

Type 2 N-acetyllactosamine dominated as glycan backbone structure in CD133+ and CD133- cells. Mass spectrometric analysis results were verifi ed by binding of Ricinus communis agglutinin-1 lectin. Moreover, type 2 N-acetyllactosamine were β1,4-galactosylated as β1,3-linked galactose was not detected. Genes encoding β1,4-galactosyltransferases such as B4GALT1, B4GALT3 and B4GALT4 were expressed in CD133+, CD133-, CD34+ and CD34- cells. B4GALT3 was downregulated in CD133+ and CD34+ cells by 2.3- and 2.0-fold respectively, when compared to their negative counterparts.

The differential expression of β1,4-galactosyltransferase encoding genes was not shown to have any effect on N-glycosylation. Yet, it may not be ruled out that the differential expression of β1,4-galactosyltransferase encoding genes still have a signifi cant function in the galactosylation of specifi c proteins not detected in glycan profi ling, or have an impact on the galactosylation of other glycan types such as O-glycans or .

CD133+ and CD34+ cell specifi c biosynthesis of N-glycan terminal epitopes (study III)

The complex type N-glycans, biantennary and larger, were very often α2,6-sialylated in CD133+ and CD133- cells. ST6Gal1 encoding α2,6- sialyltransferase was similarly expressed in CD133+/CD133- cells, and CD34+/CD34- cells. Lectin Sambucus nigra agglutinin recognizing α2,6- linked sialic acid bound to 98% of the leukocytes including CD34+ cells. Thus, α2,6-sialylation was a dominant terminal epitope on CD133+ and CD133- cells. Moreover, α2,3-sialylation had increased on CD133+ cells when compared to CD133- cells. The α2,3-sialidase digestion disclosed the presence of some solely α2,3-sialylated N-glycans in CD133+ cells. 56 RESULTS AND DISCUSSION

The α2,3-sialidase treatment was not able to entirely desialylate N- glycans in CD133- cells. CD133+ and CD34+ cells overexpressed the α2,3- sialyltransferase encoded by ST3GAL6 when compared to their negative counterparts (CD133- and CD34- cells). ST3GAL6 expression was 3.9-fold higher in CD133+ cells than CD133- cells, and 1.6-fold higher in CD34+ cells than CD34- cells. Lectin Maackia amurensis agglutinin with α2,3- linked sialic acid specifi city bound to 62% of the leukocytes and CD34+ cells demonstrating the presence of α2,3-sialyl-N-acetyllactosamine structures on the cell surface.

α2,3- and α2,6-sialyltransferases compete for the same N-glycan substrates. Lowered amount of α2,6-sialyltransferase and decreased α2,6-sialylation in murine T cells was suggested to be caused by substrate competition with α1,3-galactosyltransfase211. However, α1,3- galactosyltransferase is not present in human cells and similar substrate competition is not relevant in humans. The lower relative abundance of α2,6-sialylation in CD133+ cells is likely to be caused by increased α2,3-sialylation. ST6Gal1 or ST3Gal6 have not been shown crucial for development. B cells express CD22 (Siglec2) that specifi cally binds to α2,6-linked sialic acid. However, Siclec2 functions only in activated B cells, while it is masked by the cell's own α2,6-sialylation in resting B cells212. B cells that have reduced sialylation have been shown more susceptible to apoptosis213. Similar masking by sialic acids could also exist in other cell types to prevent Fas receptor ligand driven apoptosis. HSCs upregulate Fas upon active cell cycle, but remain resistant to Fas-induced suppression214. Fas ligand coating of HSCs improves their engraftment capacity by two- fold215. In addition, HSCs have shown to secrete Fas ligand which targets the marrow cells of the recipient in transplantation215.

Sialylation has been suggested to play a role in the initial attachment of HSCs to BM osteoblasts158. However, the participation of any single adhesion molecule in the homing of HSCs has not been unequivocally shown216. Glycosylation-based recognition may be involved in the initial steps of homing, but the binding of specifi c proteins are required for engraftment. The interaction may involve expression of stem cell factor and stromal cell-derived factor 1, activation of lymphocyte function- associated antigen 1, very late antigen 4, very late antigen 5 and CD44, cytoskeleton rearrangements and secretion of membrane type 1–matrix metalloproteinases217. In the future, it would be interesting to fi nd out whether α2,3-sialylation is the only form of sialylation in certain proteins. It is also not yet known if solely α2,3-sialylated structures are typical of BM CD133+ cells as well. More studies are needed to reveal whether RESULTS AND DISCUSSION 57 the differential sialylation observed in CB CD133+ cells is benefi cial to cell function or if these cells would benefi t from a higher level of sialylation.

Only a small amount of CD34+ cells have been shown to bind to hyaluronan in BM. The α2,3-sialylation of N-glycans has been shown reduce the binding of CD44 molecule to hyaluronan in CHO cell line218. Enriched α2,3-sialylation may have a role in the mobilization of HSCs to the circulatory system. CD34+ cells carry a specifi c glycoform of CD44 molecule on their cell surface165. It has been shown to have over 5-fold L-selectin binding when compared to P-selectin glycoprotein ligand 1168.

Selectin ligand sialyl-Lewisx is dependent on α2,3-sialylation and α1,3- fucosylation219. In addition to common α1,6-fucosylation of N-glycan core structures, one or two more fucose residues were revealed in CD133+ and CD133- cell N-glycans by mass spectrometric analysis. The fucose was expected to be α1,2- or α1,3-linked to the N-glycans as no type 1 N-acetyllactosamine was found on CD133+ or CD133- cells. FUT1 and FUT2 both encode α1,2-fucosyltransferase, but their expression was not reliably analyzed. FUT1 expression was not detected in CD133+/ CD34+ cells or their negative counterparts. For FUT2 there was no probe available on the array. Ulex europaeus agglutinin I lectin with specifi city towards α1,2-linked fucose residues labeled 53% of the leukocytes. Impaired α1,3-fucosylation and decreased α1,3-fucosyltransferase expression has been shown to be characteristic of CB CD34+ cells163,164. In order to defi ne α1,3-linked fucose on CD133+ or CD133- cell N-glycans, leukocytes were labeled with Lotus tetragonolobus agglutinin lectin. It bound to only 6% of leukocytes. Moreover only FUT4, but no FUT7 expression was detected in CD133+, CD34+, CD133- and CD34- cells. FUT4 was expressed similarly in all cell types. FUT7 encodes the main α1,3-fucosyltransferase responsible for the synthesis of sialyl-Lewisx, but FUT4 protein product is also able to synthesize it. These results suggest that poor α1,3-fucosylation is typical for the CB leukocytes including CD34+ cells. 58 CONCLUSIONS

CONCLUSIONS

The positive selection of HSCs has provided an attractive alternative to T cell depletion of transplantable cells. The selection of CD34+ cells can be used in this process. However, the selection of CD133+ cells has also shown to be promising in the enrichment of HSCs220-222. Although, CD133+ and CD34+ cells are highly similar cell populations there may be differences in their genetic regulation. In previous studies, HSCs from BM, PB and CB have shown differential gene expression related to divergent biological properties operative in HSCs10,155,223. Increased knowledge of HSC biology is crucial to improve stem cell-based therapies.

The present studies characterize human CB-derived CD133+ and CD34+ cells on a genomic level and provide gene expression profi les of CD133+ and CD34+ cells when compared to CD133-and CD34- cells, respectively. The integrated CD133+/CD34+ cell gene expression profi les reveal novel genes to specify HSCs. Moreover, the transcripts that are expressed differentially between CD133+ and CD34+ cells are associated with cell cycle, cell adhesion and differentiation. This fi nding suggests that the divergent gene expression patterns may be adjusting CD133+ and CD34+ cells in different ways in order to encompass hematopoiesis. CD133+ cells seem to be more primitive in their nature and possibly have a wider differentiation potential than CD34+ cells. CD133+/CD34+ cells express numerous transcripts coding for putative transmembrane proteins that may serve as markers for HSCs. However, during this study, specifi c markers to identify HSCs could not be found on the cell surface of CD133+ or CD34+ cells.

These studies also demonstrate N-glycan structure profi le typical for CD133+ cells. CD133+ cells have enriched amount of high mannose type and biantennary complex type N-glycans when compared to CD133- cells. CD133+ cells also have solely α2,3-sialylated N-glycans. Comparison of N-glycan structure and gene expression profi les identify key genes regulating the CD133+ cell-specifi c N-glycan synthesis. The expression pattern of genes encoding glycosyltransferases and glucosidases is similar in CD133+ and CD34+ cells, suggesting a similar N-glycosylation in CD133+ and CD34+ cells. Furthermore, the specifi c binding properties of various lectins are utilized to reveal the glycan determinants on the cell surface. CONCLUSIONS 59

Further studies to identify proteins that carry the specifi c N-glycan structures in CD133+ cells could prove useful in the recognition and isolation of HSCs. Moreover, the role of glycans and glycan binding proteins in the communication between HSCs and their environment should be elucidated. The modifi cation of HSC glycosylation could increase HSC homing and engraftment or target them to specifi c tissues. In conclusion, these results provide new knowledge of CD133+ and CD34+ cells that could help to design cells with enhanced biological properties and to further the use of HSCs in novel therapeutic applications. 60 ACKNOWLEDGEMENTS

ACKNOWLEDGEMENTS

This work was carried out at the Finnish Red Cross Blood Service, Research and Development, in Helsinki during the years 2004-2007. Partial funding support was provided by the Finnish Funding Agency for Technology and Innovation.

I am grateful to the Director of the Institute, Docent Jukka Rautonen, and Department Head, Kari Aranko, for providing excellent research facilities. I devote my warmest thanks to my supervisor, Docent Taina Jaatinen, for sharing her vast knowledge on stem cell biology, but also her enthusiasm, optimism, great patience and motivation that helped to guide me through this learning process. Very warm thanks to my supervisor, Docent Jukka Partanen, for support, numerous interesting discussions when planning the studies and all the helpful comments throughout the PhD work.

I am indebted to the reviewers, Docents Heli Skottman and Timo Tuuri, for their time, constructive criticism and valuable comments. I warmly thank Professor Riitta Lahesmaa for her kind promise to act as an opponent for the public defense of this thesis.

Acknowledgement and sincere appreciation go to Docent Jarmo Laine for his encouragement, support and valuable advice in clinical aspects, Docent Sampsa Hautaniemi for offering his expertise in data analysis, Jari Niemi for his enthusiasm in statistical analyses, Tero Satomaa and Doctor Jari Natunen for sharing their endless knowledge on glycobiology, Olli Aitio for his skillful nuclear magnetic resonance analysis, Doctor Daniel Nicorici for his fl exibility in performing data analysis, Annamari Heiskanen for her timely mass spectrometric analysis and Maria Blomqvist for her work on glycan structure analysis. Additionally, Professor Olli Yli-Harja and Juhani Saarinen are acknowledged for their fruitful collaboration. My special thanks to the undergraduate student Erkka Valo for his work on data reanalysis.

I wish to thank Docent Riitta Kekomäki and the whole staff of the Finnish Red Cross Cord Blood Bank for providing CB units to this study. I am grateful to ’STR’ and all colleagues and technicians who shared their knowledge to help carry out this study, notably; Sirkka Mannelin, Doctor Tuija Kekarainen, Noora Alakulppi, Anita Laitinen, Tanja Kaartinen, Doctor ACKNOWLEDGEMENTS 61

Johanna Nystedt and Docent Leena Valmu. Doctor Hannu Turpeinen ‘transplantation specialist’, Katri Haimila, Doctor Pekka Aroviita and Doctor Suvi Natunen ‘our own glycobiologist’ are thanked for their time and productive input.

My colleagues deserve heartfelt thanks for creating a most enjoyable and variable working atmosphere. I also thank my boss, Research and Development Manager Saara Laitinen, for her sincere care and understanding.

I address special thanks to Maarit Koutonen of Lähituki for the fast and effective support, and to both Marja-Leena Hyvönen and Maija Ekholm for excellent library services. Kevin Hanley is warmly thanked for fl exibly organizing the language revision of this thesis and I am grateful for the always warm and effi cient assistance of Pirjo Nick.

My very sincere thanks to all my friends for their invaluable companionship and the enriching life experiences we enjoy, providing balance to life inside the laboratory. Thank you all for being there for me.

I owe my deepest gratitude to my mother and father for their love and support throughout my life and for my dear little sister Nina who has been looking after the big sister, even the times when it should be the other way around. I am grateful to my grandparents for being part of my life. My sweetest thanks are to my husband Marko for his love, presence and endless support.

January 2008, Helsinki, Finland 62 ACKNOWLEDGEMENTS

REFERENCES

1. McCulloch EA, Till JE. The radiation sensitivity of normal mouse bone marrow cells, determined by quantitative marrow transplantation into irradiated mice. Radiat Res 1960;13:115-125. 2. de Bruijn MF, Speck NA, Peeters MC, Dzierzak E. Defi nitive hematopoietic stem cells fi rst develop within the major arterial regions of the mouse embryo. EMBO J 2000;19:2465-2474. 3. Mikkola HK, Gekas C, Orkin SH, Dieterlen-Lievre F. Placenta as a site for hematopoietic stem cell development. Exp Hematol 2005;33:1048- 1054. 4. Punzel M, Ho AD. Divisional history and pluripotency of human hematopoietic stem cells. Ann N Y Acad Sci 2001;938:72-81. 5. Beckmann J, Scheitza S, Wernet P, Fischer JC, Giebel B. Asymmetric cell division within the human hematopoietic stem and progenitor cell compartment: identifi cation of asymmetrically segregating proteins. Blood 2007;109:5494-5501. 6. Reya T. Regulation of hematopoietic stem cell self-renewal. Recent Prog Horm Res 2003;58:283-295. 7. Bielorai B, Trakhtenbrot L, Amariglio N, Rothman R, Tabori U, Dallal I, Golan H, Neumann Y, Reichart M, Kaplinsky C, Rechavi G, Toren A. Multilineage hematopoietic engraftment after allogeneic peripheral blood stem cell transplantation without conditioning in SCID patients. Bone Marrow Transplant 2004;34:317-320. 8. Gratwohl A, Passweg J, Bocelli-Tyndall C, Fassas A, van Laar JM, Farge D, Andolina M, Arnold R, Carreras E, Finke J, Kotter I, Kozak T, Lisukov I, Lowenberg B, Marmont A, Moore J, Saccardi R, Snowden JA, van den HF, Wulffraat NM, Zhao XW, Tyndall A. Autologous hematopoietic stem cell transplantation for autoimmune diseases. Bone Marrow Transplant 2005;35:869-879. 9. Grigull L, Beilken A, Schrappe M, Das A, Luecke T, Sander A, Stanulla M, Rehe K, Sauer M, Schmid H, Welte K, Lukacs Z, Gal A, Sykora KW. Transplantation of allogeneic CD34-selected stem cells after fl udarabine- based conditioning regimen for children with mucopolysaccharidosis 1H (M. Hurler). Bone Marrow Transplant 2005;35:265-269. 10. Ng YY, van Kessel B, Lokhorst HM, Baert MR, van den Burg CM, Bloem AC, Staal FJ. Gene-expression profi ling of CD34+ cells from various hematopoietic stem-cell sources reveals functional differences in stem- cell activity. J Leukoc Biol 2004;75:314-323. 11. Nikolova T, Wu M, Brumbarov K, Alt R, Opitz H, Boheler KR, Cross M, Wobus AM. WNT-conditioned media differentially affect the proliferation and differentiation of cord blood-derived CD133+ cells in vitro. Differentiation 2007;75:100-111. 12. Chen N, Hudson JE, Walczak P, Misiuta I, Garbuzova-Davis S, Jiang L, Sanchez-Ramos J, Sanberg PR, Zigova T, Willing AE. Human umbilical cord blood progenitors: the potential of these hematopoietic cells to become neural. Stem Cells 2005;23:1560-1570. REFERENCES 63

13. Servida F, Soligo D, Caneva L, Bertolini F, de Harven E, Campiglio S, Corsini C, Deliliers GL. Functional and morphological characterization of immunomagnetically selected CD34+ hematopoietic progenitor cells. Stem Cells 1996;14:430-438. 14. McKenzie JL, Takenaka K, Gan OI, Doedens M, Dick JE. Low rhodamine 123 retention identifi es long-term human hematopoietic stem cells within the Lin-CD34+CD38- population. Blood 2007;109:543-545. 15. Hess DA, Meyerrose TE, Wirthlin L, Craft TP, Herrbrich PE, Creer MH, Nolta JA. Functional characterization of highly purifi ed human hematopoietic repopulating cells isolated according to aldehyde dehydrogenase activity. Blood 2004;104:1648-1655. 16. Kyoto Encyclopedia of Genes and Genomes Pathway Database (1 Jan. 2008) http://www.genome.ad.jp/kegg/pathway.html (11 Jan. 2008). 17. Karsunky H, Merad M, Cozzio A, Weissman IL, Manz MG. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid- committed progenitors to Flt3+ dendritic cells in vivo. J Exp Med 2003;198:305-313. 18. Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term lymphohe- matopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 1996;273:242-245. 19. Okuno Y, Iwasaki H, Huettner CS, Radomska HS, Gonzalez DA, Tenen DG, Akashi K. Differential regulation of the human and murine CD34 genes in hematopoietic stem cells. Proc Natl Acad Sci U S A 2002;99:6246-6251. 20. Spangrude GJ, Muller-Sieburg CE, Heimfeld S, Weissman IL. Two rare populations of mouse Thy-1lo bone marrow cells repopulate the thymus. J Exp Med 1988;167:1671-1683. 21. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005;121:1109-1121. 22. Gallacher L, Murdoch B, Wu DM, Karanu FN, Keeney M, Bhatia M. Isolation and characterization of human CD34(-)Lin(-) and CD34(+)Lin(- ) hematopoietic stem cells using cell surface markers AC133 and CD7. Blood 2000;95:2813-2820. 23. Gotze KS, Schiemann M, Marz S, Jacobs VR, Debus G, Peschel C, Oostendorp RA. CD133-enriched CD34(-) (CD33/CD38/CD71)(-) cord blood cells acquire CD34 prior to cell division and hematopoietic activity is exclusively associated with CD34 expression. Exp Hematol 2007;35:1408- 1414. 24. Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, Olweus J, Kearney J, Buck DW. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 1997;90:5002-5012. 25. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, Rafi i S. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifi es a population of functional endothelial precursors. Blood 2000;95:952-958. 26. Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 2004;117:3539-3545. 64 REFERENCES

27. Corbeil D, Roper K, Hellwig A, Tavian M, Miraglia S, Watt SM, Simmons PJ, Peault B, Buck DW, Huttner WB. The human AC133 hematopoietic stem cell antigen is also expressed in epithelial cells and targeted to plasma membrane protrusions. J Biol Chem 2000;275:5512-5520. 28. Kania G, Corbeil D, Fuchs J, Tarasov KV, Blyszczuk P, Huttner WB, Boheler KR, Wobus AM. Somatic stem cell marker prominin-1/CD133 is expressed in embryonic stem cell-derived progenitors. Stem Cells 2005;23:791- 804. 29. Fateh-Moghadam S, Htun P, Tomandl B, Sander D, Stellos K, Geisler T, Langer H, Walton K, Handschu R, Garlichs C, Daniel WG, Gawaz M. Hyperresponsiveness of platelets in ischemic stroke. Thromb Haemost 2007;97:974-978. 30. Dorrell C, Gan OI, Pereira DS, Hawley RG, Dick JE. Expansion of human cord blood CD34(+)CD38(-) cells in ex vivo culture during retroviral transduction without a corresponding increase in SCID repopulating cell (SRC) frequency: dissociation of SRC phenotype and function. Blood 2000;95:102-110. 31. Zhang CC, Lodish HF. Murine hematopoietic stem cells change their surface phenotype during ex vivo expansion. Blood 2005;105:4314- 4320. 32. King MR, Heinrich V, Evans E, Hammer DA. Nano-to-micro scale dynamics of P-selectin detachment from leukocyte interfaces. III. Numerical simulation of tethering under fl ow. Biophys J 2005;88:1676-1683. 33. Juopperi TA, Schuler W, Yuan X, Collector MI, Dang CV, Sharkis SJ. Isolation of bone marrow-derived stem cells using density-gradient separation. Exp Hematol 2007;35:335-341. 34. King M. Principles of cellular engineering: understanding the biomolecular interface. Burlington, MA: Elsevier Academic Press, 2005. 35. King M, Charles N, Kanofsky J, Liesveld J.L. Using protein-functionalized microchannels for stem cell separation. Proceedings of the ASME 2006;ICNMM2006-96228. 36. Kekarainen T, Mannelin S, Laine J, Jaatinen T. Optimization of immunomagnetic separation for cord blood-derived hematopoietic stem cells. BMC Cell Biol 2006;7:30. 37. Bhatia M, Wang JC, Kapp U, Bonnet D, Dick JE. Purifi cation of primitive human hematopoietic cells capable of repopulating immune-defi cient mice. Proc Natl Acad Sci U S A 1997;94:5320-5325. 38. Yahata T, Ando K, Sato T, Miyatake H, Nakamura Y, Muguruma Y, Kato S, Hotta T. A highly sensitive strategy for SCID-repopulating cell assay by direct injection of primitive human hematopoietic cells into NOD/SCID mice bone marrow. Blood 2003;101:2905-2913. 39. Wang J, Kimura T, Asada R, Harada S, Yokota S, Kawamoto Y, Fujimura Y, Tsuji T, Ikehara S, Sonoda Y. SCID-repopulating cell activity of human cord blood-derived CD34- cells assured by intra-bone marrow injection. Blood 2003;101:2924-2931. 40. Gordon PR, Leimig T, Babarin-Dorner A, Houston J, Holladay M, Mueller I, Geiger T, Handgretinger R. Large-scale isolation of CD133+ progenitor cells from G-CSF mobilized peripheral blood stem cells. Bone Marrow Transplant 2003;31:17-22. REFERENCES 65

41. Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Effi cient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science 2007;318:1296-1299. 42. Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, Arny M, Thomas L, Boyse EA. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A 1989;86:3828-3832. 43. Hao QL, Shah AJ, Thiemann FT, Smogorzewska EM, Crooks GM. A functional comparison of CD34 + CD38- cells in cord blood and bone marrow. Blood 1995;86:3745-3753. 44. Broxmeyer HE, Hangoc G, Cooper S, Ribeiro RC, Graves V, Yoder M, Wagner J, Vadhan-Raj S, Benninger L, Rubinstein P, . Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults. Proc Natl Acad Sci U S A 1992;89:4109- 4113. 45. Bonanno G, Mariotti A, Procoli A, Corallo M, Rutella S, Pessina G, Scambia G, Mancuso S, Pierelli L. Human cord blood CD133+ cells immunoselected by a clinical-grade apparatus differentiate in vitro into endothelial- and cardiomyocyte-like cells. Transfusion 2007;47:280-289. 46. Theilgaard-Monch K, Raaschou-Jensen K, Schjodt K, Heilmann C, Vindelov L, Jacobsen N, Dickmeiss E. Pluripotent and myeloid-committed CD34+ subsets in hematopoietic stem cell allografts. Bone Marrow Transplant 2003;32:1125-1133. 47. Lewis ID, Verfaillie CM. Multi-lineage expansion potential of primitive hematopoietic progenitors: superiority of umbilical cord blood compared to mobilized peripheral blood. Exp Hematol 2000;28:1087-1095. 48. van den OS, dem Borne AE, de Haas M. Differences in megakaryocyte expansion potential between CD34(+) stem cells derived from cord blood, peripheral blood, and bone marrow from adults and children. Exp Hematol 2000;28:1054-1061. 49. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 2003;425:841-846. 50. Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S, Wiedemann LM, Mishina Y, Li L. Identifi cation of the haematopoietic stem cell niche and control of the niche size. Nature 2003;425:836-841. 51. Dao MA, Creer MH, Nolta JA, Verfaillie CM. Biology of umbilical cord blood progenitors in bone marrow niches. Blood 2007;110:74-81. 52. Vaziri H, Dragowska W, Allsopp RC, Thomas TE, Harley CB, Lansdorp PM. Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc Natl Acad Sci U S A 1994;91:9857-9860. 53. Gluckman E, Broxmeyer HA, Auerbach AD, Friedman HS, Douglas GW, Devergie A, Esperou H, Thierry D, Socie G, Lehn P. Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical- cord blood from an HLA-identical sibling. N Engl J Med 1989;321:1174- 1178. 66 REFERENCES

54. Bone Marrow Donors Worldwide (24 Dec. 2007) http://www.bmdw.org (11 Jan 2008). 55. Ballen K, Broxmeyer HE, McCullough J, Piaciabello W, Rebulla P, Verfaillie CM, Wagner JE. Current status of cord blood banking and transplantation in the United States and Europe. Biol Blood Marrow Transplant 2001;7:635- 645. 56. Foundation for the Accreditation of Cellular Therapy. http://www. factwebsite.org (11 Jan 2008). 57. Fasouliotis SJ, Schenker JG. Human umbilical cord blood banking and transplantation: a state of the art. Eur J Obstet Gynecol Reprod Biol 2000;90:13-25. 58. Broxmeyer HE, Srour EF, Hangoc G, Cooper S, Anderson SA, Bodine DM. High-effi ciency recovery of functional hematopoietic progenitor and stem cells from human cord blood cryopreserved for 15 years. Proc Natl Acad Sci U S A 2003;100:645-650. 59. Mugishima H, Harada K, Chin M, Suzuki T, Takagi K, Hayakawa S, Sato K, Klein JP, Gale RP. Effects of long-term cryopreservation on hematopoietic progenitor cells in umbilical cord blood. Bone Marrow Transplant 1999;23:395-396. 60. Barker JN, Krepski TP, DeFor TE, Davies SM, Wagner JE, Weisdorf DJ. Searching for unrelated donor hematopoietic stem cells: availability and speed of umbilical cord blood versus bone marrow. Biol Blood Marrow Transplant 2002;8:257-260. 61. Burroughs LM, Storb R, Leisenring WM, Pulsipher MA, Loken MR, Torgerson TR, Ochs HD, Woolfrey AE. Intensive postgrafting immune suppression combined with nonmyeloablative conditioning for transplantation of HLA- identical hematopoietic cell grafts: results of a pilot study for treatment of primary immunodefi ciency disorders. Bone Marrow Transplant 2007;40:633-642. 62. Ball LM, Bernardo ME, Roelofs H, Lankester A, Cometa A, Egeler RM, Locatelli F, Fibbe WE. Cotransplantation of ex vivo expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 2007;110:2764-2767. 63. Fernandez MN, Regidor C, Cabrera R, Garcia-Marco JA, Fores R, Sanjuan I, Gayoso J, Gil S, Ruiz E, Little AM, McWhinnie A, Madrigal A. Unrelated umbilical cord blood transplants in adults: Early recovery of neutrophils by supportive co-transplantation of a low number of highly purifi ed peripheral blood CD34+ cells from an HLA-haploidentical donor. Exp Hematol 2003;31:535-544. 64. Han JY, Goh RY, Seo SY, Hwang TH, Kwon HC, Kim SH, Kim JS, Kim HJ, Lee YH. Cotransplantation of cord blood hematopoietic stem cells and culture-expanded and GM-CSF-/SCF-transfected mesenchymal stem cells in SCID mice. J Korean Med Sci 2007;22:242-247. 65. Kogler G, Nurnberger W, Fischer J, Niehues T, Somville T, Gobel U, Wernet P. Simultaneous cord blood transplantation of ex vivo expanded together with non-expanded cells for high risk leukemia. Bone Marrow Transplant 1999;24:397-403. REFERENCES 67

66. Pecora AL, Stiff P, Jennis A, Goldberg S, Rosenbluth R, Price P, Goltry KL, Douville J, Armstrong RD, Smith AK, Preti RA. Prompt and durable engraftment in two older adult patients with high risk chronic myelogenous leukemia (CML) using ex vivo expanded and unmanipulated unrelated umbilical cord blood. Bone Marrow Transplant 2000;25:797-799. 67. Janowska-Wieczorek A, Majka M, Kijowski J, Baj-Krzyworzeka M, Reca R, Turner AR, Ratajczak J, Emerson SG, Kowalska MA, Ratajczak MZ. Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment. Blood 2001;98:3143-3149. 68. Kohn DB, Hershfi eld MS, Carbonaro D, Shigeoka A, Brooks J, Smogorzewska EM, Barsky LW, Chan R, Burotto F, Annett G, Nolta JA, Crooks G, Kapoor N, Elder M, Wara D, Bowen T, Madsen E, Snyder FF, Bastian J, Muul L, Blaese RM, Weinberg K, Parkman R. T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-defi cient SCID neonates. Nat Med 1998;4:775-780. 69. Dalle JH, Duval M, Moghrabi A, Wagner E, Vachon MF, Barrette S, Bernstein M, Champagne J, David M, Demers J, Rousseau P, Winikoff R, Champagne MA. Results of an unrelated transplant search strategy using partially HLA-mismatched cord blood as an immediate alternative to HLA- matched bone marrow. Bone Marrow Transplant 2004;33:605-611. 70. Gluckman E, Rocha V, Boyer-Chammard A, Locatelli F, Arcese W, Pasquini R, Ortega J, Souillet G, Ferreira E, Laporte JP, Fernandez M, Chastang C. Outcome of cord-blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med 1997;337:373-381. 71. Rubinstein P, Carrier C, Scaradavou A, Kurtzberg J, Adamson J, Migliaccio AR, Berkowitz RL, Cabbad M, Dobrila NL, Taylor PE, Rosenfi eld RE, Stevens CE. Outcomes among 562 recipients of placental-blood transplants from unrelated donors. N Engl J Med 1998;339:1565-1577. 72. Eapen M, Rubinstein P, Zhang MJ, Stevens C, Kurtzberg J, Scaradavou A, Loberiza FR, Champlin RE, Klein JP, Horowitz MM, Wagner JE. Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study. Lancet 2007;369:1947-1954. 73. Encabo A, Solves P, Carbonell-Uberos F, Minana MD. The functional immaturity of dendritic cells can be relevant to increased tolerance associated with cord blood transplantation. Transfusion 2007;47:272- 279. 74. Gluckman E, Rocha V. Cord blood transplantation for children with acute leukaemia: a Eurocord registry analysis. Blood Cells Mol Dis 2004;33:271- 273. 75. Takahashi S, Iseki T, Ooi J, Tomonari A, Takasugi K, Shimohakamada Y, Yamada T, Uchimaru K, Tojo A, Shirafuji N, Kodo H, Tani K, Takahashi T, Yamaguchi T, Asano S. Single-institute comparative analysis of unrelated bone marrow transplantation and cord blood transplantation for adult patients with hematologic malignancies. Blood 2004;104:3813-3820. 68 REFERENCES

76. Kleen TO, Kadereit S, Fanning LR, Jaroscak J, Fu P, Meyerson HJ, Kulchycki L, Slivka LF, Kozik M, Tary-Lehmann M, Laughlin MJ. Recipient- specifi c tolerance after HLA-mismatched umbilical cord blood stem cell transplantation. Transplantation 2005;80:1316-1322. 77. Lekakis L, Giralt S, Couriel D, Shpall EJ, Hosing C, Khouri IF, Anderlini P, Korbling M, Martin T, Champlin RE, de Lima M. Phase II study of unrelated cord blood transplantation for adults with high-risk hematologic malignancies. Bone Marrow Transplant 2006;38:421-426. 78. Barker JN, Weisdorf DJ, DeFor TE, Blazar BR, Miller JS, Wagner JE. Rapid and complete donor chimerism in adult recipients of unrelated donor umbilical cord blood transplantation after reduced-intensity conditioning. Blood 2003;102:1915-1919. 79. Takizawa J, Aoki S, Kurasaki T, Higashimura M, Honma K, Kitajima T, Momoi A, Takahashi H, Nakamura N, Furukawa T, Aizawa Y. Successful treatment of adult T-cell leukemia with unrelated cord blood transplantation. Am J Hematol 2007;82:1113-1115. 80. Nakagawa M, Hashino S, Takahata M, Kawamura T, Fujisawa F, Kahata K, Kondo T, Imamura M, Ando S, Asaka M. Successful reduced-intensity stem cell transplantation with cord blood for a poor-prognosis adult with refractory chronic active epstein-barr virus infection. Int J Hematol 2007;85:443-445. 81. Rodriguez R, Nademanee A, Ruel N, Smith E, Krishnan A, Popplewell L, Zain J, Patane K, Kogut N, Nakamura R, Sarkodee-Adoo C, Forman SJ. Comparison of reduced-intensity and conventional myeloablative regimens for allogeneic transplantation in non-Hodgkin’s lymphoma. Biol Blood Marrow Transplant 2006;12:1326-1334. 82. Nauta AJ, Kruisselbrink AB, Lurvink E, Mulder A, Claas FH, Noort WA, Willemze R, Fibbe WE. Enhanced engraftment of umbilical cord blood- derived stem cells in NOD/SCID mice by cotransplantation of a second unrelated cord blood unit. Exp Hematol 2005;33:1249-1256. 83. Majhail NS, Brunstein CG, Wagner JE. Double umbilical cord blood transplantation. Curr Opin Immunol 2006;18:571-575. 84. Cord Blood Center. http://www.cordbloodcenter.com (11 Jan. 2008). 85. Talvensaari K, Clave E, Douay C, Rabian C, Garderet L, Busson M, Garnier F, Douek D, Gluckman E, Charron D, Toubert A. A broad T-cell repertoire diversity and an effi cient thymic function indicate a favorable long-term immune reconstitution after cord blood stem cell transplantation. Blood 2002;99:1458-1464. 86. Giraud P, Thuret I, Reviron D, Chambost H, Brunet C, Novakovitch G, Farnarier C, Michel G. Immune reconstitution and outcome after unrelated cord blood transplantation: a single paediatric institution experience. Bone Marrow Transplant 2000;25:53-57. 87. van Burik JA, Brunstein CG. Infectious complications following unrelated cord blood transplantation. Vox Sang 2007;92:289-296. 88. Verhasselt B, Kerre T, Naessens E, Vanhecke D, De Smedt M, Vandekerckhove B, Plum J. Thymic repopulation by CD34(+) human cord blood cells after expansion in stroma-free culture. Blood 1999;94:3644- 3652. REFERENCES 69

89. Jetmore A, Plett PA, Tong X, Wolber FM, Breese R, Abonour R, Orschell- Traycoff CM, Srour EF. Homing effi ciency, cell cycle kinetics, and survival of quiescent and cycling human CD34(+) cells transplanted into conditioned NOD/SCID recipients. Blood 2002;99:1585-1593. 90. Gothot A, van der Loo JC, Clapp DW, Srour EF. Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34(+) cells in non-obese diabetic/severe combined immune-defi cient mice. Blood 1998;92:2641-2649. 91. Glimm H, Oh IH, Eaves CJ. Human hematopoietic stem cells stimulated to proliferate in vitro lose engraftment potential during their S/G(2)/M transit and do not reenter G(0). Blood 2000;96:4185-4193. 92. Beslu N, Krosl J, Laurin M, Mayotte N, Humphries KR, Sauvageau G. Molecular interactions involved in HOXB4-induced activation of HSC self- renewal. Blood 2004;104:2307-2314. 93. Zhang XB, Schwartz JL, Humphries RK, Kiem HP. Effects of HOXB4 overexpression on ex vivo expansion and immortalization of hematopoietic cells from different species. Stem Cells 2007;25:2074-2081. 94. Schofi eld KP, Rushton G, Humphries MJ, Dexter TM, Gallagher JT. Infl uence of interleukin-3 and other growth factors on alpha4beta1 integrin- mediated adhesion and migration of human hematopoietic progenitor cells. Blood 1997;90:1858-1866. 95. Nie L, Wu G, Zhang W. Correlation of mRNA expression and protein abundance affected by multiple sequence features related to translational effi ciency in Desulfovibrio vulgaris: a quantitative analysis. Genetics 2006;174:2229-2243. 96. Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, Nishida H, Yap CC, Suzuki M, Kawai J, Suzuki H, Carninci P, Hayashizaki Y, Wells C, Frith M, Ravasi T, Pang KC, Hallinan J, Mattick J, Hume DA, Lipovich L, Batalov S, Engstrom PG, Mizuno Y, Faghihi MA, Sandelin A, Chalk AM, Mottagui-Tabar S, Liang Z, Lenhard B, Wahlestedt C. Antisense transcription in the mammalian transcriptome. Science 2005;309:1564- 1566. 97. Nie L, Wu G, Zhang W. Correlation between mRNA and protein abundance in Desulfovibrio vulgaris: a multiple regression to identify sources of variations. Biochem Biophys Res Commun 2006;339:603-610. 98. Liu F, Lu J, Fan HH, Wang ZQ, Cui SJ, Zhang GA, Chi M, Zhang X, Yang PY, Chen Z, Han ZG. Insights into human CD34+ hematopoietic stem/ progenitor cells through a systematically proteomic survey coupled with transcriptome. Proteomics 2006;6:2673-2692. 99. Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science 1995;20;270:467-470. 100. Affymetrix Inc U. GeneChip Expression Analysis Technical Manual. http:// www.affymetrix.com/support/downloads/manuals/expression_analysis_ manual.pdf (11 Jan. 2008). 101. Broxmeyer HE. Cord Blood Biology, Immunology, Banking, and Clinical Transplantation. 2004 2004;AABB Press, Maryland:1-455. 102. Serial Analysis of Gene Expression. http://www.sagenet.org (11 Jan. 2008). 70 REFERENCES

103. Lonneborg A, Sharma P, Stougaard P. Construction of subtractive cDNA library using magnetic beads and PCR. PCR Methods Appl 1995;4:168- 176. 104. van Ruissen F, Ruijter JM, Schaaf GJ, Asgharnegad L, Zwijnenburg DA, Kool M, Baas F. Evaluation of the similarity of gene expression data estimated with SAGE and Affymetrix GeneChips. BMC Genomics 2005;6:91 doi:10.1186/1471-2164-6-91. 105. Yuen T, Wurmbach E, Pfeffer RL, Ebersole BJ, Sealfon SC. Accuracy and calibration of commercial oligonucleotide and custom cDNA microarrays. Nucleic Acids Res 2002;30:e48. 106. Phillips RL, Ernst RE, Brunk B, Ivanova N, Mahan MA, Deanehan JK, Moore KA, Overton GC, Lemischka IR. The genetic program of hematopoietic stem cells. Science 2000;288:1635-1640. 107. Park IK, He Y, Lin F, Laerum OD, Tian Q, Bumgarner R, Klug CA, Li K, Kuhr C, Doyle MJ, Xie T, Schummer M, Sun Y, Goldsmith A, Clarke MF, Weissman IL, Hood L, Li L. Differential gene expression profi ling of adult murine hematopoietic stem cells. Blood 2002;99:488-498. 108. Terskikh AV, Easterday MC, Li L, Hood L, Kornblum HI, Geschwind DH, Weissman IL. From hematopoiesis to neuropoiesis: evidence of overlapping genetic programs. Proc Natl Acad Sci U S A 2001;98:7934- 7939. 109. Ivanova NB, Dimos JT, Schaniel C, Hackney JA, Moore KA, Lemischka IR. A stem cell molecular signature. Science 2002;298:601-604. 110. Morrison SJ, Hemmati HD, Wandycz AM, Weissman IL. The purifi cation and characterization of fetal liver hematopoietic stem cells. Proc Natl Acad Sci U S A 1995;92:10302-10306. 111. Harrison DE, Astle CM. Short- and long-term multilineage repopulating hematopoietic stem cells in late fetal and newborn mice: models for human umbilical cord blood. Blood 1997;90:174-181. 112. Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan RC, Melton DA. “Stemness”: transcriptional profi ling of embryonic and adult stem cells. Science 2002;298:597-600. 113. Akashi K, He X, Chen J, Iwasaki H, Niu C, Steenhard B, Zhang J, Haug J, Li L. Transcriptional accessibility for genes of multiple tissues and hematopoietic lineages is hierarchically controlled during early hematopoiesis. Blood 2003;101:383-389. 114. Xi R, Xie T. Stem cell self-renewal controlled by chromatin remodeling factors. Science 2005;310:1487-1489. 115. Georgantas RW, III, Tanadve V, Malehorn M, Heimfeld S, Chen C, Carr L, Martinez-Murillo F, Riggins G, Kowalski J, Civin CI. Microarray and serial analysis of gene expression analyses identify known and novel transcripts overexpressed in hematopoietic stem cells. Cancer Res 2004;64:4434- 4441. 116. Steidl U, Kronenwett R, Rohr UP, Fenk R, Kliszewski S, Maercker C, Neubert P, Aivado M, Koch J, Modlich O, Bojar H, Gattermann N, Haas R. Gene expression profi ling identifi es signifi cant differences between the molecular phenotypes of bone marrow-derived and circulating human CD34+ hematopoietic stem cells. Blood 2002;99:2037-2044. REFERENCES 71

117. Attema JL, Papathanasiou P, Forsberg EC, Xu J, Smale ST, Weissman IL. Epigenetic characterization of hematopoietic stem cell differentiation using miniChIP and bisulfi te sequencing analysis. Proc Natl Acad Sci U S A 2007;104:12371-12376. 118. Spivakov M, Fisher AG. Epigenetic signatures of stem-cell identity. Nat Rev Genet 2007;8:263-271. 119. Baal N, Reisinger K, Jahr H, Bohle RM, Liang O, Munstedt K, Rao CV, Preissner KT, Zygmunt MT. Expression of transcription factor Oct-4 and other embryonic genes in CD133 positive cells from human umbilical cord blood. Thromb Haemost 2004;92:767-775. 120. Wagner W, Ansorge A, Wirkner U, Eckstein V, Schwager C, Blake J, Miesala K, Selig J, Saffrich R, Ansorge W, Ho AD. Molecular evidence for stem cell function of the slow-dividing fraction among human hematopoietic progenitor cells by genome-wide analysis. Blood 2004;104:675-686. 121. Parekh RB, Tse AG, Dwek RA, Williams AF, Rademacher TW. Tissue- specifi c N-glycosylation, site-specifi c oligosaccharide patterns and lentil lectin recognition of rat Thy-1. EMBO J 1987;6:1233-1244. 122. King MJ. Blood group antigens on human erythrocytes-distribution, structure and possible functions. Biochim Biophys Acta 1994;1197:15- 44. 123. Freeze HH. Genetic defects in the human glycome. Nat Rev Genet 2006;7:537-551. 124. Lowe JB. Glycosylation in the control of selectin counter-receptor structure and function. Immunol Rev 2002;186:19-36. 125. Tulp A, Barnhoorn M, Bause E, Ploegh H. Inhibition of N-linked oligosaccharide trimming mannosidases blocks human B cell development. EMBO J 1986;5:1783-1790. 126. Lowe JB, Marth JD. A genetic approach to Mammalian glycan function. Annu Rev Biochem 2003;72:643-691. 127. Kornfeld R, Kornfeld S. Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem 1985;54:631-664. 128. Metzler M, Gertz A, Sarkar M, Schachter H, Schrader JW, Marth JD. Complex asparagine-linked oligosaccharides are required for morphogenic events during post-implantation development. EMBO J 1994;13:2056-2065. 129. Chui D, Oh-Eda M, Liao YF, Panneerselvam K, Lal A, Marek KW, Freeze HH, Moremen KW, Fukuda MN, Marth JD. Alpha-mannosidase-II defi ciency results in dyserythropoiesis and unveils an alternate pathway in oligosaccharide biosynthesis. Cell 1997;90:157-167. 130. Wang Y, Tan J, Sutton-Smith M, Ditto D, Panico M, Campbell RM, Varki NM, Long JM, Jaeken J, Levinson SR, Wynshaw-Boris A, Morris HR, Le D, Dell A, Schachter H, Marth JD. Modeling human congenital disorder of glycosylation type IIa in the mouse: conservation of asparagine-linked glycan-dependent functions in mammalian physiology and insights into disease pathogenesis. Glycobiology 2001;11:1051-1070. 131. Thorens B. A toggle for type 2 diabetes? N Engl J Med 2006;354:1636- 1638. 132. Granovsky M, Fata J, Pawling J, Muller WJ, Khokha R, Dennis JW. Suppression of tumor growth and metastasis in Mgat5-defi cient mice. Nat Med 2000;6:306-312. 72 REFERENCES

133. Dennis JW, Granovsky M, Warren CE. Glycoprotein glycosylation and cancer progression. Biochim Biophys Acta 1999;1473:21-34. 134. Asano M, Nakae S, Kotani N, Shirafuji N, Nambu A, Hashimoto N, Kawashima H, Hirose M, Miyasaka M, Takasaki S, Iwakura Y. Impaired selectin-ligand biosynthesis and reduced infl ammatory responses in beta- 1,4-galactosyltransferase-I-defi cient mice. Blood 2003;102:1678-1685. 135. Hansske B, Thiel C, Lubke T, Hasilik M, Honing S, Peters V, Heidemann PH, Hoffmann GF, Berger EG, von Figura K, Korner C. Defi ciency of UDP-galactose:N-acetylglucosamine beta-1,4-galactosyltransferase I causes the congenital disorder of glycosylation type IId. J Clin Invest 2002;109:725-733. 136. Peters V, Penzien JM, Reiter G, Korner C, Hackler R, Assmann B, Fang J, Schaefer JR, Hoffmann GF, Heidemann PH. Congenital disorder of glycosylation IId (CDG-IId) -- a new entity: clinical presentation with Dandy-Walker malformation and myopathy. Neuropediatrics 2002;33:27- 32. 137. Hennet T, Chui D, Paulson JC, Marth JD. Immune regulation by the ST6Gal sialyltransferase. Proc Natl Acad Sci U S A 1998;95:4504-4509. 138. Ellies LG, Sperandio M, Underhill GH, Yousif J, Smith M, Priatel JJ, Kansas GS, Ley K, Marth JD. Sialyltransferase specifi city in selectin ligand formation. Blood 2002;100:3618-3625. 139. Ellies LG, Sperandio M, Underhill GH, Yousif J, Smith M, Priatel JJ, Kansas GS, Ley K, Marth JD. Sialyltransferase specifi city in selectin ligand formation. Blood 2002;100:3618-3625. 140. Fukumoto S, Miyazaki H, Goto G, Urano T, Furukawa K, Furukawa K. Expression cloning of mouse cDNA of CMP-NeuAc:Lactosylceramide alpha2,3-sialyltransferase, an enzyme that initiates the synthesis of gangliosides. J Biol Chem 1999;274:9271-9276. 141. Wang X, Inoue S, Gu J, Miyoshi E, Noda K, Li W, Mizuno-Horikawa Y, Nakano M, Asahi M, Takahashi M, Uozumi N, Ihara S, Lee SH, Ikeda Y, Yamaguchi Y, Aze Y, Tomiyama Y, Fujii J, Suzuki K, Kondo A, Shapiro SD, Lopez-Otin C, Kuwaki T, Okabe M, Honke K, Taniguchi N. Dysregulation of TGF-beta1 receptor activation leads to abnormal lung development and emphysema-like phenotype in core fucose-defi cient mice. Proc Natl Acad Sci U S A 2005;102:15791-15796. 142. Kudo T, Kaneko M, Iwasaki H, Togayachi A, Nishihara S, Abe K, Narimatsu H. Normal embryonic and germ cell development in mice lacking alpha 1,3-fucosyltransferase IX (Fut9) which show disappearance of stage- specifi c embryonic antigen 1. Mol Cell Biol 2004;24:4221-4228. 143. Domino SE, Zhang L, Gillespie PJ, Saunders TL, Lowe JB. Defi ciency of reproductive tract alpha(1,2)fucosylated glycans and normal fertility in mice with targeted deletions of the FUT1 or FUT2 alpha(1,2)fucosyltrans ferase . Mol Cell Biol 2001;21:8336-8345. 144. Homeister JW, Thall AD, Petryniak B, Maly P, Rogers CE, Smith PL, Kelly RJ, Gersten KM, Askari SW, Cheng G, Smithson G, Marks RM, Misra AK, Hindsgaul O, von Andrian UH, Lowe JB. The alpha(1,3)fucosyltransferases FucT-IV and FucT-VII exert collaborative control over selectin-dependent leukocyte recruitment and lymphocyte homing. Immunity 2001;15:115- 126. REFERENCES 73

145. Maly P, Thall A, Petryniak B, Rogers CE, Smith PL, Marks RM, Kelly RJ, Gersten KM, Cheng G, Saunders TL, Camper SA, Camphausen RT, Sullivan FX, Isogai Y, Hindsgaul O, von Andrian UH, Lowe JB. The alpha(1,3)fucos yltransferase Fuc-TVII controls leukocyte traffi cking through an essential role in L-, E-, and P-selectin ligand biosynthesis. Cell 1996;86:643-653. 146. Bengtson P, Larson C, Lundblad A, Larson G, Pahlsson P. Identifi cation of a missense mutation (G329A;Arg(110)--> GLN) in the human FUT7 gene. J Biol Chem 2001;276:31575-31582. 147. Galili U, Shohet SB, Kobrin E, Stults CL, Macher BA. Man, apes, and Old World monkeys differ from other mammals in the expression of alpha- galactosyl epitopes on nucleated cells. J Biol Chem 1988;263:17755- 17762. 148. Galili U, Clark MR, Shohet SB, Buehler J, Macher BA. Evolutionary relationship between the natural anti-Gal antibody and the Gal alpha 1----3 Gal epitope in primates. Proc Natl Acad Sci U S A 1987;84:1369- 1373. 149. Muchmore EA, Diaz S, Varki A. A structural difference between the cell surfaces of humans and the great apes. Am J Phys Anthropol 1998;107:187-198. 150. Heiskanen A, Satomaa T, Tiitinen S, Laitinen A, Mannelin S, Impola U, Mikkola M, Olsson C, Miller-Podraza H, Blomqvist M, Olonen A, Salo H, Lehenkari P, Tuuri T, Otonkoski T, Natunen J, Saarinen J, Laine J. N- glycolylneuraminic acid xenoantigen contamination of human embryonic and mesenchymal stem cells is substantially reversible. Stem Cells 2007;25:197-202. 151. Skottman H, Narkilahti S, Hovatta O. Challenges and approaches to the culture of pluripotent human embryonic stem cells. Regen Med 2007;2:265-273. 152. Fukuda M, Fukuda MN. Changes in cell surface glycoproteins and structures during the development and differentiation of human erythroid cells. J Supramol Struct Cell Biochem 1981;17:313- 324. 153. Rinder HM, Bonan JL, Rinder CS, Ault KA, Smith BR. Activated and unactivated platelet adhesion to monocytes and neutrophils. Blood 1991;78:1760-1769. 154. Polley MJ, Phillips ML, Wayner E, Nudelman E, Singhal AK, Hakomori S, Paulson JC. CD62 and endothelial cell-leukocyte adhesion molecule 1 (ELAM-1) recognize the same carbohydrate ligand, sialyl-Lewis x. Proc Natl Acad Sci U S A 1991;88:6224-6228. 155. Le Marer N, Skacel PO. Up-regulation of alpha2,6 sialylation during myeloid maturation: a potential role in myeloid cell release from the bone marrow. J Cell Physiol 1999;179:315-324. 156. Schwartz-Albiez R, Merling A, Martin S, Haas R, Gross HJ. Cell surface sialylation and ecto-sialyltransferase activity of human CD34 progenitors from peripheral blood and bone marrow. Glycoconj J 2004;21:451-459. 157. Hui N, Le Marer N. Alpha-2,6-sialylation regulation in CD34+ progenitor cells in the human bone marrow and granulocyte colony-stimulating factor mobilization. J Hematother Stem Cell Res 2001;10:661-668. 74 REFERENCES

158. Crean SM, Meneski JP, Hullinger TG, Reilly MJ, DeBoever EH, Taichman RS. N-linked sialyated sugar receptors support haematopoietic cell- osteoblast adhesions. Br J Haematol 2004;124:534-546. 159. Mitoma J, Bao X, Petryanik B, Schaerli P, Gauguet JM, Yu SY, Kawashima H, Saito H, Ohtsubo K, Marth JD, Khoo KH, von Andrian UH, Lowe JB, Fukuda M. Critical functions of N-glycans in L-selectin-mediated lymphocyte homing and recruitment. Nat Immunol 2007;8:409-418. 160. Goetz DJ, Greif DM, Ding H, Camphausen RT, Howes S, Comess KM, Snapp KR, Kansas GS, Luscinskas FW. Isolated P-selectin glycoprotein ligand-1 dynamic adhesion to P- and E-selectin. J Cell Biol 1997;137:509-519. 161. Spertini O, Cordey AS, Monai N, Giuffre L, Schapira M. P-selectin glycoprotein ligand 1 is a ligand for L-selectin on neutrophils, monocytes, and CD34+ hematopoietic progenitor cells. J Cell Biol 1996;135:523- 531. 162. Li F, Wilkins PP, Crawley S, Weinstein J, Cummings RD, McEver RP. Post- translational modifi cations of recombinant P-selectin glycoprotein ligand- 1 required for binding to P- and E-selectin. J Biol Chem 1996;271:3255- 3264. 163. Hidalgo A, Frenette PS. Enforced fucosylation of neonatal CD34+ cells generates selectin ligands that enhance the initial interactions with microvessels but not homing to bone marrow. Blood 2005;105:567- 575. 164. Xia L, McDaniel JM, Yago T, Doeden A, McEver RP. Surface fucosylation of human cord blood cells augments binding to P-selectin and E-selectin and enhances engraftment in bone marrow. Blood 2004;104:3091-3096. 165. Dimitroff CJ, Lee JY, Fuhlbrigge RC, Sackstein R. A distinct glycoform of CD44 is an L-selectin ligand on human hematopoietic cells. Proc Natl Acad Sci U S A 2000;97:13841-13846. 166. Dimitroff CJ, Lee JY, Rafi i S, Fuhlbrigge RC, Sackstein R. CD44 is a major E-selectin ligand on human hematopoietic progenitor cells. J Cell Biol 2001;153:1277-1286. 167. Sackstein R, Dimitroff CJ. A hematopoietic cell L-selectin ligand that is distinct from PSGL-1 and displays N-glycan-dependent binding activity. Blood 2000;96:2765-2774. 168. Dimitroff CJ, Lee JY, Schor KS, Sandmaier BM, Sackstein R. differential L-selectin binding activities of human hematopoietic cell L-selectin ligands, HCELL and PSGL-1. J Biol Chem 2001;276:47623-47631. 169. Wright C, Haddad F, Qin AX, Baldwin KM. Analysis of myosin heavy chain mRNA expression by RT-PCR. J Appl Physiol 1997;83:1389-1396. 170. Eisen MB, Spellman PT, Brown PO, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A 1998;95:14863-14868. 171. Hautaniemi S, Yli-Harja O, Astola J, Kauraniemi P, Kallioniemi A, Wolf M, Ruiz J, Mousses S, Kallioniemi OP. Analysis and visualization of gene expression microarray data in human cancer using self-organizing maps. Machine Learning 2003;52:45-66. 172. Aoki K, Perlman M, Lim JM, Cantu R, Wells L, Tiemeyer M. Dynamic developmental elaboration of N-linked glycan complexity in the Drosophila melanogaster embryo. J Biol Chem 2007;282:9127-9142. REFERENCES 75

173. Nyman TA, Kalkkinen N, Tolo H, Helin J. Structural characterisation of N- linked and O-linked oligosaccharides derived from interferon-alpha2b and interferon-alpha14c produced by Sendai-virus-induced human peripheral blood leukocytes. Eur J Biochem 1998;253:485-493. 174. Saarinen J, Welgus HG, Flizar CA, Kalkkinen N, Helin J. N-glycan structures of matrix metalloproteinase-1 derived from human fi broblasts and from HT-1080 fi brosarcoma cells. Eur J Biochem 1999;259:829-840. 175. Weikkolainen K, Aitio O, Blomqvist M, Natunen J, Helin J. Conjugation of oligosaccharides by reductive amination to amine modifi ed chondroitin oligomer and gamma-cyclodextrin. Glycoconj J 2007;24:157-165. 176. Zou J, Ichikawa H, Blackburn ML, Hu HM, Zielinska-Kwiatkowska A, Mei Q, Roth GJ, Chansky HA, Yang L. The oncogenic TLS-ERG fusion protein exerts different effects in hematopoietic cells and fi broblasts. Mol Cell Biol 2005;25:6235-6246. 177. Porras F, Urrea F, Ortiz B, Martinez-Cairo S, Bouquelet S, Martinez G, Lascurain R, Zenteno E. Isolation of the receptor for the Amaranthus leucocarpus lectin from human T lymphocytes. Biochim Biophys Acta 2005;20:155-162. 178. Kaartinen T, Lappalainen J, Haimila K, Autero M, Partanen J. Genetic variation in ICOS regulates mRNA levels of ICOS and splicing isoforms of CTLA4. Mol Immunol 2007;44:1644-1651. 179. Qiagen. RNeasy Mini Handbook. http://www1.qiagen.com/HB/ RNeasyMiniKit_EN (11 Jan. 2008). 180. Nystedt J, Makinen S, Laine J, Jolkkonen J. Human cord blood CD34+ cells and behavioral recovery following focal cerebral ischemia in rats. Acta Neurobiol Exp (Wars ) 2006;66:293-300. 181. Dai M, Wang P, Boyd AD, Kostov G, Athey B, Jones EG, Bunney WE, Myers RM, Speed TP, Akil H, Watson SJ, Meng F. Evolving gene/transcript defi nitions signifi cantly alter the interpretation of GeneChip data. Nucleic Acids Res 2005;33:e175- 182. Wu Z, Irizarry RA, Gentleman R, Martinez-Murillo F, Spencer F. A model- based background adjustment for oligonucleotide expression arrays. J Am Stat Assoc 2004;1-26. 183. GeneSifter. http://www.genesifter.net/web/ (11 Jan. 2008). 184. Chalifa-Caspi V, Shmueli O, Benjamin-Rodrig H, Rosen N, Shmoish M, Yanai I, Ophir R, Kats P, Safran M, Lancet D. GeneAnnot: interfacing GeneCards with high-throughput gene expression compendia. Brief Bioinform 2003;4:349-360. 185. Chalifa-Caspi V, Yanai I, Ophir R, Rosen N, Shmoish M, Benjamin-Rodrig H, Shklar M, Stein TI, Shmueli O, Safran M, Lancet D. GeneAnnot: comprehensive two-way linking between oligonucleotide array probesets and GeneCards genes. Bioinformatics 2004;20:1457-1458. 186. Dennis G, Jr., Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 2003;4:P3. 187. Baldus CD, Tanner SM, Kusewitt DF, Liyanarachchi S, Choi C, Caligiuri MA, Bloomfi eld CD, de La CA. BAALC, a novel marker of human hematopoietic progenitor cells. Exp Hematol 2003;31:1051-1056. 76 REFERENCES

188. He X, Gonzalez V, Tsang A, Thompson J, Tsang TC, Harris DT. Differential gene expression profi ling of CD34+ CD133+ umbilical cord blood hematopoietic stem progenitor cells. Stem Cells Dev 2005;14:188-198. 189. Liu X, Rapp N, Deans R, Cheng L. Molecular cloning and chromosomal mapping of a candidate cytokine gene selectively expressed in human CD34+ cells. Genomics 2000;65:283-292. 190. Tanner SM, Austin JL, Leone G, Rush LJ, Plass C, Heinonen K, Mrozek K, Sill H, Knuutila S, Kolitz JE, Archer KJ, Caligiuri MA, Bloomfi eld CD, de La CA. BAALC, the human member of a novel mammalian neuroectoderm gene lineage, is implicated in hematopoiesis and acute leukemia. Proc Natl Acad Sci U S A 2001;98:13901-13906. 191. Briegel K, Lim KC, Plank C, Beug H, Engel JD, Zenke M. Ectopic expression of a conditional GATA-2/estrogen receptor chimera arrests erythroid differentiation in a hormone-dependent manner. Genes Dev 1993;7:1097- 1109. 192. Stanton BR, Parada LF. The N-myc proto-oncogene: developmental expression and in vivo site-directed mutagenesis. Brain Pathol 1992;2:71- 83. 193. Toren A, Bielorai B, Jacob-Hirsch J, Fisher T, Kreiser D, Moran O, Zeligson S, Givol D, Yitzhaky A, Itskovitz-Eldor J, Kventsel I, Rosenthal E, Amariglio N, Rechavi G. CD133-positive hematopoietic stem cell “stemness” genes contain many genes mutated or abnormally expressed in leukemia. Stem Cells 2005;23:1142-1153. 194. Mohrmann L, Verrijzer CP. Composition and functional specifi city of SWI2/SNF2 class chromatin remodeling complexes. Biochim Biophys Acta 2005;1681:59-73. 195. Sif S, Saurin AJ, Imbalzano AN, Kingston RE. Purifi cation and characterization of mSin3A-containing Brg1 and hBrm chromatin remodeling complexes. Genes Dev 2001;15:603-618. 196. De Bruyn C, Delforge A, Bron D, Bernier M, Massy M, Ley P, de Hemptinne D, Stryckmans P. Comparison of the coexpression of CD38, CD33 and HLA-DR antigens on CD34+ purifi ed cells from human cord blood and bone marrow. Stem Cells 1995;13:281-288. 197. Frassoni F, Podesta M, Maccario R, Giorgiani G, Rossi G, Zecca M, Bacigalupo A, Piaggio G, Locatelli F. Cord blood transplantation provides better reconstitution of hematopoietic reservoir compared with bone marrow transplantation. Blood 2003;102:1138-1141. 198. Ueda T, Yoshida M, Yoshino H, Kobayashi K, Kawahata M, Ebihara Y, Ito M, Asano S, Nakahata T, Tsuji K. Hematopoietic capability of CD34+ cord blood cells: a comparison with CD34+ adult bone marrow cells. Int J Hematol 2001;73:457-462. 199. Passegue E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med 2005;202:1599- 1611. 200. Ambiru S, Miyazaki M, Sasada K, Ito H, Kimura F, Nakagawa K, Shimizu H, Ando K, Nakajima N. Effects of perioperative protease inhibitor on infl ammatory cytokines and acute-phase proteins in patients with hepatic resection. Dig Surg 2000;17:337-343. REFERENCES 77

201. Numanami H, Koyama S, Nelson DK, Hoyt JC, Freels JL, Habib MP, Amano J, Haniuda M, Sato E, Robbins RA. Serine protease inhibitors modulate smoke-induced chemokine release from human lung fi broblasts. Am J Respir Cell Mol Biol 2003;29:613-619. 202. Harbig J, Sprinkle R, Enkemann SA. A sequence-based identifi cation of the genes detected by probesets on the Affymetrix U133 plus 2.0 array. Nucleic Acids Res 2005;33:e31. 203. Rocha V, Wagner JE, Jr., Sobocinski KA, Klein JP, Zhang MJ, Horowitz MM, Gluckman E. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources. N Engl J Med 2000;342:1846-1854. 204. Peled A, Kollet O, Ponomaryov T, Petit I, Franitza S, Grabovsky V, Slav MM, Nagler A, Lider O, Alon R, Zipori D, Lapidot T. The chemokine SDF- 1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood 2000;95:3289-3296. 205. Lambert JF, Liu M, Colvin GA, Dooner M, McAuliffe CI, Becker PS, Forget BG, Weissman SM, Quesenberry PJ. Marrow stem cells shift gene expression and engraftment phenotype with cell cycle transit. J Exp Med 2003;197:1563-1572. 206. Skottman H, Stromberg AM, Matilainen E, Inzunza J, Hovatta O, Lahesmaa R. Unique gene expression signature by human embryonic stem cells cultured under serum-free conditions correlates with their enhanced and prolonged growth in an undifferentiated stage. Stem Cells 2006;24:151- 167. 207. Tremblay LO, Herscovics A. Characterization of a cDNA encoding a novel human Golgi alpha 1, 2-mannosidase (IC) involved in N-glycan biosynthesis. J Biol Chem 2000;275:31655-31660. 208. Jitsuhara Y, Toyoda T, Itai T, Yamaguchi H. Chaperone-like functions of high-mannose type and complex-type N-glycans and their molecular basis. J Biochem (Tokyo) 2002;132:803-811. 209. Porwoll S, Loch N, Kannicht C, Nuck R, Grunow D, Reutter W, Tauber R. Cell surface glycoproteins undergo postbiosynthetic modifi cation of their N-glycans by stepwise demannosylation. J Biol Chem 1998;273:1075- 1085. 210. Zhao Y, Li J, Wang J, Xing Y, Geng M. Role of cell surface oligosaccharides of mouse mammary tumor cell lines in cancer metastasis. Indian J Biochem Biophys 2007;44:145-151. 211. Comelli EM, Sutton-Smith M, Yan Q, Amado M, Panico M, Gilmartin T, Whisenant T, Lanigan CM, Head SR, Goldberg D, Morris HR, Dell A, Paulson JC. Activation of murine CD4+ and CD8+ T lymphocytes leads to dramatic remodeling of N-linked glycans. J Immunol 2006;177:2431- 2440. 212. Razi N, Varki A. Masking and unmasking of the sialic acid-binding lectin activity of CD22 (Siglec-2) on B lymphocytes. Proc Natl Acad Sci U S A 1998;95:7469-7474. 78 REFERENCES

213. Keppler OT, Peter ME, Hinderlich S, Moldenhauer G, Stehling P, Schmitz I, Schwartz-Albiez R, Reutter W, Pawlita M. Differential sialylation of cell surface glycoconjugates in a human B lymphoma cell line regulates susceptibility for CD95 (APO-1/Fas)-mediated apoptosis and for infection by a lymphotropic virus. Glycobiology 1999;9:557-569. 214. Dybedal I, Yang L, Bryder D, Aastrand-Grundstrom I, Leandersson K, Jacobsen SE. Human reconstituting hematopoietic stem cells up-regulate Fas expression upon active cell cycling but remain resistant to Fas-induced suppression. Blood 2003;102:118-126. 215. Pearl-Yafe M, Yolcu ES, Stein J, Kaplan O, Yaniv I, Shirwan H, Askenasy N. Fas ligand enhances hematopoietic cell engraftment through abrogation of alloimmune responses and nonimmunogenic interactions. Stem Cells 2007;25:1448-1455. 216. Orschell-Traycoff CM, Hiatt K, Dagher RN, Rice S, Yoder MC, Srour EF. Homing and engraftment potential of Sca-1(+)lin(-) cells fractionated on the basis of adhesion molecule expression and position in cell cycle. Blood 2000;96:1380-1387. 217. Lapidot T, Dar A, Kollet O. How do stem cells fi nd their way home? Blood 2005;106:1901-1910. 218. Skelton TP, Zeng C, Nocks A, Stamenkovic I. Glycosylation provides both stimulatory and inhibitory effects on cell surface and soluble CD44 binding to hyaluronan. J Cell Biol 1998;140:431-446. 219. Sasaki K, Kurata K, Funayama K, Nagata M, Watanabe E, Ohta S, Hanai N, Nishi T. Expression cloning of a novel alpha 1,3-fucosyltransferase that is involved in biosynthesis of the sialyl Lewis x carbohydrate determinants in leukocytes. J Biol Chem 1994;20;269:14730-14737. 220. Barfi eld RC, Hale GA, Burnette K, Behm FG, Knapp K, Eldridge P, Handgretinger R. Autologous transplantation of CD133 selected hematopoietic progenitor cells for treatment of relapsed acute lymphoblastic leukemia. Pediatr Blood Cancer 2005;48:349-353. 221. Feller N, van der Pol MA, Waaijman T, Weijers GW, Westra G, Ossenkoppele GJ, Schuurhuis GJ. Immunologic purging of autologous peripheral blood stem cell products based on CD34 and CD133 expression can be effectively and safely applied in half of the acute myeloid leukemia patients. Clin Cancer Res 2005;11:4793-4801. 222. Koehl U, Zimmermann S, Esser R, Sorensen J, Gruttner HP, Duchscherer M, Seifried E, Klingebiel T, Schwabe D. Autologous transplantation of CD133 selected hematopoietic progenitor cells in a pediatric patient with relapsed leukemia. Bone Marrow Transplant 2002;29:927-930. 223. Steidl U, Kronenwett R, Haas R. Differential gene expression underlying the functional distinctions of primary human CD34+ hematopoietic stem and progenitor cells from peripheral blood and bone marrow. Ann N Y Acad Sci 2003;996:89-100.