<<

Leukemia (1999) 13, 1675–1690  1999 Stockton Press All rights reserved 0887-6924/99 $15.00 http://www.stockton-press.co.uk/leu REVIEW

The role of in normal hematopoiesis and hematological malignancies JW van Oostveen1, JJ Bijl2, FM Raaphorst2, JJM Walboomers2 and CJLM Meijer2

Departments of 1Hematology and 2Pathology, University Hospital, Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands

In the last decade it has become clear that homeobox contain- Table 1 Grouping of families, coding for transcription fac- ing genes (HOX genes) not only play a significant role in reg- tors, according to their DNA binding motif,244 and their reported func- ulating body formation, but in addition, they are contributing tions to organization and regulation of hematopoiesis. Modern mol- ecular technologies showed that deregulated expression or DNA binding- Gene (family) disruption of Hox genes can lead to altered characteristics of domain blood cells or disturbance of blood development. In this paper we review the role of HOX in hematopoiesis and leukemogenesis and speculate about their possible target Helix-turn-helix HOX genes embryonic genes and involvement in lymphomagenesis. development, Keywords: transcription factors; homeobox genes; embryogen- , esis; hematopoiesis; lymphoma; adhesion hematopoiesis finger GATA genes/E2A/Pax5 hematopoiesis, endodermal differentiation245 Transcription factors Zinc finger LIM Rbtn2/Lmo2 Embryonic red blood family cells265 Runt AML1(CBFA2) Fetal liver Transcription factors are sequence-specific DNA-binding pro- 266 teins with a variety of functions: some are thought to help fold hematopoiesis Basic helix- SCL gene (tal-1)/c- hematopoietic the DNA molecule into distinct domains; others assist in the loop-helix proteins/E2A commitment and initiation of DNA replication, and many control gene tran- differentiation194,246,247 scription. Binding affinity to DNA sequences, the concen- Basic c-fos/c-jun Proliferation248 tration of transcription factors and the presence of cofactors zipper are important determinants in gene activation. Transcription Winged helix forkhead/HNF-3 family embryogenesis, liver factors can be classified according to the three-dimensional organogenesis, tumorigenesis249 structure of their DNA-binding domains. More than 80% of ETS domain ets family Hematopoiesis31,250 all transcription factors are characterized by zinc finger, helix- Rel homology Rel/NF-kappa B family regulation of immune turn-helix, helix-loop-helix, and winged helix domain and inflammatory motifs (Table 1). Moreover, the complexity of transcriptional processes, communication regulation is increased by formation of functional hetero- or 251 homodimers of transcription factors (for instance c-fos and c- between cells 1 HMG-box HMG family (Tcf-1/Lef-1) B and T cell jun ), or by participation in large complexes (for development252 instance c-myc). Transcription of a particular gene is an event regulated by a complex network of transcription factors. The HNF-3, hepatocyte nuclear factor; HMG, high mobility group; HOX, total set of transcription factors present in a cell – induced by homeobox; the Table is modified from Clevers HC, Grosschedl R. extracellular signals – provides a specific genetic imprint that Transcriptional control of lymphoid development: lessons from results in a distinct response. Transcription factors have a gene targeting. Immunol Today 1996; 17: 336–343. regulatory role in proliferation and in all kinds of differen- tiation processes, like embryogenesis, organogenesis, and also hematopoiesis (Table 1).2–9 ensure its own survival by proliferation before differentiating into all lineages of blood cells. This balance of proliferation and differentiation is highly dynamic, and pluripotent stem Hematopoiesis cells and progenitor cells are produced in relation to physio- logical stresses of the (such as bleeding, or Specialized blood cells of all lineages originate from the pluri- infection). The intriguing question arises how this process of potent stem cell by hematopoiesis. Blood cells have a limited blood cell development is controlled, and which genes are life-span, and new cells are generated continuously through- involved in this complex process. out life. The ancestral pluripotent stem cell must therefore Hematopoiesis during embryogenesis

Correspondence: JW van Oostveen, Department of Hematology, Uni- The anatomical site of hematopoiesis changes during early life versity Hospital Vrije Universiteit (VU), De Boelelaan 1117 Amster- 10 11 dam, PO Box 7057, NL-1007 MB Amsterdam, The Netherlands; Fax: phases (reviewed by Zon and Auerbach ). During 31 20 4442601 embryogenesis, early hematopoiesis occurs in the yolk sac, Received 30 December 1998; accepted 12 July 1999 starting in at day 15 and continuing for 6 weeks. Review JW van Oostveen et al 1676 Hematopoietic stem cells – derived from the ventral meso- early hematopoietic progenitor cells into the lymphoid lin- derm – and vascular progenitors migrate to the yolk sac to eage.43,44 Several other genes specifically involved in the lym- form blood cell islands.12 These islands consist of an endoder- phopoiesis have been found, such as Pax5/BSAP, E2A, Oct-2, mal layer (supporting growth), a central core of hematopoietic PU.1, Ets-1 and Tcf-1.45–53 Of these transcription factors both stem cells, and an endothelial layer that surrounds the blood Pax5 and Oct-2 genes belong to the HOX class II genes, which islands.13 Although progenitors of most blood cells are contain a homeobox in addition to another characteristic present, these stem cells predominantly differentiate into sequence (see below). erythroid cells and occasionally into monocytes and megakar- yocytes.14–16 They are primarily important for embryonic hem- atopoiesis17 and die during further development of the Development of the lymphoid lineage . In the embryo itself, an additional population of hem- atopoietic stem cells is present in the dorsal mesentery (also The stem cell first differentiates into a lymphoid and a myeloid called the aorta gonad mesonephros region, or AGM). This progenitor cell. All lymphocytes are derived from a common population initiates definitive hematopoiesis (including that of lymphoid progenitor cell. Differentiation is accompanied by the lymphoid lineage), first in fetal liver and spleen, and sub- changes in and immunophenotype, eg loss of sequently in bone marrow (BM).17 BM remains the primary stem cell marker CD34 during differentiation and induction hematopoietic organ in adult life. Developmentally controlled of lineage-specific markers such as CD19 and CD22. Lympho- changes in location of blood cell development requires coor- cytes are responsible for the body’s response to foreign anti- dinate regulatory decisions in cell fate and reorganization of gen. As the number of potential antigens is enormous, this cells. requires a large pool of lymphocytes, each displaying speci- ficity for a single antigen. Antigen responses are divided into the humoral immune response, mediated by B cells, and the Transcription factors in hematopoiesis cellular immune response, mediated by T cells. Stimulated by specific like IL–7 and Flt3 , Hematopoiesis includes many decisions in lineage commit- B cells initially develop in the BM. Their maturation is ment that depend on coordinate expression of lineage-specific accompanied by rearrangement of immunoglobulin (Ig) genes genes. This development program is governed by transcription encoding their membrane-associated antigen . This factors. Blood cell development is particularly complex produces a large population of B cells each expressing their because it occurs at different body sites, partly determined by unique Ig molecules. Immature B cells with a self-reactive developmental stage. Fetal hematopoiesis occurs in the liver, receptor are negatively selected before they leave the BM. whereas in adults it is initiated in BM. Lymphoid cells reach After migration and homing to lymph nodes, follicular B cells their final maturation stage in peripheral lymphoid organs, that recognize the appropriate antigen on follicular dendritic such as lymphnodes, spleen and extranodal lymphoid tissues. cells continue to mature in the germinal center reaction.54 The microenvironment of these specific body sites has a pro- During this process cells with an antigen-specific Ig receptor found influence on the differentiation program.18,19 Therefore, are selected to survive,55 and will further differentiate into genes that can specify positional information, such as homeo- plasma cells and memory B cells. Cells without antigen speci- box (HOX) genes, might be strongly related with the control ficity, or expressing a self-specific antigen receptor, undergo of hematopoiesis.20 (). Expression studies have revealed a number of transcription T cell development occurs primarily in a separate organ, factor genes that are active in specific cell lineages and differen- the thymus. This gland is formed from the third pharyngeal tiation stages during hematopoiesis. Their function was clarified pouch and consists of well-defined cortical and medullar in vitro by transfection studies, and in vivo by knock-out and zones. Immature prothymocytes enter the thymus, attracted by transgenic mice experiments. Disruption of chemotactic factors. Here, they interact with stromal cells that genes often resulted in a block of terminal differentiation of produce lineage-specific cytokines such as IL-7, stem cell fac- particular hematological cell types, disclosing their role in a tor (SCF), Flt3 ligand and tumor (TGF). This particular lineage. Homeobox genes were originally identified drives the T cells into further differentiation.56 T cells acquire as master control genes during , but antigen receptor diversity by rearrangement of T cell receptor they affect various steps in hematopoiesis as well.21–32 These (TCR) genes,54 followed by selection for T cells reacting with genes, the class I homeobox genes, are the subject of this non-self antigens.57 This process generates various types of T- review. Although knock-out mice have been generated for sev- helper cells (CD4 positive) and cytotoxic T cells (CD8 eral genes of the family, defects in hematopoiesis positive).58,59 Commitment and differentiation of cells to the of such knock-out mice have only recently been described for lymphoid lineage is under precise genetic control, and there the HOXA9 gene.33 HOXA9 knock-out mice exhibit disturbed is evidence that especially the HOXC cluster plays a role in differentiation of myeloid and megakaryocytic cells. Early T cell this regulatory process.26,27 development is severely affected and associated with decreased intracellular expression of CD3 and the TCR ␤-chain, while at the cell surface expression of IL-7R and E cadherin are Development of non-lymphoid lineages reduced.34 Members of the GATA gene family appear crucial for development of various cell types, like GATA-1 for erythroid All non-lymphoid blood cells are derived from a common cells,35–38 and GATA-3 for T-cells.39 By contrast, GATA-2 plays myeloid progenitor cell. Progenitors differentiate along parti- a role in differentiation of all hemopoietic lineages, suggesting cular lineages to mature blood cells, under influence of a that this gene is required for the expansion of the early progeni- multilineage stimulating factor (IL-3) and lineage-restricted tor cell pool.40–42 Lack of the Ikaros gene resulted in the com- growth factors (eg IL-5, Epo (erythropoietin) and GM-CSFs plete lack of lymphoid cells (including early lymphoid (granulocyte–macrophage colony-stimulating factors).60 This progenitors), suggesting that Ikaros affects differentiation of is accompanied with downregulation of the progenitor marker Review JW van Oostveen et al 1677 CD34 and induction of lineage-specific markers such as CD15 first discovered homeobox gene of Drosophila, called (granulocyte), CD14 and CD68 (monocyte/microphage), Antennapedia. The class II, or diverged homeobox genes, CD41 and CD42 (megakaryocyte) and H-antigen, which exhibit Ͻ50% homology to the Antennapedia gene. Class II forms the backbone of the ABO blood group proteins on homeobox genes are grouped in several families, based on erythrocytes.61 Cells also undergo morphological changes, the presence of additional conserved sequences, such as the which are most dramatic for erythroid and megakaryocytic paired box in genes of the Pax family and the POU box in development. This results in denucleated erythrocytes and the POU domain family (including members as Oct-1, 2, 3 platelets, respectively.62 Commitment and differentiation of and 4 and Pit-1).81 In this review we only discuss the homeo- myeloid cells is carefully controlled. Genes of the HOX cluster box containing class I gene cluster. are involved in this process: HOXA for the myeloid lineage, class I genes are called HOX genes in the new HOXB for the erythroid lineage. Recent studies suggest nomenclature.82,83 HOX are organized in clusters, and human that HOXC genes might also be involved in myeloid HOX elements can partly be aligned with those in Drosphila differentiation.32 (Figure 2). Drosophila homeobox genes encompass the antennapedia-complex (ANT-C) and bithorax-complex (BX- C),84 located on one . The 39 human HOX genes Homeobox genes are organized in four clusters (HOXA, -B, -C and -D), located 83,85–87 General aspects of homeobox genes on 7, 17, 12 and 2, respectively. Each cluster contains a minimum of nine HOX genes. HOX genes Homeobox genes are characterized by the presence of a con- belonging to different clusters but with highly homologous served 183 bp sequence, the homeobox (Figure 1) first disco- homeodomains, can be positionally aligned on the chromo- vered in genes of the fruit-fly .63 some. This arranges HOX genes in 13 paralogue groups; for Nuclear magnetic resonance and crystallographic studies instance, paralogue group one consists of HOXA1, HOXB1 showed that the three-dimensional structure of the homeobox and HOXD1 (Figure 2). The homology between the homeodo- is a helix-turn-helix motif, consisting of four helices.64–68 This mains of HOX genes belonging to the same paralogue group motif recognizes DNA sequences having core sequences rang- is higher than between those of one cluster, suggesting that ing from TGAT to TAAT69,70 to TTAT and TTAC. Regions out- first a tandem duplication and subsequently a duplication of 80,88–90 side the homeobox in the variable domain provide binding the complete cluster occurred during . The specificity to homeoproteins for specific DNA sequences. As genomic organization of HOX genes and cis-regulatory explained below in the process of binding to DNA, homeo- sequences suggest that processes involving enhancer sharing proteins appear to display cooperative binding to other form the basis for highly orchestrated expression of HOX regulatory proteins. genes, necessary for the guidance of sequential steps in the 91–93 Embryonic expression studies and analysis of homeotic differentiation pathways. mutants revealed that homeobox genes specify cell identity and positioning in the developing embryo.19,71–74 Some mutations in homeobox genes resulted in the duplication of HOX intact morphological structures to anterior or posterior located segments in Drosophila. For example, the Drosophila home- Each of the four clusters covers a distance of approximately 81 otic mutant called Antennapedia resulted in formation of an 150–200 kb on the chromosome. Each single gene has a additional pair of legs on the head, instead of antennae.75–78 polyadenylation site and is thought to have its own promoter. Ј The identification of homeobox genes in many other species Normally, at the 5 end of the homeobox gene, one or more indicates that homeobox genes provide an evolutionary introns are present. HOXC4, HOXC5 and HOXC6 are parti- conserved elementary regulatory mechanism for major cular because they are also under control of a master pro- 94,95 differentiation programs.78–80 moter. It is not known whether transcription from the common promoter or the individual promoters is regular for these genes. Organization of homeobox genes at the chromosomal Most HOX genes give rise to more than one RNA transcript, level generated by the use of alternative splice sites or by differen- tial promoter use, as has been shown for HOXC6 and Homeobox genes are categorized in two large groups. The HOXA9.96–98 Alternative splice variants were described that Ͼ class I homeobox genes are highly homologous ( 80%) to the are functionally equivalent.99 The distinct tissue distribution of alternative transcripts of several HOX genes implies that there are also alternative transcripts that might encode func- tionally different proteins. However, it is not known whether all alternative transcripts encode functional proteins.97,98,100 Several mechanisms have been identified that regulate expression of HOX genes. The identification of TAAT Figure 1 Structure of the homeobox protein. Homeobox proteins sequences and retinoic acid response elements in upstream are composed of several structural units, including a variable region, regions allows the binding and activation or repression by which determines a protein’s specific activity, a small connective homeoproteins101–106 (autoregulation) and activation of hinge region (IYPWM) and a homeodomain of 60 amino acid expression by retinoic acid.107–116 Early HOX gene expression sequence, encoded by the homeobox, that is similar in all these pro- in Drosophila is dependent on genes of the pair- teins. The aminoterminal region of the proteins often begin with rule (activators) and gap (repressor) families. During later MSSLYYXN and the carboxyterminal domain tends to be acidic, indi- cating that it might act as a transcriptional domain. The homeodomain stages of development, the maintenance of HOX expression consists of four alpha helices (1–4), one of which (black) recognizes is guided by the polycomb (Pc-G) and trithorax (Trx-G) and binds to a specific DNA sequence in the target genes. genes.117–122 Pc-G genes repress HOX-gene transcription, Review JW van Oostveen et al 1678

Figure 2 Comparative organization of HOX complexes. The alignments of the Drosophila and human complexes and chromosomal localiza- tions are shown. Gray intensities indicate similar homeodomain sequences. The order of the genes along the chromosome is as shown, with the order roughly corresponding to where along the anterior–posterior body axis gene is expressed or, in the case of , the anterior- most limit of expression. Categories 1–13 are called ‘paralogue’ groups and are indicative of homeodomain sequence similarity. Black ovals indicate the lack of a paralogue from a cluster. The assignment of the Drosophila homeobox genes Scr (Sex comb reduced), Ant (Antennapedia), Ubx (Ultrabithorax) and abd-A (Abdominal-A) to any particular paralogue group is uncertain. Paralogue group 10–13 do not have homologues in the Drosophila homeobox gene clusters. The direction of transcription from each gene is indicated by an arrow. Note that genes early expressed in the embryo are more sensitive to retinoic acid (RA) than later expressed genes. Modified from Manak and Scott.243

while Trx-G genes contribute to strong expression.123–125 Fol- genital malformations demonstrated the involvement of HOX lowing their discovery in Drosophila, Pc-G genes have also genes mutations in these syndromes.145–147 Furthermore, stud- been identified in mammals, including humans, where they ies with knock-out mice revealed the involvement of HOX11 appear to have a similar role.125–131 The mixed lineage-, or in the spleen,148 Pax1 and Hoxa-3 in the developing thy- myeloid-lymphoid, leukemia gene (MLL) is the only known mus149–150 while Hoxa3, -b3 and -d3 mutant mice show spe- human Trx-G gene.132,133 Human Pc-G genes are differentially cific disturbance of thymus, parathyroid and thyroid develop- transcribed during development of CD34+ into CD34− BM ment.151 Similarly, mice expressing a Hlx homeobox progenitor cells,131 but other than that little else is known transgene exhibit severe disturbance of both T and B cell about their role in hematopoiesis. development.152 Thus, HOX genes not only play a role in development of the embryo, but they also contribute to regu- lation of hematopoiesis. Importantly, this holds true for both HOX gene expression in embryogenesis and fetus and adult. In the following sections, we shall turn our organogenesis attention to the significance of HOX genes for control of hem- atopoiesis, and their involvement in oncogenesis. Furthermore The physical order of the genes is also reflected by the temporal HOX genes might be involved in different types of human sequence of their activation during embryogenesis and organo- solid tumors,142,143,153–155 however, these malignancies are genesis.134–136 The expression order along the anterior–pos- beyond the scope of this review. terior axis of the embryo is colinear with the 3Ј–5Ј organization of HOX genes on the chromosomes (Figure 2).3,4,76,137–139 Moreover, transcripts of a given HOX gene can usually not be Homeobox genes in hematopoiesis detected in the developing embryo until the product of its 3Ј neighbor is present. This so-called temporal colinearity may HOX gene expression in hematopoietic cell lines occur only in cells with high mitotic rates, resulting in a strong linkage between patterning and growth control and an uni- Analysis of human and mouse hematopoietic cell lines directional pattern process.140 The expression domains of HOX revealed that some HOX genes were only expressed in cell genes in have marked anterior boundaries, but less lines of one hematopoietic lineage, while other HOX genes sharp posterior boundaries, and this results in extensive overlap were expressed in cell lines of several lineages.42–44,156–162 of expression domains in the posterior regions. This implies that HOX genes that are expressed in cell lines of all lineages are the nature of body structures which grow out from individual HOXC8, HOXA7 and HOXB7. HOX genes showing a lineage- segments apparently depends on the specific combination of restricted expression are HOXA10 (myelomonocytic cell HOX products.141 lines),158 and HOXC4 (predominantly expressed in lymphoid Involvement of HOX genes in the formation of organs is cell lines).45 Furthermore, expression of all HOXB genes was reflected by their expression in kidney, colon, liver, spleen, mainly associated with cell lines of the erythroid lineage.42,43 skin, spinal cord and hematopoietic cells in human adults.142–144 Except for HOXB1, which is not expressed in the hematopo- In addition, recent studies of human synpolydactyly and con- ietic compartment, and HOXB13, which has been identified Review JW van Oostveen et al 1679 only recently, no expression pattern in other hematopoietic bone marrow cells than in more primitive populations. Some cell lines is yet known of HOXB genes.163 All, but one164 study of these genes (like ENX-1, M31, HP1HS␣ and HP1HS␥) appear reported that genes of the HOXD cluster are not expressed in the earliest clonogenic precursors, whereas other Pc-G in the hematopoietic compartment. Taken together, no clear genes (M32, M33, Mel-18 and Mph1/Rae3) were only detect- coordinate expression of clustered members of HOX genes able at later stages of differentiation. Interestingly, the ninth was found in particular hematopoietic cell lineages, but known human Pc-G gene, Bmi-1, was detected at low fre- instead HOX genes appear expressed in a complex pattern of quencies in mature bone marrow cells but was preferentially lineage- and non-lineage restriction. expressed in primitive long-term culture-initiating cells with high proliferative potential. Progressive upregulation of Pc-G gene expression possibly induces progressive formation of het- Functional analysis of HOX gene expression in hematopoietic erochromatin, ultimately resulting in 3Ј→5Ј closure of the cell lines: Several groups made use of the capability of HOX clusters. some hematopoietic cell lines to differentiate after induction with an appropriate stimulus. Erythroid-myeloid cell lines transfected with a HOXB6 antisense construct, showed an Functional analysis of HOX gene expression in normal hema- increase of erythroid features. Transfection with a HOXB6 topoiesis: New techniques, including retrovirus-mediated sense construct resulted in the loss of erythroid features, indi- transfection of HOX genes in mouse BM and the generation cating its involvement in erythropoiesis.165 Also, retroviral of knock-out mice, have made it possible to monitor the effect gene transfer of HOXB4 in embryonic stem cells enhanced of modulated HOX expression in vivo. Both overexpression their erythropoietic potential.166 HOXB7 is temporarily acti- and blocking of expression of one individual HOX gene in vated in promyelocytic cell line HL60 after induction with Vit bone marrow cells has an enormous effect on development

D3 or TPA leading to monocytic differentiation. By contrast, of specific cell pools. For example, mouse BM cells, over- HL60 cells induced to differentiate into the granulocytic expressing HOXB8 as a result of retroviral transfection, lineage lack HOXB7 expression. This was subsequently had enhanced capacity to form IL-3-dependent cell lines.174 confirmed by inhibition of colony formation derived from Furthermore, an increase was noted in self-renewal of early GM-CSF-stimulated bone marrow (BM) in an antisense clonogenic cells, concomitant with expansion of progenitor assay.167,168 Finally, Blatt et al169 reported that overexpression cells and a propensity to evolve into leukemia. of HOXB8 (Hox2.4) inhibited the myeloid differentiation of Sauvageau et al175 recently demonstrated that over- IL-6 inducible mouse cells. Thus, modulation of HOX gene expression of HOXB4 in normal mouse BM results in an expression in hematopoietic cell lines results in differences in increase of the most primitive cell populations, without devel- lineage commitment and maturation of these cells. opment into leukemia or other malignancies. Serial transplan- tations of bone marrow cells, initially transfected with HOXB4, revealed a 50-fold higher number of totipotent hema- HOX gene expression in normal hematopoietic bone topoietic stem cells in primary and secondary recipients, com- marrow cells pared with mice bearing a control vector expressing neomycin resistance alone. After retroviral gene transfer in embryonic Analysis of unfractionated normal human BM cells using RNase stem cells, HOXB4 was shown to influence the early progeni- protection assays demonstrated expression of HOXB2, HOXB6 tors with mixed erythroid/myeloid features and not the pro- and HOXA10,43,158 indicating that HOX expression in cell lines genitors of later stages, specific for the erythroid, granulocytic is not solely due to the transformed state. Furthermore, RT-PCR or monocytic lineage.176 Also, overexpression of the human analysis on small purified subpopulations of CD34+ hematopo- paralogue HOXC4 in human hematopoietic progenitor cells, ietic progenitor cells showed that the number of HOX genes resulted in progenitor cell expansion, without changing their expressed was the highest for the HOXA cluster, and decreased capacity to differentiate properly (Corte, personal for genes of the HOXB and HOXC clusters. No expression of communication). These results suggest that paralogue mem- the HOXD cluster genes was found.170–173 Not only the number bers HOXB4 and HOXC4 may have a similar function during of HOXA genes, but also the expression level of these genes the same time span in hematopoiesis. was higher than that of HOXB genes in enriched CD34+ subpo- Another gene investigated in this model is HOXA10, the pulations, including the self-renewing stem cell.172 expression of which is restricted to myeloid cells. Mice over- In addition, it appeared from two studies that HOX genes expressing HOXA10 in their bone marrow cells have are colinearly expressed during differentiation of myeloid and increased numbers of stem cells and myeloid progenitor erythroid cells.172,173 Downregulation of the 3Ј end located cells.177 Additionally, the number of megakaryocytic and genes (eg HOXB3) was observed in more mature fractions, primitive blast colonies was increased in the recipient ani- including the committed progenitor cells, while genes located mals. By contrast, HOXA10-overexpressing marrow cells did at the 5Ј end (eg HOXA10) seem to be more actively not contribute to the B cell population, suggesting that expressed throughout the CD34+ compartment. Their tran- HOXA10 switch-off is required for B cell differentiation. scription decreases only when cells progress to mature CD34− Moreover, the HOXA10-transfected animals were extremely stages.172 Induction of human progenitors into erythroid and susceptible to a myeloproliferative disorder that resembled granulocytic lineages in vitro revealed a 3Ј to 5Ј end wave of acute myeloblastic leukemia. HOXB gene expression colinear with differentiation,173 similar An alternative approach to functional analysis of HOX to the expression order in the embryonic development genes is to reduce or knock-out their normal expression. Using (Figure 2). homologous recombination in embryonic stem cells, mice Expression of HOX regulatory genes, like Pc-G genes, dur- with a defect in HOXA9 were created.178 Mutants were heal- ing human hematopoiesis remains relatively unexplored. Les- thy and fertile, but had reduced numbers of peripheral blood sard and colleagues131 demonstrated that the expression level granulocytes and lymphocytes. Lymphoid tissues, spleen and of eight out of nine Pc-G genes is much higher in more mature thymus were smaller, and the number of myeloid and B cells Review JW van Oostveen et al 1680 was reduced. By contrast, no significant effect on pluripotent required. Recently, the mechanism by which HOX11 inter- stem cells seemed to be observed.52 However, more recent feres in the regulation of the cell cycle was shown by Kawabe data of the same investigators (ASH 1998) indicate a major et al.193 By using a two hybrid system they found that HOX11 stem cell defect in these animals. These results indicated that can interact with the protein phosphatases PP2A and PP1, HOXA9 activates differentiation of stem cells, myeloid pro- resulting in disruption of the G2/M cell cycle checkpoint. genitors and, in contrast to HOXA10, lymphoid progenitor The third translocation, t(1;19)(q23;p13.3), produces a cells. chimeric fusion protein that links the amino-terminal trans- An antisense oligodeoxynucleotides (ASOs) approach has activating domain of E2A to the carboxy-terminal DNA bind- been used to diminish HOX gene function in vivo as well. ing domain of the diverged class II homeobox containing Blocking expression of HOXA5 with ASOs had a dual effect regulatory gene, PBX1.194–196 The translocation correlates with on growth of BM cells. Firstly, myeloid colony forming was 25% of pre-B ALL.197 Five alternative splice variants of E2A- inhibited, and secondly, the erythroid progenitor population PBX1 have been identified. Protein products were shown to was expanded.179 In contrast to HOXA5, an ASO blockade of possess various transforming potentials.198 Whether PBX1 several HOXB and C genes led to inhibition of erythroid col- alone has transforming potential is unknown, but PBX proteins ony formation.20,180 Some pitfalls using the antisense ASOs can form DNA-binding complexes with HOX proteins.199 treatment must be considered, however. First, only a 50% These complexes are thought to form the basis for autoregu- reduction of mRNA levels is generally achieved, and an effect lation and HOX-induced cellular transformation.199,200 of HOX inhibition may not necessarily be observed. Secondly, Inordinate HOX gene expression as a result of abnormal ASOs could bind non-specifically and affect the function of regulation has been observed in leukemic cells compared other genes. with their normal counterparts.181,201 For example, the diverged class II homeobox containing gene HLX (formerly known as HB24) is strongly expressed in activated lympho- Expression of HOX genes in hematological malignancies cytes and hematopoietic progenitor cells.202,203 In samples of ALL and AML, however, very high levels of mRNA transcripts It will not be surprising that genes which are involved in regu- were found, in contrast to the normal levels detected in CML lation of differentiation and proliferation can contribute to the material (which represent more mature cells).204 Transient malignant transformation of cells, because their deregulated expression of HLX into a subpopulation of hematopoietic pro- expression could lead to defective control of cell growth. genitors resulted in impairment of differentiation into mature HOX genes not only control the differentiation and prolifer- hematopoietic cells, suggesting that downregulation of HLX is ation of embryonic cells, but their expression in adult necessary for normal differentiation of hematopoietic progeni- organs142,143,181,182 also suggests a function in the mainte- tors. That this gene also has oncogenic transformation nance of the differentiation state of cells. Aberrant expression capacity was proven by the fact that HLX-transfected Jurkat of these genes might be a result of DNA amplification, translo- cells induced tumor formation when injected into nude cated genes, mutations or deregulated gene expression. mice.205 Another example of leukemic transformation through deregulated HOX gene expression is presented by HOXA9. Recent studies have shown that co-activation of the Hoxa9 Leukemias: HOX gene mutations associated with leukem- and Meis1 genes is potentially sufficient for transformation ias are not known, but some leukemias are associated with and co-transfection of these genes in mouse hematopoietic irregular expression of HOX genes due to translocation or cells rapidly leads to development of AML.206,207 abnormal gene regulation. Translocations have been reported There is scarce evidence that trx-G and Pc-G genes are for class I and II homeobox genes. Class I gene HOXA9 was directly involved in oncogenesis by HOX proteins. The MLL found to be involved in translocation t(7;11)(p15;p15) present (Hrx) trithorax gene is implicated in mixed lineage (myeloid- in AML FAB M2-M4, linking the complete HOXA9 gene to a lymphoid) leukemia.208–210 Knocking out the Bmi-1 Pc-G gene gene for nucleoporin NUP98.183,184 Diverged, class II homeo- in mice specifically affects expression of HOXC5 and box gene HOX11 is juxtaposed to the J␦1 region of the TCR HOXC6,211 while expression of Bmi-1 as a transgene results delta gene in the translocation t(10;14)(q24;q11). This translo- in a high incidence of lymphomas.212 This is most likely cation has been found in 5–7% of T-ALL.185–187 related to downregulation of the /INKA4 gene, an inhibitor A variant translocation t(7;10)(q35;q24) fuses HOX11 to the of the cell cycle.213 TCR␤ gene on chromosome 7.188 Since the coding region of The expression of HOXC genes and in particular of the HOX11 is not affected by the translocation,189 deregulated HOXC4 gene,26 seems to be associated with commitment to HOX11 expression is thought to be a key event in the devel- the lymphoid lineage. We focussed on the expression of opment of leukemias. The amino- and carboxyl termini of HOXC4, HOXC5 and HOXC6 since these three genes have a HOX11, however, seem dispensable for transformation.190 common promoter, and produce one mRNA molecule that is The transforming capacity of HOX11 was demonstrated in spliced in separate transcripts. The expression pattern of these vitro by transfection of HOX11 into NIH 3T3 mouse cells. 3Ј end cluster-C genes were studied by RT-PCR in normal HOX11 can transactivate various mammalian and yeast pro- hematopoiesis and hematopoietic leukemias. Acute B- and T- moters,191 suggesting that the oncogenic activity of HOX11 lymphatic leukemias showed HOXC4 and -C6 expression but might not be restricted to T cell leukemias. Retroviral lacked HOXC5 expression like normal lymphoid cells.27 In transduction of HOX11 in mouse BM cells, enriched for the myeloid lineage, the expression patterns of HOXC4 and progenitors, yielded cell lines at high frequency consisting of -C6 were largely similar in neoplastic cells of AML.32 How- immature cell lines of the myeloid lineage.192 However, these ever, HOXC5 gene transcripts were poorly expressed in these cells are IL-3-dependent and showed no leukemogenicity, neoplastic cells and in normal myeloid cells. Surprisingly, suggesting that HOX11 promotes proliferation and blocks ter- none of the three genes were expressed in CML. Expression minal differentiation of hematopoietic cells.192 For further pro- of all three genes could generally be detected in immature gression to malignancy secondary events (eg mutations) are AMLs (FAB M1–M4 and M6). In mature (normal) myeloid cells Review JW van Oostveen et al 1681 the expression of these genes seemed to be downregulated,32 contain the t(2;5) translocation and normally have an agreeing with the results of studies reported in the literature. intermediate prognosis. Mature myeloid leukemias localized in tissue simultaneously Given the involvement of HOX genes in hematopoiesis, expressed HOXC4, HOXC5 and HOXC6 genes, in contrast to organogenesis of hematopoietic organs, and pathogenesis of leukemic myeloid cells in the same stage of maturity. These tumors and leukemias, investigation of HOX gene expression findings are suggestive for a relationship between HOXC gene in lymphomas seemed warranted. expression and the presence of adhesion molecules at the sur- We studied the expression pattern of the 3Ј end cluster-C face of these cells (see below). genes HOXC4, HOXC5 and HOXC6 in lymphoid hematopo- In conclusion, if HOX regulatory genes are of central impor- iesis to see whether the expression pattern of HOXC genes in tance in normal hematopoietic development, then disruption NHL, the neoplastic equivalents of normal lymphoid cells, is of this pathway might be a common feature in human altered compared to normal lymphoid cells. Lymphoid B and leukemias.207 T cell lines, arrested at different stages of maturity, early ALL, and healthy lymphoid cells from peripheral blood and hyp- erplastic tonsillar tissue were used as a model for normal Non-Hodgkin’s lymphomas: To explain our interest in the lymphoid development. Semiquantitative RT-PCR demon- relationship between HOXC genes and non-Hodgkin’s lym- strated increased expression of HOXC4 and HOXC6 with pro- phomas (NHL), we would like to briefly discuss the develop- gressive maturation of both B- and T-lymphocytes. In contrast ment of the present conceptual definition of NHL. to HOXC4 and HOXC6 gene expression in neoplastic and NHL form a heterogeneous group of malignant lymphoprol- normal lymphoid cells, we detected expression of HOXC5 iferations. They can be roughly divided into B and T cell lym- only in lymphoma-derived cell lines and some B cell lym- phomas depending on lineage markers, like Ig- or TCR-gene phomas.27 These findings suggested a role for HOXC5 in lym- rearrangements, and markers expressed on the cell surface. In phomagenesis. the past, several classifications have been used to describe To distinguish HOXC gene expression in normal cells from these various types of lymphomas. The updated Kiel classi- that in lymphoma cells, we used non-radioactive RNA in situ fication is nowadays widely used in Europe. The concept of hybridization (RISH) to determine HOXC transcription at the this classification was that various types of non-Hodgkin’s single cell level. We further analyzed expression of HOXC4, lymphomas are thought to represent malignant counterparts of HOXC5 and HOXC6 in histologic subtypes of primary lymphoid cells. They are arrested at various stages of normal cutaneous lymphomas, in benign cutaneous lymphoid lesions differentiation, while maintaining their proliferating and hyperplastic lymphoid organs. HOXC4 and HOXC6 were activity.214 The Kiel classification was mainly based on nodal detected by RT-PCR and RISH in almost all samples of primary lymphomas. Extranodal lymphomas, such as mycosis fungo- cutaneous NHL, benign lymphoid lesions and hyperplastic ides and maltomas that display unique clinicopathological lymphoid organs.223 However, expression of HOXC5 was features, did not fit in this concept. Present insights in the only detectable by RISH in primary cutaneous anaplastic large biology of lymphocytes, in particular the recirculation of lym- T cell lymphomas. Semiquantitative RT-PCR revealed phocytes through the body and homing of lymphocytes to spe- expression of HOXC5 in some other subtypes of cutaneous B- cific body sites, increased our understanding of extranodal and T-NHL, but this was much weaker than in primary lymphomas.215,216 The recirculation pattern of lymphocytes is cutaneous anaplastic large T cell lymphomas. Since we could specific for particular body compartments, meaning that there not induce HOXC5 expression in PBL stimulated with ␣- are subpopulations of lymphocytes that home specifically to CD3/CD28 mAbs in the presence of IL-2, HOXC5 might act lymph nodes, to the MALT (mucosa-associated lymphoid as a putative oncogen in the genesis of cutaneous anaplastic tissue) or other body compartments. This homing is mediated large T cell lymphomas.223 by specific adhesion molecules, which react with site-specific Keeping in mind that HOX genes provide cells with pos- adhesion molecules on the endothelial cells of high endo- itional as well as developmental information, different histol- thelial venules (HEV), named vascular adressins or tissue pos- ogic subtypes of B- and T-NHL were analyzed for expression ition markers. For example, lymphocytes expressing of HOXC4, HOXC5 and HOXC6. Expression of HOXC4 and cutaneous lymphocyte antigen CLA (the proposed skin hom- HOXC6 could be detected by RISH in the majority of lym- ing receptor217,218), interact with E-selectin on the superficial phomas. Interestingly, HOXC6 could not be detected in dermal vessels.219 Those expressing the ␣4␤7 , serving lymphnode-based B-CLL. HOXC5 gene expression appeared as the homing receptor for mucosal sites, interacts with MAd- associated with primary cutaneous anaplastic large T cell lym- CAM-1, present on the HEV of the Peyer’s patch.220,221 This phomas and B cell lymphomas with plasmocytic differen- compartmentalization of the immune system is accompanied tiation from the oro-digestive tract.30 Thus, HOXC5 showed a with different functions and the existence of distinct type- and site-restricted expression pattern in NHL and may immune cell populations. be involved in formation of these lymphomas. Extending the concept of the ‘neoplastic equivalent’ with this aspect of tissue-restricted lymphocytes, we can now define NHL as neoplastic counterparts of functional recirculat- ing tissue restricted lymphocytes activated after antigen stimu- Target genes of homeoproteins lation.222 From this point of view it is conceivable that lym- phomas may arise with similar histologic features, but with Although expression patterns of many class I HOX genes have different clinical behavior, different translocations, different been described in embryogenesis and hematopoiesis, the tar- adhesion molecule expression pattern, or differences in the gets of the majority of their protein products have yet to be presence of viral sequences (such as EBV or HTLV-1). Thus, identified. Genes that have been shown to be controlled by anaplastic lymphomas arising in the skin express CLA, lack homeoproteins encode adhesion molecules, transcription the t(2;5) translocation and have a good prognosis. By factors (in particular the HOX genes themselves) and growth contrast, nodal anaplastic lymphomas do not express CLA, factors. Review JW van Oostveen et al 1682 Modulation of adhesion molecule expression Modulation of transcription factor expression Another group of target genes of the homeoproteins are genes By modulation of HOX gene expression in vitro it was coding for transcription factors (Table 3). The most obvious observed that homeoproteins are able to influence the transcription factors under regulation of homeoproteins are adhesive capacity of myeloid leukemia cells.224 Adhesion the homeobox genes themselves. Moreover, coordinate molecules are categorized in the following families: the expression of HOX genes in embryogenesis is achieved in part immunoglobulin (Ig) gene superfamily, cadherins, , by cross-talk between the various members of the HOX selectins and the CD44 family. Analysis of adhesion molecule complexes.7,72,134 For several HOX genes auto- and/or cross- profile of cells, overexpressing HOX genes, revealed that regulatory interactions within the homeotic network have members from different adhesion molecule families were been described by genetic analysis and in transfection affected. Examples are given in Table 2. assays.234–237 For example, human HOXD9 (formerly HOX4C) Cotransfection experiments demonstrated that the is transactivated by the HOXD9 protein itself and by HOXD10 expression of N-CAM, a member of the Ig gene superfamily, (formerly HOX4D) protein by cotransfection. Conversely, is involved in the development of the neural network in mouse Hoxd-8 (formerly Hox-4.3) represses activation of embryogenesis. This protein, present on natural killer (NK) HOXD9.106 Furthermore, HOXC5 can be transactivated by cells, could be activated by increased levels of Xenopus HOX genes from the same cluster (HOXC6), as well as from HoxC6 and HoxB9.225,226 In contrast, HoxB8 had an inhibi- the HOXD cluster (HOXD9 and HOXD10).102 In vivo experi- tory effect on the expression of N-CAM, indicating that tan- ments with Drosophila and mice indicated that a is demly arranged genes, such as HOXB8 and HOXB9, act necessary for autoregulation of Hoxb-1.101 These experiments together to control one gene by differential regulation.225 In demonstrate that the HOX cross-talk region (HCR) can be addition, a study of HOX gene expression by the human meta- occupied simultaneously by several protein molecules and static melanoma cell line Me 655/2 demonstrated that mela- allows various combinations of HOX proteins to interact. noma clones could be classified into two populations: one Although no data are present on cross- or auto-regulatory group expressed HOX-C genes, which correlated with low events in hematopoietic cells, these regulatory circuits may be level expression of ICAM and absent expression of VLA-2, active in hematopoiesis as well. -5 and -6. By contrast, high level expression of VLA-2, -5 and In addition to regulation of homeobox gene expression by -6, and ICAM coincided with silence of the HOX-C locus.227 homeoproteins, gene expression of other transcription factors, In addition to these class I genes, class II homeobox genes involved in organogenesis and proliferation, have also been (Phox2 and Cux) also showed cross-talk by either activating shown to be under control of homeoproteins (Table 3). Con- (Phox2) or inactivating (Cux) N-CAM expression.228 Further- cerning hematopoiesis, it is of interest that knock-out mice for more, decreased expression of ICAM-1 (CD54) was found in HOXA3 have a reduced expression of the Pax1 gene, suggest- myeloid FDC-P1 cells after enforced expression of the mouse ing that Pax1 is a target of HOXA3. Moreover, these mutants homeobox genes Hlx.229 In the same study, the expression of were athymic, which is in agreement with the of CD44 also decreased as a result of the Hlx overexpression.229 Pax1 mutant mice.150 Since Pax1 mutants also had reduced CD44 is an integral membrane protein, which is widely dis- + + + numbers of CD4 /CD8 immature and CD4 mature thymo- tributed over hematopoietic and non-hematopoietic cells. In cytes, Pax1 is also involved in the maturation of thymo- addition to important adhesion events necessary for migration, cytes.149 Moreover, the gene encoding transcription factor T cell activation230 and probably lymphopoiesis231,232 within Thy-1, involved in the maturation of thymocytes, was dis- the bone marrow, a variant of CD44 was shown to have meta- closed as a target of Pax1. The expression order from HOXA3, static potential.233 Pax1 to Thy-1 in the development of the thymus and thymo- Integrins can also be modulated by HOX genes. For cytes indicates an early function as master control genes for instance, increased expression of ␣IIb␤3, a surface fibronectin HOX genes in organogenesis and lymphopoiesis. receptor, was induced by HOXD3 in the erythroleukemia cell lines HEL and K562.164 No example of HOX-mediated regu- Modulation of expression of other targets of lation of selectins are known. All adhesion molecules have homeoproteins important functions in embryogenesis, , lin- eage commitment, hematopoiesis and cell migration, stressing Other targets of homeoproteins, not belonging to adhesion the importance of homeoproteins in these cellular events. molecules or transcription factors are mentioned in Table 4.

Table 2 Genes of adhesion molecules as targets of homeoproteins

Adhesion Species HOX Function Reference molecules

N-CAM human/xenopus HOXC6/HOXB8/HOXB9 neural development 226, 253, 254 mice Pax6/Pax8/Phox2/Cux ICAM-1 mice Hlx embryogenesis 229 CD44 mice Hlx embryogenesis 229 ␣IIb␤3 human HOXD3 embryogenesis 164 L-CAM human HOXD9/HNF-1 liver development 255 Cytotactin mice Pax6/Pax8/evx-1 (counter)adhesion, ordering tissues 239, 254 E-cadherin mice HOXA9 Cell adhesion 34 mgl-1 mice HOXC8 putative adhesion 256 Connectin drosophila Ubx cell adhesion 257

CAM, cell adhesion molecule; Ubx, Ultrabithorax. Review JW van Oostveen et al 1683 Table 3 Genes of transcription factors as targets of homeoproteins

Target gene Species Regulatory homeoprotein Function Reference

HOXD9 human HOXD9/HOXD10/HOXD8 embryogenesis, limb development 106 HOXC5 human HOXC6/HOXD9/HOXD10 embryogenesis 102 Pax1 mice HOXA3 thymus development 150 TTF-1 human/mice HOXB3 expression of and thyroperoxidase 258 Thy-1 mice Pax1 maturation of thymocytes 149 c-fos mice Pax1/Pax6/Pax8 proliferation 254 fkh human/drosophila HOXA5/Scr liver development 259, 260 salm drosophila Ant transcription factor 261 fkh, forkhead; SCR, sex comb reduced; TTF-1, thyroid transcription factor; Ant, Antennapedia; Ubx, Ultrabithorax.

Table 4 Genes encoding other proteins as targets of homeoproteins

Other genes Species Regulatory homeoprotein Function Reference

␤-APP Mice HOXC8 cellulary regulatory processes 262 CD19 Mice Pax5 (BSAP) B cell development 48 Thyroglobulin/thyroperoxidase Mice Pax8 thyroid development 263 Drosophila Ubx growth factor (TGF␤) 264

␤-APP, ␤-amyloid precursor protein (involved in Alzheimer disease and Down syndrome); Ubx, Ultrabithorax.

Interestingly, the Pax5 protein (BSAP, B-cell lineage-specific embryogenesis, starting with the 3Ј genes and progressing to activator protein), is able to induce the expression of B cell 5Ј genes of the HOX cluster. This ‘wave’ of HOX gene membrane protein CD19.34 BSAP is essential for B cell expression has been found in CD34+ BM cells173 and T development34 and deregulated BSAP expression has been cells238 after activation, suggesting a temporal axis of HOX implicated in formation of large-cell lymphomas and some gene expression in hematopoiesis. Moreover, most mature follicular lymphomas.39 The finding that many target genes of CD34+ subpopulations showed a preference for expression of homeoproteins are involved in hematopoiesis emphasizes 5Ј end genes compared to expression of more 3Ј end genes their regulatory role in this process. Furthermore, deregulated in more immature fractions.172 HOX gene expression might have profound effects on prolifer- Concerning positional information, we have considered a ation and differentiation of a cell, and promote neoplasia. role of homeobox genes in homing of lymphocytes to specific body sites by regulation of adhesion molecules, which would give these cells a positional imprint. Immature blood cells lay Epilogue ordered in the three-dimensional structure of the BM, inter- acting with a stromal meshwork through adhesion molecules In this part we will discuss two major aspects which have not and cytokines.19 Progression of maturation is accompanied been dealt with in the previous sections: general aspects of with migration within the BM, requiring downregulation of HOX gene expression in hematopoiesis, and the significance adhesion molecules determined by the given BM localization, of HOXC5 in NHL. in combination with expression of other adhesion molecules. Thus, blood cell differentiation is tightly associated with a spe- cific position and a spatial axis, that might be coordinated by General aspects of HOX gene expression in hematopoiesis HOX genes. However, the release of hematopoietic cells from BM to peripheral blood requires downregulation or shedding of Our study on HOXC4, HOXC5 and HOXC6 gene expression in myeloid and lymphoid hematopoietic compartments adhesion molecules. Since it seems possible that adhesion revealed a lineage- and stage-restricted expression pattern of molecule expression can be controlled by homeoproteins, downregulation of the expression of (particular) HOX genes in these genes. Other groups showed commitment of some HOX + genes to particular lineages or stages of hematopoiesis. An mature BM populations is also necessary. Compared to CD34 immature BM cells HOXA10 is only found in CD34− mature interesting question is whether there are parallels between the + − function of HOX genes in the formation of the embryonic cells. Indeed, transition from the CD34 to the CD34 com- and their regulatory role in hematopoiesis. In other partment was accompanied with the downregulation of words, does the homeotic network regulate the process of HOXA10 gene expression.201 This indicates that HOXA10 is + blood cell maturation by providing positional and temporal temporarily expressed during maturation of immature CD34 + information? Secondly, are HOX genes expressed in a spatial BM cells to mature CD34 BM cells. and temporal manner during hematopoiesis? When mature BM cells are released in the peripheral blood Stage- and lineage-restricted expression patterns of HOX stream, adhesion molecules are required for the homing of genes in hematopoiesis indicates that their transcription is lymphocytes to lymphoid organs and extranodal lymphoid coordinated in time. Indeed, HOX gene expression during tissues. Therefore, we suggest that a second ‘wave’ of HOX hematopoiesis appears to occur in a pattern similar to that in gene expression is activated, reinducing (other) adhesion mol- Review JW van Oostveen et al 1684 ecules. This view is supported by our observation of HOXC4 pendent functions: the C-terminus represses immediate–early and HOXC6 expression in mature lymphocytes. gene expression via CArG elements. Mol Cell Biol 1990; 10: 4243–4255. 2 Driever W, Nu¨sslein-Volhard C. The bicoid protein determines Significance of HOXC5 expression in non-Hodgkin’s position in the Drosophila embryo in a concentration-dependent manner. Cell 1988; 54: 95–104. lymphomas 3 St Johnston D, Nu¨sslein-Volhard C. The origin of pattern and polarity in the Drosophila embryo. Cell 1992; 68: 201–219. The regulatory function of HOX genes in cell differentiation 4Nu¨sslein-Volhard C. Determination of the embryonic axes of and proliferation, made them possible candidates for a role in Drosophila. Development Sup 1991; 1: 1–10. development of tumors and leukemias. We have proposed a 5 Driever W, Thoma G, Nu¨sslein-Volhard C. Determination of spa- role for HOXC genes in lymphomagenesis. HOXC5 tial domains of zygotic gene expression in the Drosophila embryo expression was specifically found in anaplastic large T cell by the affinity of binding sites for the bicoid . Nature 1989; 340: 363–367. lymphomas (predominantly localized in the skin) and in B cell 6 Grossniklaus U, Cadigan KM, Gehring WJ. Three maternal coor- lymphomas with plasmacytic differentiation (mainly originat- dinate systems cooperate in the patterning of the Drosophila ing from the oro-digestive tract). These specific expression pat- head. Development 1994; 120: 3155–3171. terns of HOXC5 indicate a relationship with (1) lymphomas 7 Hunt P, Krumlauf R. Hox codes and positional specification in equivalent to terminal differentiation stages of both B and T embryonic axes. Annu Rev Cell Biol 1992; 8: 227– lymphocytes; and (2) with development of lymphomas at spe- 256. cific body sites, ie the skin and the oro-digestive tract. 8 Krumlauf R. Mouse HOX genetic functions. Curr Opin Genet Dev 1993; 3: 621–625. In this respect the link between homeoproteins and acti- 225,226,239 9 Krumlauf R. HOX genes and in the branchial vation of adhesion molecule expression is extremely region of the vertebrate head. Trends Genet 1993; 9: 106–112. interesting. In both primary cutaneous and primary oro- 10 Zon LI. Development biology of hematopoiesis. Blood 1995; 86: digestive tract lymphomas, site-specific adhesion molecules 2876–2891. are expressed, such as the skin-homing receptor CLA and 11 Auerbach R, Huang H, Lu L. Hematopoietic stem cells in the ␣4␤7, the mucosa-associated homing receptor.240,241 It is mouse embryonic yolk sac. Stem Cells 1996; 14: 269–280. possible that HOXC5 is directly or indirectly involved in 12 Palis J, McGrath KE, Kingsley PD. Initiation of hematopoiesis and expression of these site-specific adhesion molecules. A vasculogenesis in murine yolk sac explants. Blood 1995; 86: 156–163. relationship between HOX gene expression and integrin mol- 13 Tavassoli M. Embryonic and fetal hematopoiesis. An overview. ecules is supported by the finding that genes coding for inte- Blood Cells 1991; 17: 269–283. grin chains are clustered in regions of the chromosomes that 14 Liu CP, Auerbach R. of murine T cells. Thymus-regu- also contain HOX genes. This suggests co-evolution of these lated development of T cell receptor bearing cells derived from two gene families.242 Moreover, the position of the ␤7 chain embryonic yolk sac. Eur J Immunol 1991; 21: 1849–1861. gene near the HOXC cluster on chromosome 12q13,242 sup- 15 Johnson GR, Barker DC. Erythroid progenitor cells and stimulat- ports the hypothesis that homeoprotein C5 might be involved ing factors during murine embryonic and fetal development. Exp ␣ ␤ Hematol 1985; 13: 200–209. in modulation of 4 7 expression. To test this hypothesis, 16 Wong PMC, Chung SW, Chui DHK, Eaves CJ. Properties of the transfection experiments should be carried out using HOXC5 earliest clonogenic hematopoietic precursors to appear in the constructs, and additionally ␣4␤7 and CLA expression analy- developing murine yolk sac. Proc Natl Acad Sci USA 1986; 83: sis of the transfected cells should be performed. We trans- 3851–3854. fected a vector containing a HOXC5 construct driven by a 17 Detrich HW, Kieran MW, Chan FY, Barone LM, Yee K, Rund- CMV promoter in pre-B cells. Unfortunately, it was not poss- statler JA, Pratt S, Ransom D, Zon LI. Intraembryonic hematopo- ible to select for cells containing the construct because trans- ietic cell migration during vertebrate development. Proc Natl Acad Sci USA 1995; 92: 10713–10717. fectants died. Moreover, limiting dilution assays produced cell 18 Boyd RL, Tucek CL, Godfrey DI, Izon DJ, Wilson TJ, Davidson populations without the HOXC5 construct, indicating that NJ, Bean AG, Ladyman HM, Ritter MA, Hugo P. The thymic these cells died as a result of high expression of HOXC5 pro- microenvironment. Immunol Today 1993; 14: 445–459. tein due to the strong CMV promoter. Therefore, HOXC5-con- 19 Tavassoli M, Hardy CL. Molecular basis of homing of intravene- taining vectors should be constructed containing a weaker or ously transplanted stem cells to the marrow. Blood 1990; 76: inducible promoter. 1059–1070. Another interesting finding is that HOXC5 is mainly 20 Rich IN, Zimmerman F. Homeobox gene expression during hem- opoietic ontogeny in the mouse. Blood 1993; 82: 68a. expressed in B and T cell lymphomas equivalent to mature 21 Lawrence HJ, Largman C. Homeobox genes in normal hematopo- lymphoid differentiation stages. Since HOXC5 could not be iesis and leukemia. Blood 1992; 80: 2445–2453. found in normal and activated lymphoid cells or benign 22 Keller G, Wall C, Fong AZC, Hawley TS, Hawley RG. Over- lymphoid skin lesions, it is not likely that HOXC5 expression expression of HOX11 leads to the immortalization of embryonic in these lymphomas is induced by local micro-environmental precursors with both primitive and definitive hematopoietic factors. Therefore, we suggest that HOXC5 acts as an onco- potential. Blood 1998; 92: 877–887. gene in these lymphomas. In addition, HOXC5 activation – 23 Magli MC, Barba P, Celetti A, De VG, Cillo C, Boncinelli E. Coor- dinate regulation of HOX genes in human hematopoietic cells. resulting in expression of site-specific adhesion molecules – Proc Natl Acad Sci USA 1991; 88: 6348–6352. might keep cutaneous anaplastic T cell lymphomas and mal- 24 Mathews CH, Detmer K, Boncinelli E, Lawrence HJ, Largman C. tomas of the oro-digestive tract predominantly localized in Erythroid-restricted expression of homeobox genes of the human particular body compartments (skin and oro-digestive tract). HOX 2 locus. Blood 1991; 78: 2248–2252. This could favourably affect their prognosis. 25 Petrini M, Quaranta MT, Testa U, Samoggia P, Tritarelli E, Care` A, Cianetti L, Valtieri M, Barletta C, Peschle C. Expression of selected human HOX-2 genes in B/T acute lymphoid leukemia References and interleukin-2/interleukin-1␤-stimulated natural killer lym- phocytes. Blood 1992; 80: 185–193. 1 Gius D, Cao X, Rauscher III FJ, Cohen DR, Curran T, Sukhatme 26 Lawrence HJ, Stage KM, Mathews CH, Detmer K, Scibienski R, VP. Transcriptional activation and repression by Fos are inde- Mackenzie M, Migliaccio E, Boncinelli E, Largman C. Expression Review JW van Oostveen et al 1685 of HOX C homeobox genes in lymphoid cells. Cell Growth Differ M, Valk van der M, Riele te HPJ, Berns A, Murre C. E2A proteins 1993; 4: 665–669. are required for proper B cell development and initiation of 27 Bijl JJ, Van Oostveen JW, Kreike M, Rieger E, Van der Raaij- immunoglobulin gene rearrangements. Cell 1994; 79: 885–892. Helmer L, Walboomers JMM, Corte G, Boncinelli E, Van den 47 Zuang Y, Soriano P, Weintraub H. The helix-loop-helix gene E2A Brule AJC, Meijer CJLM. Expression of HOXC4, -C5, and -C6 in is required for B cell formation. Cell 1994; 79: 875–884. human lymphoid cell lines, leukemias and in benign and malig- 48 Urba´nek P, Wang Z-Q, Fetka I, Wagner EF, Busslinger M. Com- nant lymphoid tissue. Blood 1996; 87: 1737–1745. plete block of early B cell differentiation and altered patterning 28 Krishnaraju K, Hoffman B, Liebermann DA. Lineage-specific of the posterior midbrain in mice lacking Pax5/BSAP. Cell 1994; regulation of hematopoiesis by HOX-B8 (HOX-2.4) inhibition of 79: 901–912. granulocytic differentiation and potentiation of monocytic differ- 49 Mckercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, entiation. Blood 1997; 90: 1840–1849. Baribault H, Klemsz M, Feeney AJ, Wu GE, Paige CJ, Maki RA. 29 Zimmermann F, Rich IN. Mammalian homeobox B6 expression Targeted disruption of the PU.1 gene results in multiple hemato- can be correlated with erythropoietin production sites and poietic abnormalities. Embo J 1996; 15: 5647–5658. erythropoiesis during development, but not with hematopoietic 50 Bories J-C, Willerford DM, Grevin D, Davidson L, Camus A, Mar- or nonhematopoietic stem cell populations. Blood 1997; 89: tin P, Stehelin D, Alt FW. Increased T-cell apoptosis and terminal 2723–2735. B cell differentiation induced by inactivation of the Ets-1 proto- 30 Bijl JJ, Van Oostveen JW, Walboomers JMM, Horstman A, Van . Nature 1995; 377: 635–638. den Brule AJC, Willemze R, Meijer CJLM. HOXC4, HOXC5 and 51 Muthusamy N, Barton K, Leiden JM. Defective activation and HOXC6 expression in non-Hodgkin’s lymphoma: preferential survival of T cells lacking the Ets-1 transcription factor. Nature expression of the HOXC5 gene in primary cutaneous anaplastic 1995; 377: 639–643. T-cell and oro-gastrointestinal tract mucosa-associated B-cell 52 Anderson KL, Smith KA, Conners K, McKercher SR, Maki RA, lymphomas. Blood 1997; 90: 4116–4125. Torbett BE. Myeloid development is selectively disrupted in PU.1 31 Quaranta MT, Petrini M, Tritarelli E, Samoggia P, Care´ A, Bottero null mice. Blood 1998; 91: 3702–3710. L, Testa U, Peschle C. HOXB cluster genes in activated natural 53 Krenacs L, Himmelmann AW, Quintanilla-Martinez L, Fest T, killer lymphocytes: expression from 3Ј → 5Ј cluster side and pro- Riva A, Wellmann A, Bagdi E, Kerl JH, Jaffe ES, Raffeld M. Tran- liferative function. J Immunol 1996; 157: 2462–2469. scription factor B-cell-specific activator protein (BSAP) is differen- 32 Bijl JJ, Van Oostveen JW, Walboomers JMM, Brink ATP, Vos W, tially expressed in B cells and in subsets of B-cell lymphomas. Ossenkoppele GJ, Meijer CJLM. Differentiation and cell-type Blood 1998; 92: 1308–1316. restricted expression of HOXC4, HOXC5 and HOXC6 in myeloid 54 Zouali M, MacLennan CM. Molecular events in the development leukemias and normal myeloid cells. Leukemia 1998; 12: of the lymphocyte repertoire. Immunol Today 1992; 13: 41–43. 1724–1732. 55 Liu Y-J, Joshua DE, Williams GT, Smith CA, Gordon J, MacLen- 33 Lawrence HJ, Helgason CD, Sauvageau G, Fong S, Izon DJ, nan IC. Mechanism of antigen-driven selection in germinal Humphries RK, Largman C. Mice bearing a targeted interruption centres. Nature 1989; 342: 929–931. of the homeobox gene have defects in myeloid, erythroid, 56 Haks MC, Oosterwegel MA, Blom B, Spits HM, Kruisbeek AM. and lymphoid hematopoiesis. Blood 1997; 89: 1922–1930. Cell-fate decisions in early T cell development: regulation by 34 Izon DJ, Rozenfield S, Fong ST, Ko¨mu¨ves L, Langman C, Lawr- receptors and the pre-TCR. Semin Immunol 1999; 11: ence HJ. Loss of function of the homeobox gene Hoxa-9 perturbs 23–37. early T-cell development and induces apoptosis in primitive thy- 57 Boehmer H. Thymic selection: a matter of life and death. Immu- mocytes. Blood 1998; 92: 383–393. nol Today 1992; 13: 454–458. 35 Simon MC, Pevny L, Wiles MV, Keller G, Costatini F, Orkin SH. 58 Rodewald H-R. Pathways from hematopoietic stem cells to thym- Rescue of erythroid development in gene targeted GATA-1- ocytes. Curr Opin Immunol 1995; 7: 176–187. mouse embryonic stemm cells. Nat Genet 1992; 1: 92–98. 59 Hugo P, Petrie HT. Multiple routes for late intrathymic precursors 36 Weiss MJ, Keller G, Orkin SH. Novel insights into erythroid to generate CD4+CD8+ thymocytes. Adv Mol Cell Biol 1992; 5: development revealed through in vitro differentiation of GATA- 37–53. 1 embryonic stem cells. Genes Dev 1994; 8: 1184–1197. 60 Campana D, Behm FG. Leukaemia immunophenotyping: uses for 37 Pevny L, Lin C-S, D’Agati V, Simon MC, Orkin SH, Constantini F. establishing diagnosis, predicting outcome and assessing treat- Development of hematopoietic cells lacking transcription factor ment response. In: Whittaker JA (ed). Leukaemia. Blackwell GATA-1. Development 1995; 121: 163–172. Scientific: Oxford, 1992, pp 203–226. 38 Weiss MJ, Orkin SH. Transcription factor GATA-1 permits sur- 61 Testa NG, Dexter TM. Clonal analysis of cell development in vival and maturation of erythroid precursors by preventing hemapoiesis. In: Whittaker JA (ed). Leukaemia. Blackwell Scien- apoptosis. Proc Natl Acad Sci USA 1995; 92: 9623–9627. tific: Oxford, 1992, pp 153–172. 39 Ting CN, Olson MC, Barton KP, Leiden JM. Transcription factor 62 Beug H, Mu¨llner EW, Hayman MJ. Insights into erythroid differ- GATA-3 is required for development of the T-cell lineage. Nature entiation obtained from studies on avian erythroblastosis virus. 1996; 384: 474–478. Curr Opin Cell Biol 1994; 6: 816–824. 40 Shivdasani RA, Orkin SH. The transcriptional control of hemato- 63 McGinnis W, Levine MS, Hafen E, Kuroiwa A, Gehring WJ. A poiesis. Blood 1996; 87: 4025–4039. conserved DNA sequence in homeotic genes of the Drosophila 41 Briegel K, Lim K-C, Plank C, Beug H, Engel J, Zenke M. Ectopic Antennapedia and bithorax complexes. Nature 1984; 308: expression of a conditional GATA-2/ receptor chimera 428–433. arrests erythroid differentiation in a -dependent manner. 64 Qian YQ, Otting G, Furukubo-Tokunaga K, Affolter M, Gehring Genes Dev 1993; 7: 1079–1109. WJ, Wuthrich K. NMR structure determination reveals that the 42 Tsai F-Y, Keller G, Kuo FC, Weiss MJ, Chen J-Z, Rosenblatt M, homeodomain is connected through a flexible linker to the main Alt F, Orkin SH. An early hematopoietic defect in mice lacking body in the Drosophila Antennapedia protein. Proc Natl Acad the transcription factor GATA-2. Nature 1994; 371: 221–226. Sci USA 1992; 89: 10738–10742. 43 Georgopoulos K, Moore DD, Derfler B. Ikaros, an early lymph- 65 Wolberger C, Vershon AK, Liu B, Johnson AD, Pabo CO. Crystal oid-specific transcription factor and a putative mediator for T cell structure of a MAT␣2 homeodomain–operator complex suggests commitment. Science 1992; 258: 808–812. a general model for homeodomain–DNA interactions. Cell 1991; 44 Georgopoulos K, Bigby M, Wang J-H, Molnar A, Wu P, Winandy 67: 517–528. S, Sharpe A. The Ikaros gene is required for the development of 66 Qian YQ, Billeter M, Otting G, Mu¨ller M, Gehring WJ, Wu¨tthrick all lymphoid lineages. Cell 1994; 79: 143–156. K. The structure of the Antennapedia homeodomain determined 45 Hromas R, Orazi A, Neiman RS, Maki R, Van Beveran C, Moore by NMR spectroscopy in solution: comparison with prokaryotic J, Klemsz M. Hematopoietic lineage- and stage-restricted repressors. Cell 1989; 59: 573–580. expression of the ETS oncogene family member PU.1. Blood 67 Kissinge CR, Lui B, Martin-Blanco E, Kornberg TB, Pabo CO. 1993; 82: 2998–3004. Crystal structure of an homeodomain–DNA complex at 46 Bain G, Robanus Maandag EC, Izon DJ, Amsem D, Kruisbeek 2.8A resolution: a framework for understanding homeodomain– AM, Weintraub BC, Krop I, Schlissel MS, Feeney AJ, Roon van DNA interactions. Cell 1990; 63: 579–590. Review JW van Oostveen et al 1686 68 Billeter M, Qian YQ, Otting G, Mu¨ller M, Gehring WJ, Wu¨thrick 95 Simeone A, Mavilio F, Acampora D, Giampaolo A, Faiella A, K. Determination of the nuclear magnetic resonance solution Zappavigna V, D’Esposito M, Pannese M, Russo G, Boncinelli E, structure of an Antennapedia homeodomain–DNA complex. J Peschle C. Two human homeobox genes, c1 and c8: structure Mol Biol 1993; 234: 1084–1093. analysis and expression in embryonic development. Proc Natl 69 Desplan C, Theis J, O’Farrell PH. The sequence specificity of Acad Sci USA 1987; 84: 4914–4918. homeodomain–DNA interaction. Cell 1988; 54: 1081–1090. 96 Shimeld SM, Gaunt SJ, Coletta PL, Geada AMC, Sharpe PT. Spa- 70 Affolter M, Schier A, Gehring WJ. Homeodomain proteins and tial localisation of transcripts of the Hox-C6 gene. J Anat 1993; the regulation of gene expression. Curr Opin Cell Biol 1990; 2: 183: 515–523. 485–495. 97 Fujimoto S, Araki K, Chisaka O, Araki M, Takagi K, Yamamura 71 Harding K, Wedeen C, McGinnis W, Levine M. Spatially regu- K. Analysis of the murine Hoxa-9 cDNA: an alternatively spliced lated expression of homeotic genes in Drosophila. Science 1985; transcript encodes a truncated protein lacking the homeodomain. 229: 1236–1242. Gene 1998; 209: 77–85. 72 McGinnis W, Krumlauf R. Homeobox genes and axial patterning. 98 Kim MH, Chang HH, Shin C, Cho M, Park D, Park HW. Genomic Cell 1992; 68: 283–302. structure and sequence analysis of human HOXA-9. DNA Cell 73 Scott MP, Tamkun JW, Hartzell GW. The structure and function Biol 1998; 17: 407–414. of the homeodomain. Biochim Biophys Acta Rev Cancer 1989; 99 Busturia A, Vernos I, Macias A, Casanova J, Morata G. Different 989: 25–48. forms of Ultrabithorax proteins generated by alternative splicing 74 Malicki J, Schughart K, McGinnis W. Mouse Hox-2.2 specifies are functionally equivalent. EMBO J 1990; 9: 3551–3555. thoracic segmental identity in drosophila and larvae. 100 Chariot A, Castronovo V, Le P, Gillet C, Sobel ME, Gielen J. Cell 1990; 63: 961–967. Cloning and expression of a new HOXC6 transcript encoding a 75 Dessain S, McGinnis W. Regulating the expression and function repressing protein. Biochem J 1996; 319: 91–97. of homeotic genes. Curr Opin Genet Dev 1991; 1: 275–282. 101 Popperl H, Bienz M, Studer M, Chan S-K, Aparicio S, Brenner 76 Gehring WJ. The homeobox in perspective. Trends Biochem Sci S, Mann RS, Krumlauf R. Segmental expression of Hoxb-1 is con- 1992; 17: 277–280. trolled by a highly conserved autoregulatory loop dependent 77 Gehring WJ, Hiromi Y. Homeotic genes and the homeobox. upon exd/pox. Cell 1995; 81: 1031–1042. Annu Rev Genet 1986; 20: 147–173. 102 Arcioni I, Simeone A, Guazzi S, Zappavigna V, Boncinelli E, 78 Gehring WJ. The homeobox: a key to the understanding of devel- Mavilio F. The upstream region of the human homeobox gene opment. Cell 1985; 40: 3–5. HOX3D is a target for regulation by retinoic acid and HOX 79 McGinnis W, Garber RL, Wirz J, Kuroiwa A, Gehring WJ. A hom- homeoproteins. EMBO J 1992; 11: 265–277. ologous protein-coding sequence in Drosophila homeotic genes 103 Simeone A, Acampora D, Arcioni L, Andrews PW, Boncinelli E, and its conservation in other metazoans. Cell 1984; 37: 403–408. Mavilio F. Sequential activation of HOX2 homeobox genes by 80 Holland P. Homeobox genes in vertebrate evolution. Bioessays retinoic acid in human embryonal carcinoma cells. Nature 1990; 1992; 14: 267–273. 346: 763–766. 81 Duboule D (ed). Guidebook to the Homebox Genes. Oxford Uni- 104 Popperl H, Featherstone MS. An autoregulatory element of the versity Press: Oxford, 1994. murine Hox-4.2 gene. Embo J 1992; 11: 3673–3680. 82 Scott MP. Vertebrate homeobox . Cell 1992; 105 Cianetti L, Di Cristofaro A, Zappavigna V, Bottero L, Boccoli G, 71: 551–553. Testa U, Russo G, Boncinelli E, Peschle C. Molecular mech- 83 Scott MP. A rational nomenclature for vertebrate homeobox anisms underlying the expression of the human HOX-5.1 gene. (HOX) genes. Nucleic Acids Res 1993; 21: 1687–1688. Nucleic Acids Res 1990; 18: 4361–4368. 84 Regulski M, Harding K, Kostriken R, Karch F, Levine M, 106 Zappavigna V, Renucci A, Izpisu`a-Belmonte JC, Urier G, Peschle McGinnis W. Homeo box genes of the Antennapedia and Bitho- C, Duboule D. HOX4 genes encode transcription factors with rax complexes of Drosophila. Cell 1985; 43: 71–80. potential auto-and cross-regulatory capacities. Embo J 1991; 10: 85 Boncinelli E, Acampora D, Pannese M, D’Esposito M, Somma 4177–4187. R, Gaudino G, Stornaiuolo A, Cafiero M, Faiella A, Simeone A. 107 Marshall H, Studer M, Po¨pperl H, Aparicio S, Kuroiwa A, Brenner Organization of human class I homeobox genes. Genome 1989; S, Krumlauf R. A conserved retinoic acid response element 31: 745–756. required for early expression of the homeobox gene Hoxb-1. 86 Acampora D, D’Esposito M, Faiella A, Pannese M, Migliaccio E, Nature 1994; 370: 567–571. Morelli F, Stornaiuolo A, Nigro V, Simeone A, Boncinelli E. The 108 Popperl H, Featherstone MS. Identification of a retinoic acid human HOX gene family. Nucleic Acids Res 1989; 17: response element upstream of the murine Hox-4.2 gene. Mol 10385–10402. Cell Biol 1993; 13: 257–265. 87 Apiou F, Flagiello D, Cillo C, Malfoy B, Poupon MF, Dutrillaux 109 Bottero L, Simeone A, Arcioni L, Acampora D, Andrews PW, B. Fine mapping of human HOX gene clusters. Cytogenet Cell Boncinelli E, Mavilio F. Differential activation of homeobox Genet 1996; 73: 114–115. genes by retinoic acid in human embryonal carcinoma cells. Rec 88 Shughart K, Kappen C, Ruddle FH. Duplication of large genomic Res Cancer Res 1991; 123: 133–143. regions during the evolution of vertebrate homeobox genes. Proc 110 Simeone A, Acampora D, Nigro V, Faiella A, D’Esposito M, Stor- Natl Acad Sci USA 1989; 86: 7067–7071. naiuolo A, Mavilio F, Boncinelli E. Differential regulation by reti- 89 Holland PWH, Garcia-Fernandez J, Williams NA, Sidow A. Gene noic acid of the homeobox genes of the four HOX loci in human duplications of the origins of vertebrate development. Develop- embryonal carcinoma cells. Mech Dev 1991; 33: 215–227. ment Suppl 1994; 125–133. 111 Boncinelli E, Simeone A, Acampora D, Mavilio F. HOX gene 90 Kappen C, Schughart K, Ruddle FH. Early evolutionary origin of activation by retinoic acid. Trends Genet 1991; 7: 329–334. major homeodomain sequence classes. Genomics 1993; 18: 112 Tabin CJ. , , and growth factors: toward 54–70. molecular models for limb development. Cell 1991; 66: 199– 91 Mann RS. Why are Hox genes clustered? Bioessays 1997; 19: 217. 661–664. 113 Guazzi S, Pintonello ML, Vigano A, Boncinelli E. Regulatory 92 Gould A, Morrison A, Sproat G, White RA, Krumlauf R. Positive interactions between the human HOXB1, HOXB2 and HOXB3 cross-regulation and enhancer sharing: two mechanisms for proteins and the upstream sequence of the Otx2 gene in embry- specifying overlapping Hox expression patterns. Genes Dev onal carcinoma cells. J Biol Chem 1998; 273: 11092–11099. 1997; 11: 900–913. 114 Packer AI, Crotty DA, Elwell VA, Wolgemuth DJ. Expression of 93 Sharpe J, Nonchev S, Gould A, Whiting J, Krumlauf R. Selectivity, the murine Hoxa4 gene requires both autoregulation and a con- sharing and competitive interactions in the regulation of Hoxb served retinoic acid response element. Development 1998; 125: genes. EMBO J 1998; 17: 1788–1798. 1991–1998. 94 Simeone A, Pannese M, Acampora D, D’Esposito M, Boncinelli 115 Lufkin T. Transcriptional regulation of vertebrate Hox genes dur- E. At least three human homeoboxes on belong ing embryogenesis. Crit Rev Eukaryot Gene Exp 1997; 7: 195– to the same transcription unit. Nucleic Acids Res 1988; 16: 213. 5379–5390. 116 Maconochie MK, Nonchev S, Studer M, Chan SK, Popperl H, Review JW van Oostveen et al 1687 Shan MH, Mann RS, Krumlauf R. Cross-regulation in the mouse 140 Duboule D. Temporal colinearity and the phylotypic pro- HoxB complex: the expression of Hoxb2 in rhobomere 4 is regu- gression: a basis for the stability of a vertebrate Bauplan and the lated by Hoxb1. Genes Dev 1997; 11: 1885–1895. evolution of morphologies through . Development 117 Simon J, Chiang A, Bender W. Ten different polycomb group 1994; (Suppl) 135–142. genes are required for spatial control of the abdA and abdB 141 Kessel M, Gruss P. Homeotic transformation of murine vertabrae homeotic products. Development 1992; 114: 493–505. and concomitant alteration of Hox code induced by retinoic 118 Kennison JA. Transcriptional activation of Drosophila homeotic acid. Cell 1991; 67: 89–104. genes from distinct regulatory elements. Trends Genet 1993; 9: 142 Cillo C, Barba P, Freschi G, Bucciarelli G, Magli MC, Boncinelli 75–79. E. HOX gene expression in normal and neoplastic human kidney. 119 Paro R. Imprinting a determined state into the chromatin of Dro- Int J Cancer 1992; 51: 892–897. sophila. Trends Genet 1990; 6: 416–421. 143 De Vita G, Barba P, Odartchenko N, Givel J-C, Freschi G, Bucci- 120 Pirotta V. Chromatin complexes regulating gene expression in arelli G, Magli MC, Boncinelli E, Cillo C. Expression of homeo- Drosophila. Curr Opin Gen Dev 1995; 5: 466–472. box-containing genes in primary and metastatic colorectal can- 121 van der Lugt N, Alkema M, Berns A, Deschamps J. The poly- cer. Eur J Cancer 1993; 29A: 887–893. comb-group homolog Bmi-1 is a regulator of murine Hox gene 144 Kehrl JH. Homeobox genes in hematopoiesis. Crit Rev Oncol expression. Mech Dev 1996; 58: 153–164. Hematol 1994; 16: 145–156. 122 van Lohuizen M. Functional analysis of mouse Polycomb group 145 Innis JW. Role of HOX genes in human development. Curr Opin genes. Cell Mol Life Sci 1998; 54: 71–79. Pediatr 1997; 9: 617–622. 123 Jenuwein T, Laible G, Dorn R, Reuter G. SET domain proteins 146 Mark M, Rijli FM, Chambon P. Homeobox genes in embryogen- modulate chromatin domains in eu- and heterochromatin. Cell esis and pathogenesis. Pediatr Res 1997; 42: 421–429. Mol Life Sci 1998; 54: 80–93. 147 Boncinelli E. Homeobox genes and disease. Curr Opin Genet 124 Pirotta V. Polycombing the genome. PcG, trxG and chromatin Dev 1997; 7: 331–337. silencing. Cell 1998; 93: 333–336. 148 Roberts CWM, Sonder AM, Lumsden A, Korsmeyer SJ. Develop- 125 Yu BD, Hess JL, Horning SE, Brown GA, Korsmeyer SJ. Altered mental expression of Hox11 and specification of splenic cell fate. HOX expression and segmental identity in MLL-mutant mice. Am J Pathol 1995; 146: 1089–1101. Nature 1995; 378: 505–508. 149 Wallin J, Eibel H, Neubu¨ser A, Wilting J, Koseki H, Balling R. 126 Alkema MJ, van der Lugt NM, Bobeldijk RC, Berns A, van Lohu- Pax1 is expressed during development of the thymus epithelium izen M. Transformation of axial skeleton due to overexpression and is required for normal T-cell maturation. Development 1996; of Bmi-1 in transgeneic mice. Nature 1995; 374: 724–727. 122: 23–30. 127 Saunders WS, Chue C, Goebl M, Craig C, Clark RF, Powers JA, 150 Manley NR, Capecchi MR. The role of Hoxa-3 in mouse thymus Eissenberg JC, Elgin SC, Rothfield NF, Earnshaw WC. Molecular and thyroid development. Development 1995; 121: 1989–2003. cloning of a human homologue of Drosophila heterochromatin 151 Manley NR, Capecchi MR. Hox group 3 paralogues regulate the protein HP1 using anti-centromere autoantibodies with anti- development and migration of the thymus, thyroid and para- chromo specificity. J Cell Sci 1993; 104: 573–582. thyroid glands. Dev Biol 1998; 195: 1–15. 128 van Lohuizen M, Frasch M, Wientjens E, Berns A. Sequence simi- 152 Allen JD, Harris AW, Bath ML, Strasser A, Scollay R, Adams JM. larity between the mammalian Bmi-1 proto-oncogene and the Perturbed development of T and B cells in mice expressing an Drosophila regulatory genes Psc and Su(z)2. Nature 1991; 353: Hlx homeobox transgene. J Immunol 1995; 154: 1531–1542. 353–355. 153 Chariot A, Castronovo V, Le P, Gillet C, Sobel ME, Gielen J. 129 Akasaka T, Tsuji K, Kawahira H, Kanno M, Harigaya K, Hu L, Cloning and expression of a new HOXC6 transcript encoding a Ebihara Y, Nakahata T, Tetsu O, Taniguchi M, Koseiki H. The repressing protein. Biochem J 1996; 319: 91–97. role of mel-18, a mammalian Polycomb group gene, during IL- 154 Vider BZ, Zimber A, Hirsch D, Estlein D, Chastre E, Prevot S, 7-dependent proliferation of lymphocyte precursors. Immunity Gespach C, Yaniv A, Gazit A. Human colorectal carcinogenesis 1997; 7: 135–146. is associated with deregulation of homeobox gene expression. 130 Schumacher A, Faust C, Magnuson T. Positional cloning of a Biochem Biophys Res Commun 1997; 232: 742–748. global regulator of anterior-posterior patterning in mice. Nature 155 Cillo C. HOX genes in human cancers (review). Invas Metast 1996; 383: 250–253. 1994–95; 14: 38–49. 131 Lessard J, Baban S, Sauvageau G. Stage-specific expression of 156 Kongsuwan K, Webb E, Housiaux P, Adams JM. Expression of polycomb group genes in human bone marrow cells. Blood multiple homeobox genes within diverse mammalian haemopo- 1998; 9: 1216–1224. ietic lineages. Embo J 1988; 7: 2131–2138. 132 Nam DK, Honoki K, Yu M, Yunis JJ. Alternative RNA splicing of 157 Shen W-F, Largman C, Lowney P, Corral JC, Detmer K, Hauser the MLL gene in normal and malignant cells. Gene 1996; 178: CA, Simonitch TA, Hack FM, Lawrence HJ. Lineage-restricted 169–175. expression of homeobox-containing genes in human hematopo- 133 Hess JL, Benjamin D, Yi BL, Hanson R, Korsmeyer SJ. Defects in ietic cell lines. Proc Natl Acad Sci USA 1989; 86: 8536–8540. yolk sac hematopoiesis in Mll-null embryos. Blood 1997; 90: 158 Lowney P, Corral J, Detmer K, LeBeau MM, Deaven L, Lawrence 1799–1806. HJ, Largman C. A human Hox 1 homeobox gene exhibits 134 Duboule D, Morata G. Colinearity and functional hierarchy myeloid-specific expression of alternative transcripts in human among genes of the homeotic complexes. Trends Genet 1994; hematopoietic cells. Nucleic Acids Res 1991; 19: 3443–3449. 10: 358–364. 159 Baier LJ, Hannibal MC, Hanley EW, Nabel GJ. Lymphoid 135 Dolle´ P, Izpisu´a-Belmonte J-C, Falkenstein H, Renucci A, expression and TATAA binding of a human protein containing Duboule D. Coordinate expression of the murine Hox-5 complex an Antennapedia homeodomain. Blood 1991; 78: 1047–1055. homeobox-containing genes during limb pattern formation. Nat- 160 Deguchi Y, Moroney JF, Kehrl JH. Expression of HOX-2.3 ure 1989; 342: 767–772. homeobox gene in human lymphocytes and lymphoid tissues. 136 Izpisu´a-Belmonte J-C, Tickle C, Dolle´ P, Wolpert L, Duboule D. Blood 1991; 78: 445–450. Expression of the homeobox Hox-4 genes and the specification 161 Inamori K, Takeshita K, Chiba S, Yazaki Y, Hirai H. Identification of position in chick wing development. Nature 1991; 350: of Homeobox genes expressed in human T-lymphocytes. 585–589. Biochem Biophys Res Commun 1993; 196: 203–208. 137 Krumlauf R. Hox genes in vertebrate development. Cell 1994; 162 Takeshita K, Bollekens JA, Hijiya N, Ratajczak M, Ruddle FH, 78: 191–201. Gewirtz AM. A homeobox gene of the Antennapedia class is 138 Shashikant CS, Utset MF, Violette SM, Wise TL, Einat P, Einat M, required for human adult erythropoiesis. Proc Natl Acad Sci USA Pendleton JW, Schughart K, Ruddle FH. Homeobox genes in 1993; 90: 3535–3538. mouse development. Crit Rev Eukaryot Gene Exp 1991; 1: 163 Zeltser L, Despian C, Heintz N. HOXB-13: a new Hox gene in 207–245. a distant region of the HOXB cluster maintains colinearity. 139 Duboule D, Dolle´ P. The structural and functional organization Development 1996; 122: 2475–2484. of the murine HOX gene family resembles that of Drosophila 164 Taniguchi Y, Komatsu N, Moriuchi T. Overexpression of the homeotic genes. Embo J 1989; 8: 1497–1505. HOX4A (HOXD3) homeobox gene in human erythroleukemia Review JW van Oostveen et al 1688 HEL cells results in altered adhesive properties. Blood 1995; 85: cation in acute myeloid leukaemia fuses the genes for 2786–2794. nucleoporin NUP98 and class I homeoprotein HOXA9. Nat 165 Shen WF, Detmer K, Mathews CH, Hack FM, Morgan DA, Larg- Genet 1996; 12: 159–167. man C, Lawrence HJ. Modulation of homeobox gene expression 185 Lu M, Gong ZY, Shen WF, Ho AD. The tcl-3 proto-oncogene alters the phenotype of human hematopoietic cell lines. Embo J altered by chromosomal translocation in T-cell leukemia codes 1992; 11: 983–989. for a homeobox protein. Embo J 1991; 10: 2905–2910. 166 Helgason CD, Sauvageau G, Lawrence HJ, Langman C, Humphr- 186 Hatano M, Roberts CWM, Minden M, Crist WM, Korsmeyer SJ. ies RK. Overexpression of HOXB4 enhances the hematopoietic Deregulation of a homeobox gene, HOX11, by the t(10;14) in T potential of embryonic stem cells differentiated in vitro. Blood cell leukemia. Science 1991; 253: 79–82. 1996; 87: 2740–2749. 187 Dube´ ID, Kamel-Reid S, Yuan CC, Lu M, Wu X, Corpus G, Rai- 167 Lill MC, Fuller JF, Herzig R, Crooks GM, Gasson JC. The role of mondi SC, Crist WM, Carroll AJ, Minowada J, Baker JB. A novel the homeobox gene, HOX B7, in human myelomonocytic differ- human homeobox gene lies at the chromosome 10 breakpoint entiation. Blood 1995; 85: 692–697. in lymphoid neoplasia with chromosomal translocation t(10;14). 168 Wu J, Zhu J-Q, Zhu D-X, Scharfman A, Lamblin G, Han K-K. Blood 1991; 78: 2996–3003. Selective inhibition of normal murine myelopoiesis ‘in vitro’by 188 Kennedy MA, Gonzalez-Sarmiento R, Kees UR, Lampert F, Dear a HOX 2.3 antisense oligodeoxynucleotide. Cell Mol Biol 1992; N, Boehm T, Rabbitts TH. HOX11, a homeobox-containing T- 38: 367–376. cell oncogene on human chromosome 10q24. Proc Natl Acad 169 Blatt C, Lotem J, Sachs L. Inhibition of specific pathways of Sci USA 1991; 88: 8900–8904. myeloid cell differentiation by an activated Hox-2.4 homeobox 189 Lu M, Zhang N, Ho AD. Genomic organization of the putative gene. Cell Growth Diff 1992; 3: 671–676. human homeobox proto-oncogene HOX-11 (TCL-3) and its 170 Vieille-Grosjean I, Roullot V, Courtois G. Identification of endogenous expression in T cells. Oncogene 1992; 7: 1325– homeobox-containing genes expressed in hematopoietic blast 1330. cells. Biochem Biophys Res Commun 1992; 185: 785–792. 190 Hawley RG, Fong AZ, Reis MD, Zhang N, Lu M, Hawley TS. 171 Moretti P, Simmons P, Thomas P, Haylock D, Rathjen P, Vadas Transforming function of the HOX11/TCL3 homeobox gene. M, D’Andrea R. Identification of homeobox genes expressed in Cancer Res 1997; 57: 337–345. human haemopoietic progenitor cells. Gene 1994; 144: 213– 191 Zhang N, Shen W, Ho AD, Lu M. Three distinct domains in the 219. HOX-11 homeobox oncoprotein are required for optimal trans- 172 Sauvageau G, Lansdorp PM, Eaves CJ, Hogge DE, Dragowska activation. Oncogene 1996; 13: 1781–1787. WH, Reid DS, Largman C, Lawrence HJ, Humphries RK. Differ- 192 Hawley R, Fong AZC, Lu M, Hawley TS. The HOX11 homeobox- ential expression of homeobox genes in functionally distinct containing gene of human leukemia immortalizes murine hema- CD34+ subpopulations of human bone marrow cells. Proc Natl topoietic precursors. Oncogene 1994; 9: 1–12. Acad Sci USA 1994; 91: 12223–12227. 193 Kawabe T, Muslin AJ, Korsmeyer SJ. HOX11 interacts with pro- 173 Giampaolo A, Sterpetti P, Bulgarini D, Samoggia P, Pelosi E, Val- tein phosphatases PP2A and PP1 and disrupts a G2/M cell-cycle tieri M, Peschle C. Key functional role and lineage-specific checkpoint. Nature 1997; 385: 454–458. expression of selected HOXB genes in purified hematopoietic 194 Murre C, McCaw PS, Vaessin H, Caudy M, Jan LY, Jan YN, Cab- progenitor differentiation. Blood 1994; 84: 3637–3647. rera CV, Buskin JN, Hauschka SD, Lassar AB, Baltimore D. Inter- 174 Perkins AC, Cory S. Conditional immortalization of mouse myel- actions between heterologous helix-loop-helix proteins generate omonocytic, megakaryocytic and mast cell progenitors by the complexes that bind specifically to a common DNA sequence. Hox-2.4 homeobox gene. Embo J 1993; 12: 3835–3846. Cell 1989; 58: 537–544. 175 Sauvageau G, Thorsteinsdottir U, Eaves CJ, Lawrence HJ, Larg- 195 Nourse J, Mellentin JD, Galili N, Wilkinson J, Stanbridge E, Smith man C, Lansdorp PM, Humphries RK. Overexpression of HOXB4 SD, Cleary ML. Chromosomal translocation t(1;19) results in syn- in hematopoietic cells causes the selective expansion of more thesis of a homeobox fusion mRNA that codes for a potential primitive populations in vitro and in vivo. Gene Dev 1995; 9: chimeric transcription factor. Cell 1990; 60: 535–545. 1753–1765. 196 Kamps MP, Murre C, Sun X-H, Baltimore D. A new homeobox 176 Helgason CD, Sauvageau G, Lawrence HJ, Largman C, Humphr- gene contributes the DNA binding domain of the t(1;19) translo- ies RK. Overexpression of enhances the hematopoietic cation protein in pre-B ALL. Cell 1990; 60: 547–555. potential of embryonic stem cells differentiated in vitro. Blood 197 Carroll AJ, Crist WM, Parmley RT, Roper M, Cooper MD, Finley 1996; 87: 2740–2749. WM. Pre-B cell leukemia associated with chromosome translo- 177 Thorsteinsdottir U, Sauvageau G, Hough MR, Dragowska W, cation. Blood 1984; 63: 721–724. Lansdorp PM, Lawrence HJ, Largman C, Humphries RK. Over- 198 Kamps MP, Baltimore D. E2A-Pbx1, the t(1;19) translocation pro- expression of hoxa10 in murine hematopoietic cells perturbs tein of human pre-B-cell acute lymphocytic leukemia, causes both myeloid and lymphoid differentiation and leads to acute acute myeloid leukemia in mice. Mol Cell Biol 1993; 13: myeloid leukemia. Mol Cell Biol 1997; 17: 495–505. 351–357. 178 Lawrence HJ, Helgason CD, Sauvageau G. Mice homozygous 199 Di Rocco G, Mavilio F, Zappavigna V. Functional dissection of for a targeted disruption of the homeobox gene HOXA9 have a trasncriptionally active, target-specific Hox-Pbx complex. defects in lymphoid and myeloid hematopoiesis. Blood 1995; EMBO J 1997; 16: 3644–3654. 86: 254a. 200 Krosl J, Baban S, Krosl G, Rozenfeld S, Largman C, Sauvageau 179 Fuller JF, Fraser JK, Gasson JC. Regulation of human myelopoiesis G. Cellular proliferation and transformation induced by HOXB4 by HOX A5. Blood 1995; 86: 255a. and HOXB3 proteins involves cooperation with PBX1. Oncogene 180 Lawrence HJ, Johnson RA, Perrine S. The role of homeobox 1998; 16: 3403–3412. genes in erythropoiesis. Ann NY Acad Sci 1994; 718: 165–180. 201 Lawrence HJ, Sauvageau G, Ahmadi N, Lopez AR, LeBeau MM, 181 Celetti A, Barba P, Cillo C, Rotoli B, Boncinelli E, Magli MC. Link M, Humphries K, Largman C. Stage- and lineage-specific Characteristic patterns of HOX gene expression in different types expression of the HOXA10 homeobox gene in normal and leu- of human leukemia. Int J Cancer 1993; 53: 237–244. kemic hematopoietic cells. Exp Hematol 1995; 23: 1160–1166. 182 Scott GA, Goldsmith LA. Homeobox genes and skin develop- 202 Deguchi Y, Kehrl JH. sequence of a novel diverged ment: a review. J Invest Dermatol 1993; 101: 3–8. human homeobox gene encodes a DNA binding protein. 183 Nakamura T, Largaespada DA, Lee MP, Johnson LA, Ohyashiki Nucleic Acids Res 1991; 19: 3742. K, Toyama K, Chen SJ, Willman CL, Chen I-M, Feinberg AP, Jenk- 203 Deguchi Y, Thevenin C, Kehrl JH. Stable expression of HB24, a ins NA, Copeland NG, Shaughnessy JD. Fusion of the nucleopo- diverged human homeobox gene in T lymphocytes induces rin gene NUP98 to HOXA9 by the chromosome translocation genes involved in T cell activation and growth. J Biol Chem t(7;11)(p15;p15) in human myeloid leukaemia. Nat Genet 1996; 1992; 267: 8222–8229. 12: 154–158. 204 Deguchi Y, Yamanaka Y, Theodossiou C, Najfeld V, Kehrl JH. 184 Borrow J, Shearman AM, Stanton VP, Becher R, Collins T, Willi- High expression of two diverged homeobox genes, HB24 and ams AJ, Dube I, Katz F, Kwong YL, Morris C, Ohyashiki K, Toy- HB9 in acute leukemias: molecular markers of hematopoietic ama K, Rowley J, Housman DE. The t(7;11)(p15;p15) translo- cell immaturity. Leukemia 1993; 7: 446–451. Review JW van Oostveen et al 1689 205 Deguchi Y, Kehrl JH. High level expression of the homeobox adhesion molecule promoter activity is modulated by cotransfec- gene HB24 in a human T-cell line confers the ability to form tion with Hox-2.5 and −2.4. Proc Natl Acad Sci USA 1992; 89: tumors in nude mice. Cancer Res 1993; 53: 373–377. 2086–2090. 206 Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Buchberg AM, Sau- 226 Jones FS, Holst BD, Minowa O, De REM, Edelman GM. Binding vageau G. HOXA9 transforms primary bone marrow cells and transcriptional activation of the promoter for the neural cell through specific collaboration with Meisla but not Pbxlb. EMBO adhesion molecule by HoxC6 (Hox-3.3). Proc Natl Acad Sci USA J 1998; 17: 3714–3725. 1993; 90: 6557–6561. 207 Look AT. Oncogenic transcription factors in the human acute 227 Cillo C, Cantile M, Mortarini R, Barba P, Parmiani G, Antichini leukemias. Science 1997; 278: 1059–1064. A. Differential patterns of HOX gene expression are associated 208 Yu BD, Hanson RD, Hess JL, Horning SE, Korsmeyer SJ. MLL, a with specific integrin and ICAM profiles in clonal populations mammalian trithorax-group gene, functions as a transcriptional isolated from a single human melanoma metatstasis. Int J Cancer maintenance factor in morphogenesis. Proc Natl Acad Sci USA 1996; 66: 692–697. 1998; 95: 10632–10636. 228 Valarche´ I, Tissier-Seta J-P, Hirsch M-R, Martinez S, Goridis C, 209 Corral J, Forster A, Thompson S, Lampert F, Kaneko Y, Slater R, Brunet J-F. The mouse homeodomain protein Phox2 regulates Kroes WG, Vanderschoot CE, Ludwig WD, Karpas A, Pocock C, Ncam promoter activity in concert with Cux/CDP and is a puta- Cotter F, Rabbits TH. Acute leukemias of different lineages have tive determinant of phenotype. Development similar MLL gene fusions encoding related chimeric proteins 1993; 119: 881–896. resulting from chromosomal translocation. Proc Natl Acad Sci 229 Allen JD, Adams JM. Enforced expression of Hlx homeobox gene USA 1993; 90: 8538–8542. prompts myeloid cell maturation and altered adherence proper- 210 Nam DK, Honoki K, Yu M, Yunis JJ. Alternative RNA splicing of ties of T cells. Blood 1993; 81: 3242–3251. the MLL gene in normal and malignant cells. Gene 1996; 178: 230 Koopman G, Kooyk van Y, Graaf de M, Meijer CJLM, Figdor 169–175. CG, Pals ST. Triggering of the CD44 antigen on T lymphocytes 211 van der Lugt N, Alkema M, Berns A, Deschamps J. The poly- promoted the T cell adhesion through the LFA-1 pathway. J comb-group homolog Bmi-1 is a regulator of murine Hox gene Immunol 1990; 145: 3589–3595. expression. Mech Dev 1996; 58: 153–164. 231 Miyake K, Medina KL, Hayasho S-I, Ono S, Hamaola T, Kincade 212 Alkema MJ, Jacobs H, van Lohuizen M, Berns A. Pertubation of PW. Monoclonal to Pgp1/CD44 block lympho-hema- B and T cell development and predisposition to lymphomagen- topoiesis in long-term bone marrow cultures. J Exp Med 1990; esis in Emu Bmil transgenic mice require the Bmil RING finger. 171: 477–485. Oncogene 1997; 15: 899–910. 232 Horst ter E, Meijer CJLM, Radaszkiewicz T, Dongen van JJ, Piet- 213 Jacobs JJL, Kieboom K, Marino S, DePinho RA, van Lohuizen M. ers R, Figdor CG, Hooftman A, Pals ST. Expression of a human The oncogene and Polycomb-group gene bmi-1 regulates cell homing receptor (CD44) in lymphoid malignancies and related proliferation and senescence through the ink4a locus. Nature stages of lymphoid development. Leukemia 1990; 4: 383–389. 1999; 397: 164–168. 233 Guernt U, Hoffman M, Rudt W, Reber S, Zoeller M, Haussman 214 Habeshaw JA. Hypothesis: non-Hodgkin’s lymphomas are abnor- I, Matzku S, Wenzel A, Ponta H, Herrlich P. A new variant of mal immune responses. Cancer Immunol Immunother 1979; 2: glycoprotein CD44 confers metastatic potential to rat carcinoma 37–42. cells. Cell 1991; 65: 13–25. 215 Picker LJ, Treer JR, Ferguson-Darnell B, Collins PA, Buck D, Ter- 234 Jaynes JB, O’Farrell PH. Activation and repression of transcrip- stappen LWMM. Control of lymphocyte recirculation in man. J tion by homoeodomain-containing proteins that bind a common Immunol 1993; 150: 1105–1121. site. Nature 1988; 336: 744–749. 216 Picker LJ, Treer JR, Ferguson-Darnell B, Collins PA, Bergstresser 235 Han K, Levine MS, Manley JL. Synergistic activation and PR, Terstappen LWMM. Control of lymphocyte recirculation in repression of transcription by drosophila homeobox proteins. man. J Immunol 1993; 150: 1122–1136. Cell 1989; 56: 573–583. 217 Picker LJ, Michie SA, Rott LS, Butcher EC. A unique phenotype 236 Krasnow MA, Saffman EE, Kornfeld K, Hongness DS. Transcrip- of skin-associated lymphocytes in humans. Preferential tional activation and repression by ultrabithorax proteins in cul- expression of the HECA-452 epitope by benign and malignant T tured drosophila cells. Cell 1989; 57: 1031–1043. cells at cutaneous sites. Am J Pathol 1990; 136: 1053–1058. 237 Winslow GM, Hayashim S, Krasnow M, Hongness DS, Scott MP. 218 Berg EL, Yoshino T, Rott LS. The cutaneous lymphocyte antigen Transcriptional activation by the antennapedia and fushi tarazu is a skin lymphocyte homing receptor for the vascular lectin proteins in cultured drosophila cells. Cell 1989; 57: 1017–1030. endothelial cell-leukocyte adhesion molecule. J Exp Med 1991; 238 Care´ A, Testa U, Bassani A, Tritarelli E, Montesore E, Samoggia 174: 3247–3252. P, Cianetti I, Peschle C. Coordinate expression and proliferative 219 Picker LJ, Kishimoto TK, Smith CW, Warnock RA, Butcher EC. role of HOXB genes in activated adult T lymphocytes. Mol Cell ELAM-1 is an adhesion molecule for skin-homing T cells. Nature Biol 1994; 14: 4872–4877. 1991; 349: 706–799. 239 Edelman GM, Jones FS. Cytotactin: a morphoregulatory molecule 220 Berlin C, Berg EL, Briskin MJ. alpha4␤7 integrin mediates lym- and a target for regulation by homeobox gene products. Trends phocyte binding to the mucosal vascular addressin MadCAM-1. Biochem Sci 1992; 17: 228–232. Cell 1993; 74: 185–195. 240 Drillenburg P, Vandervoort R, Koopman G, Dragosics B, Vank- 221 Briskin MJ, McEvoy LM, Butcher EC. MadCAM-1 has homology rieken JHJM, Kluin P, Meenan J, Lazarovits AI, Radaszkiewicz T, to immunoglobulin and mucin-like adhesion receptors and to Pals ST. Preferential expression of the mucosal homing receptor IgA1. Nature 1993; 363: 461–465. integrin alpha(4)beta(7) in gastrointestinal non-hodgkin lym- 222 Bruin de PC, Kummer JA, Valk van der P, Heerde van P, Kluin phomas. Am J Pathol 1997; 150: 919–927. PM, Willemze R, Ossenkoppele GJ, Radaszkiewicz T, Meijer 241 Pals ST, Drillenburg P, Radaszkiewicz T, Manten-Horst E. CJLM. Granzyme B expressing peripheral T-cell lymphomas: Adhesion molecules in the dissemination of non-Hodgkin’s lym- neoplastic equivalents of activated cytotoxic T cells with prefer- phomas. Acta Haematol 1997; 97: 73–80. ence for mucosa associated lymphoid tissue localization. Blood 242 Wang W, Wu W, Desai T, Ward DC, Kaufman SJ. Localization 1994; 84: 3785–3791. of the ␣7 integrin gene (ITGA7) on human chromosome 12q13: 223 Bijl JJ, Rieger E, Van Oostveen JW, Walboomers JMM, Kreike M, clustering of integrin and Hox genes implies parallel evolution Willemze R, Meijer CJLM. HOXC4, HOXC5 and HOXC6 of these gene families. Genomics 1995; 26: 563–570. expression in primary cutaneous lymphoid lesions. High 243 Manak JR, Scott MP. A class act: conservation of homeodomain expression of HOXC5 in anaplastic large-cell lymphomas. Am J protein functions. Development Sup 1994; 61–71. Pathol 1997; 151: 1067–1074. 244 Johnson PF, McKnight SL. Eukaryotic transcriptional regulatory 224 Blatt C, Lotem J, Sachs L. Inhibition of specific pathways of proteins. Annu Rev Biochem 1989; 58: 799–839. myeloid cell differentiation by an activated Hox-2.4 homeobox 245 Rehorn KP, Thelen H, Michelson AM, Reuter R. A molecular gene. Cell Growth Diff 1992; 3: 671–676. aspect of hematopoiesis and endoderm development common 225 Jones FS, Prediger EA, Bittner DA, De REM, Edelman GM. Cell to vertebrates and drosophila. Development 1996; 122: 4023– adhesion molecules as targets for Hox genes: neural cell 4031. Review JW van Oostveen et al 1690 246 Robb L, Begley CG. The helix-loop-helix gene SCL-implicated in 258 Guazzi S, Loigro R, Pintonello L, Boncinelli E, Di Lauro R, Mav- T-cell acute lymphoblastic leukaemia and in normal haematopo- illo F. The thyroid transcription factor-1 gene is a candidate target ietic development. Int J Biochem Cell Biol 1996; 28: 609–618. for regulate by Hox proteins. Embo J 1994; 13: 3339–3347. 247 Porcher C, Swat W, Rockwell K, Fujiwara Y, Alt FW, Orkin SH. 259 Zhao G-Q, Zhou X, Eberspaecher H, Solursh M, Crombrugghe The T cell leukema oncoprotein SCL/TAL-1 is essential for devel- B. Cartilage homeoprotein 1, a homeoprotein selectively opment of all hematopoietic lineages. Cell 1996; 86: 47–57. expressed in chondrocytes. Proc Natl Acad Sci USA 1993; 90: 248 De Groot R, Foulkes N, Mulder M, Kruijer W, Sassone-Corsi P. 8633–8637. Positive regulation of Jun/AP-1 by E1A. Mol Cell Biol 1991; 11: 260 Lai E, Prezioso VR, Tao W, Chen WS, Darnell JJ. Hepatocyte 192–201. nuclear factor 3alpha belongs belongs to a gene family in mam- 249 Scheidler S, Fredericks WJ, Rauscher FJ, Barr FG, Vogt PK. The mals that is homologous to the forkhead. Genes hybrid Pax3-FKHR fusion protein of alveolar rhabdomyosarcoma Dev 1991; 5: 416–427. transforms fibroblasts in culture. Proc Natl Acad Sci USA 1996; 261 Wagner-Bernholz JT, Wilson C, Gibson G, Shuh R, Gehring W. 93: 9805–9809. Identification of target genes of the homeotic gene Antennapedia 250 Hromas R, Klemsz M. The ETS oncogene family in development, by enhancer detection. Genes Dev 1991; 5: 2467–2480. proliferation and neoplasia (review). Int J Hematol 1994; 59: 262 Violette SM, Shashikant CS, Salbaum JM, Belting H-G, Wang JC, 257–265. Ruddle FH. Repression of the ␤-amyloid gene in a Hox-3.1-pro- 251 Callaerts P, Halder G, Gehring WJ. PAX-6 in development and ducing cell line. Proc Natl Acad Sci USA 1992; 89: 3805–3809. evolution (review). Ann Rev Neurosci 1997; 20: 483–532. 252 Clevers HC, Owen MJ. Towards a molecular understanding of 263 Zannini M, Francis-Lang H, Plachov D, Di Lauro R. Pax-8, a T-cell differentiation. Immunol Today 1991; 12: 86–92. paired domain-containing protein, binds to a sequence overlap- 253 Valarche´ I, Tissier-Seta J-P, Hirsch M-R, Martinez S, Goridis C, ping the recognition site of a homeodomain and activates tran- Brunet J-F. The mouse homeodomain protein Phox2 regulates scription from two thyroid-specific promoters. Mol Cell Biol Ncam promoter activity in concert with Cux/CDP and is a puta- 1992; 12: 4230–4241. tive determinant of neurotransmitter phenotype. Development 264 Capovilla M, Brandt M, Botas J. Direct regulation of decapenta- 1993; 119: 881–896. plegic by Ultrabithorax and its role in midgut morphogenesis. 254 Chalepakis G, Tremblay P, Gruss P. , mutants and mol- Cell 1994; 76: 461–475. ecular function. J Cell Sci 1992; 16: 61–67. 265 Warren AJ, Colledge WH, Carlton MBL, Evans MJ, Smith AJH, 255 Goomer RS, Holst BD, Wood IC, Jones FS, Edelman GM. Regu- Rabbits TH. The oncogenic -rich LIM domain protein lation in vitro of an L-CAM enhancer by homeobox genes HoxD9 Rbtn2 is essential for erythroid development. Cell 1994; 78: and HNF-1. Proc Natl Acad Sci USA 1994; 91: 7985–7989. 45–57. 256 Tomotsune D, Shoji H, Wakamatsu Y, Kondoh H, Takahashi N. 266 Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. A mouse homoloque of the Drosophila tumour-suppressor gene AML1, the target of multiple chromosomal translocations in l(2)gl controlled by Hox-C8 in vivo. Nature 1993; 365: 69–72. human leukemia, is essential for normal fetal liver hematopo- 257 Gould AP, White R. Connectin, a target of homeotic gene control iesis. Cell 1996; 84: 321–330. in Drosophila. Development 1992; 116: 78–80.