THIS STORY BEGINS with a seminal case report describing a 16-year-old boy who presented with acute lymphoblastic leukemia (ALL).1 He had an elevated white blood cell (WBC) count, thymic enlargement, and leukemic cells with an early T-cell phenotype (CD2 and CD7 positive; CD3, CD4 and CD8 negative). Standard chemotherapeutic agents failed to induce remission, and treatment commenced with the adenosine deaminase inhibitor 2′-deoxycoformycin. Over the next 7 days, the leukemic cells underwent an abrupt and dramatic transformation. The lymphoid leukemic cells acquired morphological features of myeloid cells, gained myeloid antigens, and lost CD7 expression. Unfortunately, the patient died of extensive tissue infiltration with cells of promyelocyte morphology. This “stem cell leukemia” phenotype was also evident in vitro because exposure of the leukemic cells to 2′-deoxycoformycin stimulated multi-lineage differentiation. Before and after the phenotypic conversion, the leukemic cells carried a translocation between chromosome 1p32-33 and the T-cell receptor (TCR) α/δ locus on chromosome 14q11.

The investigators established a cell line that carried the same chromosomal translocation and recapitulated the stem cell phenotype during in vitro culture.2 They further showed that other cases of ALL, with a similar cell surface phenotype, did not show this specific cytogenetic abnormality.3 Although rearrangements of chromosome 1p32-33 are now recognized to occur frequently in T-cell ALL, there have been no reports of these leukemias being treated with similar agents, nor reports of such a striking morphological conversion. Based on what is now known about the molecular genetics of this event, it seems reasonable to speculate that other cases may respond in a similar manner and that ultimately this might be exploited for therapeutic advantage. However, there is still no clear explanation for the mechanism by which 2′-deoxycoformycin induced this dramatic change.

At that time, the recent successes in identifying genes involved in B-cell tumors as a result of their translocation into the Ig gene loci inspired a search for the gene responsible for the stem cell phenotype. By analogy with B-cell tumors, it seemed likely that the responsible gene was located on chromosome 1 and, as a result of the translocation into the TCR locus, it was aberrantly activated when transcription of the TCR locus was attempted. However, analysis of the translocation first required characterization of the TCR α and δ loci and recognition that the α locus contained the δ locus embedded within it.4-8 Once probes to the human TCR δ locus became available, the translocation breakpoint was isolated and a new gene on chromosome 1 was identified.9,10 The name “stem cell leukemia” gene or SCL was assigned, a name that has turned out to be remarkably appropriate given the essential function of this gene in normal hematopoietic stem cells (see below). The same gene was independently identified by several other groups.11-13 

The t(1;14) translocation was the first example of what has since transpired to be a common theme in a significant proportion of T-ALL cases; the aberrant activity of recombinase enzymes with abnormal expression of SCL as a result.

The leukemic cells were captured at a very early stage of T-cell development and frozen while in the process of rearranging/deleting both their TCR δ loci. On one chromosome 14 the cells were caught in the process of attempting to delete the entire TCR δ locus.14 Deletion of the TCR δ locus is a necessary prerequisite for TCR α rearrangement15 and although this transient intermediate state had been predicted to occur (ie, δ deleted but incomplete α rearrangement),16 it had not actually been previously observed.

The second chromosome 14 was the site of the translocation event.9 Here the recombinase enzymes had successfully juxtaposed two TCR δ diversity (or D region) genes and completed the required excision of the intervening DNA. However, during or immediately after this event, chromosome 1 had been inadvertently involved in this otherwise normal recombination process. The presence of additional, nontemplated nucleotides provided circumstantial evidence implicating enzymes such as terminal deoxytidyl transferase (TDT) as also being active at that moment.

There are mitigating circumstances that in part explain the “error” on the part of the recombinase enzymes. The recombinases normally recognize specific DNA sequences that flank the segments of DNA to be rearranged. These serve as specific signals that allow the enzymes to correctly identify and juxtapose segments of DNA in the process of assembling a functional TCR (or Ig) gene. Very similar sequences are present in several regions of the human SCL gene, but intriguingly are not seen in equivalent regions of the mouse gene.17-19 Their normal function in the human SCL gene, if any, is unknown. However, the presence of “signal-sequences” in the human SCL gene does provide an explanation for the case of mistaken identity in this chromosomal translocation. There is a second reason that perhaps also partly explains this event. It is now known that SCL is crucially important at the earliest stages of hematopoietic stem cell development and differentiation. Therefore, the SCL gene was likely to be hypomethylated in an “open,” chromatin configuration, thus making it vulnerable to the recombinase enzymes.20 

This patient’s translocation disrupted the 3′ untranslated region of the SCL gene, preserving the SCL protein intact and thus allowing production of the normal protein to be inappropriately regulated by elements within the TCR δ locus. As a consequence, SCL expression was readily detected within the leukemic T cells while SCL is not detected in the vast majority of normal thymocytes.10 21-24 

Other investigators had also noted rare cases of T-ALL with translocations between chromosomes 1 and 14 occurring in about 3% of cases,25-27 and Bernard et al13 went on to show that SCL was also dysregulated in some of these cases. At least 10 chromosomal translocations involving SCL have since been defined, with the majority of these occurring into the TCR α/δ locus.12,28-31 However, translocations involving SCL and chromosomes 3, 5, and 7 (the TCR β locus) have also been documented.32-34 These translocations all have in common the errant expression of SCL in a T-cell environment. In addition, the fact that breakpoints within the SCL gene lie at the site of signal sequences strongly suggests that the recombinase enzyme complex is again likely to be the primary culprit. Because of the frequent involvement of SCL in leukemias with a T-cell phenotype, the names TCL-5 and TAL-1 were also proposed.11,12 30 

In addition to chromosomal translocations, there is a second mechanism by which the SCL gene can be disrupted and aberrantly activated in leukemic T lymphocytes. This involves deletions of approximately 90 kb of DNA from chromosome 1 that brings SCL under the control of the upstream, ubiquitously expressed SIL (for SCL interrupting locus) gene.35-38 Again, recombinase enzymes are the primary suspect in the genesis of the SIL/SCL deletions (also known as Tald): the presence of specific recombination signal sequences in both the SIL and SCL genes ensure that a limited number of SIL/SCL deletional events are seen recurrently in independent leukemic samples. Although this “limited number” could theoretically be as high as 18, with 3 recombination sites observed within the SIL gene and at least 6 different recombination sites observed within SCL, only two types of deletion are commonly observed. The SIL/SCL deletional event in T-cell leukemia occurs with a frequency estimated to be between 6% and 26% of cases.35,39-43 The generation of unique sequences at the site of the SIL/SCL junction can serve as a useful leukemia-specific and patient-specific marker for polymerase chain reaction (PCR)-based techniques to detect residual leukemic cells.44 More recently it was proposed that SCL may be aberrantly expressed in up to 60% of childhood T-ALL samples in many cases without detectable rearrangements of the gene.45 This potentially provided an explanation for earlier studies that failed to identify a significant clinical, phenotypic, or prognostic subgroup of T-ALL associated with SCL rearrangement. However, it has been suggested that SCL expression observed in the absence of SCL rearrangements was probably caused by contamination of the leukemic blasts with normal hematopoietic cells.46 

Despite the lack of a clearly distinct clinical subgroup of T-ALL associated with SCL rearrangement, there is one characteristic that is highly correlated with the presence of the SIL/SCL deletion; deletion of one or both TCR δ gene(s). Therefore, SCL rearrangements are observed predominantly, but not exclusively, in CD3 positive, TCR α/β positive T-ALL.41,42,47 The striking concordance with deletion of the TCR δ locus, a prerequisite for TCR α rearrangement, raises the possibility that SCL may even play a role in regulating deletion of the TCR δ loci and in commitment to the TCR α/β T-cell lineage.20 47 

Although the structural alterations of the SCL gene are seen in T-ALL, they are not seen in B-lineage ALL, acute myeloid leukemia (AML), T-cell non-Hodgkin’s lymphomas, or in a range of other solid tumors.43,44,48 Although expression of SCL has been observed in samples of AML and may even correlate with poor outcome,49 this probably reflects malignant transformation of myeloid progenitor/stem cells that normally express SCL.23,24 50 

Although the genetic lesions outlined above result in ectopic expression of SCL in a T-cell environment, the precise mechanism(s) by which this transcription factor contributes to leukemia development remains uncertain.

There is clear evidence that SCL can provide a proliferative advantage to cells in a number of different contexts. Expression of antisense SCL in a multipotent human cell line caused a significant decrease in cellular proliferation, decreased progression of cells through the cell cycle, and resulted in a 50-fold decrease in self-renewal potential, thus suggesting that SCL normally influences these functions.51 Similarly, aberrant SCL expression in a T-cell environment increased clonogenic ability.52 In murine myeloid cells, retrovirally enforced SCL expression allowed cells to “escape” differentiation induction and overcome growth-factor–induced suppression of clonogenicity.53-55Likewise, SCL expression as a consequence of an insertion of a retroviral-like element, an intra-cisternal A particle, conferred a growth advantage on myeloid cells and resistance to differentiation-inducing agents.56,57 Thus, in these various contexts enforced SCL expression stimulates proliferation and opposes differentiation. Within a different cellular context, erythroid cells, enforced SCL expression also enhances cell proliferation, although in this setting differentiation is stimulated.58-60 In addition to its action to stimulate proliferation in a variety of contexts, SCL expression delayed apoptosis55,61,62; again, this action may depend on the cell type in which SCL is acting.55 Therefore, as with other oncogenes, it is likely that SCL contributes to leukemogenesis by multiple mechanisms that include an increase in proliferation, cell cycling, and self-renewal potential with a decrease in cell death.

These actions of SCL translate into increased leukemia development and subsequent death in a variety of animal models. In some transgenic models expression of SCL alone did not cause leukemia to occur, presumably as a consequence of the regulatory elements that were used to direct SCL expression.63-65 However, several groups have demonstrated cooperation in vivo between SCL and a variety of additional genes including ABL,52 casein kinase II,66 p53,62,67 and RAS.67 Whether any of these cooperating genes contribute to human T-ALL is unclear. However, SCL also collaborates with the LIM domain proteins LMO-1 and LMO-2 to generate tumors.64,65 This interaction is particularly intriguing because SCL, LMO-1, and LMO-2 are implicated in human T-ALL. However, all the mouse models suggest that additional events are still needed, and in the context of SCL and the LIM domain proteins, defects in the DNA mismatch repair gene MSH2 are probably important.68 

The biochemical basis for the tumorigenic cooperation described above remains unclear. However, it is known that SCL interacts with the products of the E2A gene69-71 and related proteins72 to form heterodimeric DNA-binding complexes. The heterodimer can form part of a larger protein complex in which LMO-2 acts as a molecular bridge between the zinc finger transcription factor GATA-1 and the SCL/E2A heterodimer (Fig1).73,74 Using a chromatin immuno-precipitation approach, Cohen-Kaminsky et al75 have recently identified a novel gene of unknown function that is expressed in erythroid cells and that is directly regulated by a complex containing SCL and GATA-1. Although these interactions have been characterized primarily in erythroid cells, where SCL is normally expressed, a large protein complex occurs in T cells, but with altered DNA binding specificity.76 The existence of such multi-component protein complexes is an emerging theme in the area of transcriptional regulation. The constituents of the complex are likely to vary in different cell types, differentiation states, or cell cycle stages, perhaps explaining in part the multiplicity of functions ascribed to SCL and other transcription factors.

Fig. 1.

(A) SCL forms part of a multiprotein complex. SCL normally interacts with other transcription factors in erythroid cells to form a multiprotein complex that contains transcription factors such as GATA-1, LMO-2, LDB-1, and E2A proteins.74 As a result, “Gene A” is appropriately regulated. The precise stoichiometry of the complex is not known. (B and C) Models for the leukemogenic effect of SCL expression in T cells. (B) The leukemogenic effect of ectopic SCL expression in T cells may involve aberrant activation of target genes (Gene B), perhaps by a complex involving additional bHLH proteins (eg, X and Y).76 (C) Alternatively ectopic SCL expression may sequester E2A and/or other T-cell proteins (eg, Z), thereby upregulating or downregulating expression of their normal target genes (Gene C).

Fig. 1.

(A) SCL forms part of a multiprotein complex. SCL normally interacts with other transcription factors in erythroid cells to form a multiprotein complex that contains transcription factors such as GATA-1, LMO-2, LDB-1, and E2A proteins.74 As a result, “Gene A” is appropriately regulated. The precise stoichiometry of the complex is not known. (B and C) Models for the leukemogenic effect of SCL expression in T cells. (B) The leukemogenic effect of ectopic SCL expression in T cells may involve aberrant activation of target genes (Gene B), perhaps by a complex involving additional bHLH proteins (eg, X and Y).76 (C) Alternatively ectopic SCL expression may sequester E2A and/or other T-cell proteins (eg, Z), thereby upregulating or downregulating expression of their normal target genes (Gene C).

Close modal

The leukemogenic effect of SCL expression in T cells may reflect either activation of target genes that should normally be silent, or alternatively the sequestration of other components of the multi-protein complex (Fig 1). Several lines of circumstantial evidence support the latter view: (1) SCL can sequester E2A proteins, thereby inhibiting transactivation of genes that normally require E2A proteins.77-79 (2) Mice lacking a functional E2A gene develop T-cell tumors.80 (3) As mentioned above, casein kinase II cooperates with SCL to hasten T-cell tumors,66and casein kinase II is also known to inhibit activity of proteins derived from the E2A gene.81 (4) Mice carrying an SCL transgene lacking its transactivation domain still develop lymphoid tumors65 (although it remains possible that SCL/E2A heterodimers aberrantly transactivate critical target genes via the E2A transactivation domain).

If SCL does act as a molecular sink in T-ALL, one would predict that this property would not be unique to SCL and that other transcription factors capable of interacting with E2A proteins would also be leukemogenic if ectopically expressed in T cells. The proteins LYL-1 and TAL-2 proteins share this characteristic and are indeed involved in translocations in T-ALL, but only rarely.82 Therefore, the particular prevalence of SCL rearrangements in T-ALL probably reflects the presence of the specific signal sequences in the human SCL gene that are recognized by the recombinase complex and dictate the site of translocation and rearrangement events.

Given the unusual phenotype of the leukemic cells from which SCL was first identified, it seemed possible that the protein encoded by the SCL gene would also prove an important molecule in normal hematopoietic cells. The first evidence that this was the case came from analysis of the amino acid sequence for the predicted SCL protein.9 Two important domains within the protein were immediately apparent. One domain is a helix-loop-helix (HLH) region that is common to a large family of functionally important transcription factors. This region allows SCL to heterodimerize with E12 and E47, products of the E2A gene and themselves HLH transcription factors. In fact, the E2A gene [which is involved in the t(1;19) translocation in human B-ALL] was a founding member of the HLH family along with C-MYC [involved in the t(8;14) translocation of Burkitt’s lymphoma], MYO-D (an important regulator of muscle differentiation), and daughterless (aDrosophila gene involved in sex determination).83 

The second conspicuous domain within the SCL protein is a basic region, which is also present in many other HLH proteins: the SCL/E2A heterodimer binds DNA using the basic domains present within these proteins.84 The site on the DNA to which the SCL/E2A heterodimer binds conforms to an “E-box” motif, which is also common to other proteins of the HLH class.83 85 

A role for SCL in the regulation of normal hematopoiesis was suggested by its normal expression pattern; SCL is restricted to hematopoietic cells, endothelial cells, the central nervous system, and embryonic skeleton.23,86,87 Within the hematopoietic compartment, SCL is normally expressed within erythroid cells, mast cells, megakaryocytes, and progenitor/“stem” cells.10,21-24 50 

The first clue to a normal function of the SCL protein came from antisense experiments showing that SCL regulates proliferation and self-renewal in a multipotent hematopoietic cell line.51 A positive role for SCL in erythroid differentiation was suggested by the observation that SCL mRNA levels increased during erythroid differentiation induced either by chemicals or erythropoietin,22,23,50,88-90 although a concurrent decrease in levels of SCL protein implies additional levels of control.91 A more direct role for SCL in erythroid differentiation was provided by gene delivery experiments where enforced expression of SCL enhanced erythroid differentiation of hematopoietic cell lines.58,90 Conversely, as cells differentiated along the myeloid/macrophage pathway, levels of SCL mRNA and protein decreased, becoming essentially undetectable.50,53,54,90,92 The decrease in SCL expression not only accompanies macrophage differentiation, but is also necessary because enforced SCL expression prevents macrophage differentiation from proceeding normally.53,54,90 These results in cell lines have recently been confirmed and extended using normal human CD34-positive “stem” cells. In these cells, retrovirally directed SCL expression enforces and hastens erythroid differentiation and also directs megakaryocytic differentiation, with less striking effects on the myeloid compartment.59 60 

Dramatic confirmation that the SCL gene encodes a vital stem cell regulator came from studies in which SCL function was destroyed (Fig 2). This was achieved using gene targeting in embryonic stem (ES) cells to derive SCL-null mutant animals (“knock-out” mice). Mice lacking SCL function died at about 8.5 days of embryonic development (E8.5) with a complete failure of yolk sac hematopoiesis,93,94 suggesting that SCL is crucial for the generation of hematopoietic cells in the early embryo (“primitive hematopoiesis”). SCL plays a similar essential role in adult, “definitive” hematopoiesis. After injection of SCL-null ES cells into blastocysts, analysis of the resulting chimeric animals showed the complete failure of the SCL-null ES cells to give rise to hematopoietic cells in the adult animals, although the mutant ES cells did contribute to all other tissues examined.95,96Furthermore, these SCL-null ES cells failed to give rise to hematopoietic cells during in vitro culture and failed to express hematopoietic-specific genes.97 Taken together, these observations show that SCL plays a critical role in the generation and/or subsequent behavior of pluripotent hematopoietic stem cells. This role is “cell-autonomous,” and occurs within the stem cell itself. These results also predict that normal SCL-expressing progenitors should function as multipotent cells, displaying the ability to generate hematopoietic cells committed to multiple lineages. This prediction has recently been confirmed using a gene-targeting approach; a marker gene (Lac-Z) was introduced into the SCL locus, thereby allowing the isolation of SCL-expressing cells for functional analysis.98 These results confirm the status of SCL as a “master regulator” for hematopoiesis.99 

Fig. 2.

SCL is required for development of hematopoietic stem cells. (A) Wild type (+/+) and SCL mutant (−/−) mouse embryos. Note the red-brown coloration of the normal embryo due to blood within the fetal liver and vessels. This is absent in the mutant which also has a runted appearance. (B) Histological section through the mouse yolk sac with blood islands (arrows) present in the wild-type and lacking in the SCL mutant yolk sac. (C) Histological section through the embryo showing hematopoietic cells in the paired dorsal aortae in the wild type (arrows) and the absence of these cells in the dorsal aortae of the SCL mutant (arrows). (Reprinted with permission from Robb L, Lyons I, Li R, Hartley L, Kontgen F, Harvey RP, Metcalf D, Begley CG: Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene. The Proceedings of the National Academy of Sciences, U.S.A., vol 92, p 7075, 1995. Copyright 1995 National Academy of Sciences, U.S.A.93)

Fig. 2.

SCL is required for development of hematopoietic stem cells. (A) Wild type (+/+) and SCL mutant (−/−) mouse embryos. Note the red-brown coloration of the normal embryo due to blood within the fetal liver and vessels. This is absent in the mutant which also has a runted appearance. (B) Histological section through the mouse yolk sac with blood islands (arrows) present in the wild-type and lacking in the SCL mutant yolk sac. (C) Histological section through the embryo showing hematopoietic cells in the paired dorsal aortae in the wild type (arrows) and the absence of these cells in the dorsal aortae of the SCL mutant (arrows). (Reprinted with permission from Robb L, Lyons I, Li R, Hartley L, Kontgen F, Harvey RP, Metcalf D, Begley CG: Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene. The Proceedings of the National Academy of Sciences, U.S.A., vol 92, p 7075, 1995. Copyright 1995 National Academy of Sciences, U.S.A.93)

Close modal

The results with SCL are remarkably similar to those seen when the LMO-2 gene is destroyed. LMO-2 null animals also lack any detectable hematopoietic cells within the yolk sac and embryo, and die at about E8.5.100 Similarly, LMO-2 null ES cells fail to contribute to adult hematopoiesis,101 thus reinforcing the parallels between SCL and LMO-2. In addition to interacting in erythroid and some leukemic T cells, it therefore seems likely that SCL and LMO-2 are components of a critical multiprotein complex present in hematopoietic stem cells.

Thus, the phenotype of the initial stem cell leukemia implied SCL was capable of regulating hematopoietic differentiation in the leukemic cells. Pursuit of this early clue has unmasked a master gene which functions as a critical regulator of hematopoiesis.

Given the crucial function of SCL within hematopoietic stem cells, the expression of SCL within endothelial cells raised the possibility that SCL could also function within an even more primitive cell, the putative common hematopoietic and endothelial precursor, the hemangioblast. There is a long-recognized, close relationship between the development of blood and endothelium, suggesting that they may well share a common precursor. Both cell types emerge at a similar time during the formation of the yolk sac blood islands,102-104and early intra-embryonic sites of hematopoiesis and are closely associated with vessels.105-111 In addition, a number of molecules are expressed in both endothelium and in hematopoietic progenitors including CD34.112,113 Furthermore, mice lacking the early endothelial marker Flk-1 display a defect in both hematopoietic cells and vasculature,114 and Flk-1 appears to play an important and cell-autonomous role in the formation of both lineages.115 Consistent with this concept VEGF, the ligand for Flk-1, can stimulate the generation of hematopoietic cells from ES cells116 and single Flk-1 positive avian cells can generate either hematopoietic or endothelial cells in vitro.117Moreover, in the zebrafish mutant cloche, the numbers of both endothelial and hematopoietic cells are severely reduced.118 These studies are all consistent with the notion of a common precursor for hematopoietic and endothelial cells.

Several lines of evidence suggest a functional role for SCL in endothelial cell development. SCL is expressed in normal endothelium86,119 and in SCL-null animals there is a defect in the development of yolk sac blood vessels with the failure of formation of large vitelline vessels.93 This was shown to reflect a primary consequence of SCL loss in elegant transgenic experiments that rescued the hematopoietic, but not the vascular defect, in the SCL-null mice.120 Furthermore, in the zebrafish mutant cloche, both the endothelial and hematopoietic defects are partially rescued by ectopic expression of SCL.121 

Direct evidence for a role for SCL in hemangioblast formation has been reported by Gering et al122(Fig 3). SCL was coexpressed with Flk-1 in presumptive hemangioblasts within early postero-lateral mesoderm of zebrafish embryos. Ectopic expression of SCL mRNA during zebrafish development resulted in an expansion of hematopoietic and endothelial precursors at the expense of somitic and pronephric duct tissues. Taken together, these data show that SCL is capable of specifying or directing hemangioblast development from early mesoderm, and underline the striking similarities between the role of SCL in hematopoiesis/vasculogenesis and the function of other HLH proteins in muscle and neural development.123 124 

Fig. 3.

SCL specifies hemangioblast development from early zebrafish mesoderm. Appearance of uninjected zebrafish embryos (a) and embryos injected with SCL mRNA at the 2-4 cell stage (b). SCL injected embryos developed a marked excess of hemoglobinized blood cells (arrowhead), a striking absence of blood circulation despite a beating heart and an abnormally curved axis. In situ hybridization experiments with SCL alone (c) or SCL and Flk-1 (d) showed a marked increase in cells coexpressing both endogenous SCL and Flk-1 in SCL-injected embryos. This increase was often unilateral (as illustrated in this figure), because the injected mRNA was frequently localized to one half of an embryo. NT, neural tube; NC, notochord; SM, somitic mesoderm; LM, lateral mesoderm. (Reprinted from Gering M, Rodaway ARF, Göttgens B, Patient RK, Green AR: The SCL gene specifies haemangioblast development from early mesoderm. The EMBO Journal, vol 17, p 4029, 1998, by permission of Oxford University Press.122)

Fig. 3.

SCL specifies hemangioblast development from early zebrafish mesoderm. Appearance of uninjected zebrafish embryos (a) and embryos injected with SCL mRNA at the 2-4 cell stage (b). SCL injected embryos developed a marked excess of hemoglobinized blood cells (arrowhead), a striking absence of blood circulation despite a beating heart and an abnormally curved axis. In situ hybridization experiments with SCL alone (c) or SCL and Flk-1 (d) showed a marked increase in cells coexpressing both endogenous SCL and Flk-1 in SCL-injected embryos. This increase was often unilateral (as illustrated in this figure), because the injected mRNA was frequently localized to one half of an embryo. NT, neural tube; NC, notochord; SM, somitic mesoderm; LM, lateral mesoderm. (Reprinted from Gering M, Rodaway ARF, Göttgens B, Patient RK, Green AR: The SCL gene specifies haemangioblast development from early mesoderm. The EMBO Journal, vol 17, p 4029, 1998, by permission of Oxford University Press.122)

Close modal

It is interesting to contrast the loss of function phenotype, in which endothelial cells develop but fail to contribute normally to vessel formation, with the gain of function experiments described above. These results are consistent with previous studies of HLH proteins in myogenesis and neurogenesis, suggesting a cascade of HLH proteins that may have overlapping functions and expression patterns.123 124 In the case of SCL, functional redundancy with related HLH proteins may explain the relatively normal development of endothelial cells in SCL null mice.

Multiple tiers of regulation.

Although multiple mechanisms exist and at different levels, to ensure that the critical functions of SCL are tightly controlled, these mechanisms and their significance are poorly understood. However, the importance of these mechanisms is amply demonstrated when they go awry in the T lymphocyte.

At the level of SCL protein, there is a requirement for HLH partner proteins such as E12 and E47. Additional control is provided via a subgroup of HLH proteins, the prototypic example of which is Id. These HLH family members lack a DNA-binding basic domain and thereby isolate other HLH proteins in inactive complexes.125 Moreover, the SCL protein itself exists in several different forms as a result of multiple sites of protein initiation within the SCL gene.17-19,28,126-128 These proteins can occur in a cell-type–specific manner and may differ in their ability to transactivate downstream genes. Regulation of protein turnover91 and phosphorylation on serine127 are probably also important in regulating the function of SCL protein.

Signaling pathways.

Several lines of evidence suggest links between SCL and a number of signal transduction pathways. During early development the formation of blood progenitors is thought to be initiated by growth factors, including bone morphogenetic protein 4 (BMP-4). During hematopoietic development in Xenopus, SCL expression is induced by BMP-4 and its expression is inhibited by a dominant-negative BMP-4 receptor.129 However, the precise position of SCL within the hierarchy of transcription factors modulated by BMP-4 is not known.

In the context of adult hematopoiesis, SCL has been linked to growth factor signaling pathways that suggest a “downstream” role for SCL. During erythroid differentiation, levels of SCL increase in response to stem cell factor130,131 and erythropoietin.88,89 Conversely, levels of SCL mRNA and protein decrease during growth-factor–induced macrophage differentiation.53,54 SCL is also a target for MAP kinases, including ERK-1.132 

By contrast, antisense and dominant-negative SCL constructs reduced levels of c-kit (the receptor for stem cell factor) and SCL transactivated the c-kit promoter.134 These data raise the possibility that c-kit is regulated by SCL.

These observations are relevant to current views of hematopoietic stem cell behavior and lineage commitment. Several lines of evidence suggest that multiple components of distinct differentiation programs may be primed and undergo low-level transcription within individual multipotent stem cells.135 Commitment to a particular lineage may result from poorly understood and possibly stochastic processes that result in a single program becoming dominant and reinforced with competitive feedback loops. Transcription factors play a pivotal role in these processes. Thus, upregulation of c-kit or other growth factor receptors by SCL may modulate the behavior of multipotent cells and, as part of a self-sustaining feedback loop, participate in the lineage-commitment process.

Cis-acting elements.

The functions of many transcription factors hinge on their cellular context, the presence or absence of interacting proteins, and the accessibility of potential target genes. Therefore, the biological function of a given transcription factor is likely to depend critically upon its pattern of expression. This reasoning has focused attention on the cis-acting elements that regulate SCL transcription. Characterization of SCL regulatory elements that drive expression in hemangioblasts or multipotent progenitors will illuminate the molecular basis for the formation of these cell types and also provide important tools for experimental or therapeutic manipulation of hematopoietic stem cells.

Two promoters in alternative 5′ exons have been identified in both murine and human SCL genes,17,19,136-138 together with a third promoter within the body of the gene, the normal function of which remains unclear.133 Both 5′ promoters exhibit lineage restricted activity. Promoter 1a is regulated by GATA-1, SP1, and SP3, and is active in transient reporter assays in erythroid and mast cells.58,136,137,139 Promoter 1b is regulated by PU-1, SP1, and SP3, and is active in mast and primitive myeloid cells.138 139 Thus, in committed hematopoietic lineages, SCL appears to be regulated at least in part by a transcription factor with a pivotal role in erythroid cells (GATA-1) and by a critical myeloid transcription factor (PU-1).

However, although these promoters recapitulate aspects of the lineage-restricted pattern of SCL expression in transient reporter assays, they are not sufficient to direct reporter gene expression once integrated in chromatin.138 This observation stimulated a search for distant regulatory elements (“enhancers”) in the vicinity of the SCL gene. Therefore, a systematic survey of chromatin structure surrounding the SCL locus was undertaken140,141and resulted in identification of a number of enhancers active in both transient and stable transfection assays.141 More recently, transgenic assays have been used to study the activities of these elements and have identified spatially distinct enhancers that mimic components of normal SCL expression in endothelium, midbrain, hindbrain/spinal cord, and hematopoietic progenitors (A. Sinclair, M.-J. Sanchez, and A.R. Green, unpublished data, 1998). One element is particularly exciting because it appears to target hematopoietic progenitors throughout ontogeny.

SCL is clearly pivotal for normal hematopoiesis and vasculogenesis and is also likely to have a critical role in the nervous system. A plethora of questions remain unanswered. How does SCL induce an undifferentiated mesodermal cell to become a hemangioblast and then a multipotent hematopoietic stem cell? Does SCL perform the same role in different classes of hematopoietic stem cells throughout ontogeny? What is the role of SCL in the nervous system? Why is ectopic SCL expression leukemogenic in T-cell progenitors?

Answers to these and other questions are likely to require advances in at least three areas. First, we need to understand the significance of the multiprotein complexes that include SCL. How do the complexes differ in distinct cell types and what are the functional consequences of those differences? Second, it seems likely that different complexes will activate distinct target genes, and so it will also be essential to identify direct target genes downstream of SCL in different SCL-expressing cell types. Only in this way will we be able to put molecular flesh onto our skeletal ideas of SCL’s various functions. Third, how is SCL itself regulated? What triggers SCL expression in early mesoderm and therefore results in hemangioblast specification? How is SCL downregulated during differentiation along specific hematopoietic lineages, and is the modulation cause or consequence? Is there an SCL enhancer that can be used to target hematopoietic stem cells? At the current rate of progress, we should not have long to wait for some answers. Hopefully this will lead back to the bedside with new approaches to the manipulation of hematopoiesis and the treatment of T-ALL.

Supported by The Wellcome Trust, Leukaemia Research Fund (UK), Kay Kendall Leukaemia Fund (UK), Medical Research Council (UK), National Health and Medical Research Council (Canberra), Anti-Cancer Council of Victoria, and the Bone Marrow Donor Institute, Melbourne.

1
Hershfield
MS
Kurtzberg
J
Harden
E
Moore
JO
Whang-Peng
J
Haynes
BF
Conversion of a stem cell leukemia from a T-lymphoid to a myeloid phenotype induced by the adenosine deaminase inhibitor 2′-deoxycoformycin.
Proc Natl Acad Sci USA
81
1984
253
2
Kurtzberg
J
Bigner
SH
Hershfield
MS
Establishment of the DU.528 human lymphohemopoietic stem cell line.
J Exp Med
162
1985
1561
3
Kurtzberg
J
Waldmann
TA
Davey
MP
Bigner
SH
Moore
JO
Hershfield
MS
Haynes
BF
CD7+, CD4−, CD8− acute leukemia: A syndrome of malignant pluripotent lymphohematopoietic cells.
Blood
73
1989
381
4
Hata
A
Brenner
MB
Krangel
MS
Identification of putative human T cell receptor delta complementary DNA clones.
Science
238
1987
678
5
Loh
EY
Lanier
LL
Turck
CW
Littman
DR
Davis
MM
Chien
YH
Weiss
A
Identification and sequence of a fourth human T cell antigen receptor chain.
Nature
330
1987
569
6
Chien
YH
Iwashima
M
Kaplan
KB
Elliott
JF
Davis
MM
A new T-cell receptor gene located within the alpha locus and expressed early in T-cell differentiation.
Nature
327
1987
677
7
Boehm
T
Baer
R
Lavenir
I
Forster
A
Waters
JJ
Nacheva
E
Rabbitts
TH
The mechanism of chromosomal translocation t(11;14) involving the T-cell receptor (C. delta locus on human chromosome 14q11) and a transcribed region of chromosome 11p15.
EMBO J
7
1988
385
8
Croce
CM
Isobe
M
Palumbo
A
Puck
J
Ming
J
Tweardy
D
Erikson
J
Davis
M
Rovera
G
Gene for alpha-chain of human T-cell receptor: location on chromosome 14 region involved in T-cell neoplasms.
Science
227
1985
1044
9
Begley
CG
Aplan
PD
Davey
MP
Nakahara
K
Tchorz
K
Kurtzberg
J
Hershfeld
MS
Haynes
BF
Cohen
DI
Waldmann
TA
Kirsch
IR
Chromosomal translocation in a human leukemic stem cell line disrupts the T-cell antigen receptor δ-chain diversity region and results in a previously unreported fusion transcript.
Proc Natl Acad Sci USA
86
1989
2031
10
Begley
CG
Aplan
PD
Denning
SM
Haynes
B
Waldmann
TA
Kirsch
IR
The gene, SCL, is expressed during early hematopoiesis and encodes a differentiation-related DNA-binding motif.
Proc Natl Acad Sci USA
86
1989
10128
11
Finger
LR
Kagan
J
Christopher
G
Kurtzberg
J
Hershfield
MS
Nowell
PC
Croce
C
Involvement of the TCL5 gene on chromosome 1 in T-cell leukemia and melanoma.
Proc Natl Acad Sci USA
86
1989
5039
12
Chen
Q
Cheng
JT
Tsai
LH
Schneider
N
Buchanan
G
Carroll
A
Crist
W
Ozanne
B
Siciliano
MJ
Baer
R
The tal gene undergoes chromosome translocation in T cell leukemia and potentially encodes a helix-loop-helix protein.
EMBO J
9
1990
415
13
Bernard
O
Guglielmi
P
Jonveaux
P
Cherif
D
Gisselbrecht
S
Mauchauffe
M
Berger
R
Larsen
CJ
Mathieu-Mahul
D
Two distinct mechanisms for the SCL gene activation in the t(1;14) translocation of T-cell leukemias.
Genes Chromosomes Cancer
1
1990
194
14
Begley
CG
Aplan
PD
Davey
MP
de Villartay
JP
Cohen
DI
Waldmann
TA
Kirsch
IR
Demonstration of delta rec-pseudo J alpha rearrangement with deletion of the delta locus in a human stem-cell leukemia.
J Exp Med
170
1989
339
15
de Villartay
JP
Lewis
D
Hockett
R
Waldmann
TA
Korsmeyer
SJ
Cohen
DI
Deletional rearrangement in the human T-cell receptor alpha-chain locus.
Proc Natl Acad Sci USA
84
1987
8608
16
de Villartay
JP
Hockett
RD
Coran
D
Korsmeyer
SJ
Cohen
DI
Deletion of the human T-cell receptor delta-gene by a site-specific recombination.
Nature
335
1988
170
17
Aplan
PD
Begley
CG
Bertness
V
Nussmeier
MA
Ezquerra
A
Coligan
J
Kirsch
IR
The SCL gene is formed from a transcriptionally complex locus.
Mol Cell Biol
10
1990
6426
18
Begley
CG
Visvader
J
Green
AR
Aplan
PD
Metcalf
D
Kirsch
IR
Gough
NM
Molecular cloning and chromosomal localisation of the murine homolog of the human “helix-loop-helix” gene SCL.
Proc Natl Acad Sci USA
88
1991
869
19
Begley
CG
Robb
L
Rockman
S
Visvader
J
Bockamp
E-O
Chan
YS
Green
AR
Structure of the gene encoding the murine SCL protein.
Gene
138
1994
93
20
Breit
TM
Wolvers-Tettero
IL
van Dongen
JJ
Lineage specific demethylation of tal-1 gene breakpoint region determines the frequency of tal-1 deletions in alpha beta lineage T-cells.
Oncogene
9
1994
1847
21
Visvader
J
Begley
CG
Adams
JM
Differential expression of the LYL, SCL and E2A helix-loop-helix genes within the hemopoietic system.
Oncogene
6
1991
195
22
Green
AR
Salvaris
E
Begley
CG
Erythroid expression of the “helix-loop-helix” gene, SCL.
Oncogene
6
1991
475
23
Green
AR
Visvader
J
Lints
T
Harvey
R
Begley
CG
SCL is co-expressed with GATA-1 in haemopoietic cells but is also expressed in developing brain.
Oncogene
7
1992
653
24
Mouthon
M-A
Bernard
O
Mitjavila
M-T
Romeo
P-H
Vainchenker
W
Mathieu-Mahul
D
Expression of Tal-1 and GATA-binding proteins during human hematopoiesis.
Blood
81
1993
647
25
Mathieu-Mahul
D
Bernheim
A
Sigaux
F
Daniel
MT
Larsen
CJ
Berger
R
Translocation t(1;14) and rearrangement of the gene for the alpha chain of the T-cell receptor in acute T-lymphoblastic leukemia.
C R Acad Sci III
302
1986
525
26
Lampert
F
Harbott
J
Ritterbach
J
Ludwig
WD
Fonatsch
C
Schwamborn
D
Stier
B
Gnekow
A
Gerein
V
Stollmann
B
T-cell acute childhood lymphoblastic leukemia with chromosome 14q11 anomaly: A morphologic, immunologic, and cytogenetic analysis of 10 patients.
Blut
56
1988
117
27
Carroll
AJ
Crist
WM
Link
MP
Amylon
MD
Pullen
DJ
Ragab
AH
Buchanan
GR
Wimmer
RS
Vietti
TJ
The t(1;14)(p34;q11) is nonrandom and restricted to T-cell acute lymphoblastic leukemia: A Pediatric Oncology Group study.
Blood
76
1990
1220
28
Bernard
O
Lecointe
N
Jonveaux
P
Souyri
M
Mauchauffe
M
Berger
R
Larsen
CJ
Mathieu-Mahul
D
Two site-specific deletions and t(1;14) translocation restricted to human T-cell acute leukemias disrupt the 5′ part of the tal-1 gene.
Oncogene
6
1991
1477
29
Bernard
O
Barin
C
Charrin
C
Mathieu-Mahul
D
Berger
R
Characterization of translocation t(1;14)(p32;q11) in a T and in a B acute leukemia.
Leukemia
7
1993
1509
30
Chen
Q
Yang
CY
Tsan
JT
Ragab
AH
Peiper
SC
Carroll
A
Baer
R
Coding sequences of the tal-1 gene are disrupted by chromosome translocation in human T cell leukemia.
J Exp Med
172
1990
1403
31
Xia
Y
Brown
L
Tsan
JT
Yang
CY
Siciliano
MJ
Crist
WM
Carroll
AJ
Baer
R
The translocation (1;14)(p34;q11) in human T-cell leukemia: Chromosome breakage 25 kilobase pairs downstream of the TAL1 proto oncogene.
Genes Chromosomes Cancer
4
1992
211
32
Aplan
PD
Raimondi
SC
Kirsch
IR
Disruption of the SCL gene by a t(1;3) translocation in a patient with T cell acute lymphoblastic leukemia.
J Exp Med
176
1992
1303
33
Fitzgerald
TJ
Geoffrey
AM
Neale
SC
Raimondi
SC
Goorha
RM
c-tal, a helix-loop-helix protein, is juxtaposed to the T-cell receptor-β chain gene by a receiprocal chromosomal translocation: t(1;7) (p32;q35).
Blood
10
1991
2686
34
Francois
S
Delabesse
E
Baranger
L
Dautel
M
Foussard
C
Boasson
M
Blanchet
O
Bernard
O
Macintyre
EA
Ifrah
N
Deregulated expression of the TAL1 gene by t(1;5)(p32;31) in patient with T-cell acute lymphoblastic leukemia.
Genes Chromosomes Cancer
23
1998
36
35
Brown
L
Cheng
JT
Chen
Q
Siciliano
MJ
Crist
W
Buchanan
G
Baer
R
Site-specific recombination of the tal-1 gene is a common occurrence in human T cell leukemia.
EMBO J
9
1990
3343
36
Aplan
PD
Lombardi
DP
Ginsberg
AM
Cossman
J
Bertness
VL
Kirsch
IR
Disruption of the human SCL locus by “illegitimate” V-(D)-J recombinase activity.
Science
250
1990
1426
37
Collazo-Garcia
N
Scherer
P
Aplan
PD
Cloning and characterization of a murine SIL gene.
Genomics
30
1995
506
38
Izraeli
S
Colaizzo-Anas
T
Bertness
VL
Mani
K
Aplan
PD
Kirsch
IR
Expression of the SIL gene is correlated with growth induction and cellular proliferation.
Cell Growth Differ
8
1997
1171
39
Aplan
PD
Lombardi
DP
Reaman
GH
Sather
HN
Hammond
GD
Kirsch
IR
Involvement of the putative hematopoietic transcription factor SCL in T-cell acute lymphoblastic leukemia.
Blood
79
1992
1327
40
Borkhardt
A
Repp
R
Harbott
J
Keller
C
Berner
F
Ritterbach
J
Lampert
F
Frequency and DNA sequence of tal-1 rearrangement in children with T-cell acute lymphoblastic leukemia.
Ann Hematol
64
1992
305
41
Breit
TM
Mol
EJ
Wolvers-Tettero
IL
Ludwig
WD
van Wering
ER
van Dongen
JJ
Site-specific deletions involving the tal-1 and sil genes are restricted to cells of the T cell receptor alpha-beta lineage: T cell receptor delta gene deletion mechanism affects multiple genes.
J Exp Med
177
1993
965
42
Janssen
JW
Ludwig
WD
Sterry
W
Bartram
CR
SIL-TAL1 deletion in T-cell acute lymphoblastic leukemia.
Leukemia
7
1993
1204
43
Kikuchi
A
Hayashi
Y
Kobayashi
S
Hanada
R
Moriwaki
K
Yamamoto
K
Fujimoto
J
Kaneko
Y
Yamamori
S
Clinical significance of TAL1 gene alteration in childhood T-cell acute lymphoblastic leukemia and lymphoma.
Leukemia
7
1993
933
44
Breit
TM
Beishuizen
A
Ludwig
WD
Mol
EJ
Adriaansen
HJ
van Wering
ER
van Dongen
JJ
tal-1 deletions in T-cell acute lymphoblastic leukemia as PCR target for detection of minimal residual disease.
Leukemia
7
1993
2004
45
Bash
RO
Hall
S
Timmons
CF
Crist
WM
Amylon
M
Smith
RG
Baer
R
Does activation of the TAL1 gene occur in a majority of patients with T cell acute lymphoblastic leukemia? A pediatric oncology group study.
Blood
86
1995
666
46
Delabesse
E
Bernard
M
Meyer
V
Smit
L
Pulford
K
Cayuela
JM
Ritz
J
Bourqueiot
P
Strominger
JL
Valensi
F
Macintyre
EA
TAL1 expression does not occur in the majority of T-ALL blasts.
Br J Haematol
102
1998
449
47
Macintyre
EA
Smit
L
Ritz
J
Kirsch
IR
Strominger
JL
Disruption of the SCL locus in T-lymphoid malignancies correlates with commitment to the T-cell receptor alpha beta lineage.
Blood
80
1992
1511
48
Rockman
S
Begley
CG
Kannourakis
G
Mann
GJ
Dobrovic
AN
Kefford
RF
McGrath
K
SCL gene in human tumors.
Leukemia
6
1992
623
49
Shimamoto
T
Ohyashiki
K
Ohyashiki
JH
Kawakubo
K
Fujimura
T
Iwama
H
Nakazawa
S
Toyama
K
The expression pattern of erythrocyte/megakaryocyte-related transcription factors GATA-1 and the stem cell leukemia gene correlates with hematopoietic differentiation and is associated with outcome of acute myeloid leukemia.
Blood
86
1995
3173
50
Cross
MA
Heyworth
CM
Murrell
AM
Bockamp
E-O
Cobley
UT
Dexter
TM
Green
AR
Expression of lineage restricted transcription factors precedes lineage specific differentiation in a multipotent haemopoietic progenitor cell line.
Oncogene
9
1994
3013
51
Green
AR
De Luca
E
Begley
CG
Antisense SCL suppresses self-renewal and enhances spontaneous erythroid differentiation of the human leukaemic cell line K562.
EMBO J
10
1991
4153
52
Elwood
NJ
Cook
WD
Metcalf
D
Begley
CG
SCL, the gene implicated in human T-cell leukaemia, is oncogenic in a murine T-lymphocyte cell line.
Oncogene
8
1993
3093
53
Tanigawa
T
Elwood
N
Metcalf
D
Cary
D
DeLuca
E
Nicola
NA
The SCL gene product is regulated by and differentially regulates cytokine responses during myeloid leukemic cell differentiation.
Proc Natl Acad Sci USA
90
1993
7864
54
Tanigawa
T
Nicola
N
McArthur
GA
Strasser
A
Begley
CG
Differential regulation of macrophage differentiation in response to leukemia inhibitory factor/oncostatin-M/interleukin-6: The effect of enforced expression of the SCL transcription factor.
Blood
85
1995
379
55
Condorelli
GL
Tocci
A
Botta
R
Facchiano
F
Testa
U
Vitelli
L
Valtieri
M
Croce
CM
Peschle
C
Ectopic TAL-1/SCL expression in phenotypically normal or leukemic myeloid precursors: Proliferative and anti-apoptotic effects coupled with a differentiation blockade.
Mol Cell Biol
17
1997
2954
56
Tanigawa
T
Robb
L
Green
AR
Begley
CG
Constitutive expression of the putative transcription factor SCL associated with proviral insertion in the myeloid leukemic cell line WEHI-3BD-.
Cell Growth Differ
5
1994
557
57
Li
J
Finch
RA
Xiao
W
Sartorelli
AC
Identification of a repressor of the differentiation of WEHI-3B D-leukemia cells.
Exp Cell Res
242
1998
274
58
Aplan
PD
Nakahra
K
Orkin
SH
Kirsch
IR
The SCL gene product: A positive regulator of erythroid differentiation.
EMBO J
11
1992
4073
59
Elwood
NJ
Zogos
H
Pereira
DS
Dick
JE
Begley
CG
Enhanced megakaryocyte and erythroid development from normal human CD34+ cells: Consequence of enforced expression of SCL.
Blood
91
1998
3756
60
Valtieri
M
Tocci
A
Gabbianelli
M
Luchetti
L
Masella
B
Vitelli
L
Botta
R
Testa
U
Condorelli
GL
Peschle
C
Enforced TAL-1 expression stimulates primitive, erythroid and megakaryocytic progenitors but blocks the granulopoietic differentiation program.
Cancer Res
58
1998
562
61
Leroy-Viard
K
Vinit
M-A
Lecointe
N
Jouault
H
Hidner
U
Romeo
P-H
Mathieu-Mahul
D
Loss of TAL-1 protein activity induces premature apoptosis of Jurkat leukemic T cells upon medium depletion.
EMBO J
14
1995
2341
62
Condorelli
GL
Facchiano
F
Valtieri
M
Proietti
E
Vitelli
L
Lulli
V
Huebner
K
Peschle
C
Croce
CM
T-cell-directed TAL-1 expression induces T-cell malignancies in transgenic mice.
Cancer Res
56
1996
5113
63
Robb
L
Rasko
JEJ
Bath
ML
Strasser
A
Begley
CG
SCL, a gene frequently activated in human T cell leukaemia does not induce lymphomas in transgenic mice.
Oncogene
10
1995
205
64
Larson
RC
Lavenir
I
Larson
TA
Baer
R
Warren
AJ
Wadman
I
Nottage
K
Rabbitts
TH
Protein dimerization between Lmo2 (Rbtn2) and Tal1 alters thymocyte development and potentiates T cell tumorigenesis in transgenic mice.
EMBO J
15
1996
1021
65
Aplan
PD
Jones
CA
Chervinsky
DS
Zhao
XF
Ellsworth
M
Wu
CZ
McGuire
EA
Gross
KW
An scl gene product lacking the transactivation domain induces bony abnormalities and cooperates with LMO1 to generate T-cell malignancies in transgenic mice.
EMBO J
16
1997
2408
66
Kelliher
MA
Seldin
DC
Leder
P
Tal-1 induces T cell acute lymphoblastic leukemia accelerated by casein kinase IIalpha.
EMBO J
15
1996
5160
67
Curtis
DJ
Robb
L
Strasser
A
Begley
CG
The CD2-scl transgene alters the phenotype and frequency of T-lymphomas in N-ras transgenic or p53 deficient mice.
Oncogene
15
1997
2975
68
Lowsky
R
DeCoteau
JF
Reitmair
AH
Ichinohasama
R
Dong
WF
Xu
Y
Mak
TW
Kadin
ME
Minden
MD
Defects of the mismatch repair gene MSH2 are implicated in the development of murine and human lymphoblastic lymphomas and are associated with the aberrant expression of rhombotin-2 (Lmo-2) and Tal-1 (SCL).
Blood
89
1997
2276
69
Hsu
H-L
Wadman
I
Baer
R
Formation of in vivo complexes between the TAL1 and E2A polypeptides of leukemic T cells.
Proc Natl Acad Sci USA
91
1994
3181
70
Voronova
AF
Lee
F
The E2A and tal-1 helix-loop-helix proteins associate in vivo and are modulated by Id proteins during interleukin 6-induced myeloid differentiation.
Proc Natl Acad Sci USA
91
1994
5952
71
Condorelli
G
Vitelli
L
Valtieri
M
Marta
I
Montesoro
E
Lulli
V
Baer
R
Peschle
C
Coordinate expression and developmental role of Id2 protein and TAL1/E2A heterodimer in erythroid progenitor differentiation.
Blood
86
1995
164
72
Nielsen
AL
Norby
PL
Pedersen
FS
Jorgensen
P
E-box sequence and context-dependent TAL1/SCL modulation of basic helix-loop-helix protein-mediated transcriptional activation.
J Biol Chem
271
1996
31463
73
Valge-Archer
VE
Osada
H
Warren
AJ
Forster
A
Li
J
Baer
R
Rabbitts
TH
The LIM proteins RBTN2 and the basic helix-loop-helix protein TAL-1 are present in a complex in erythroid cells.
Proc Natl Acad Sci USA
91
1994
8617
74
Wadman
IA
Osada
H
Grutz
GG
Agulnick
AD
Westphal
H
Forster
A
Rabbitts
TH
The LIM-only protein Lmo2 is a bridging molecule assembling an erythroid, DNA-binding complex which includes the TAL1, E47, GATA-1 and Ldb1/NL1 proteins.
EMBO J
16
1997
3145
75
Cohen-Kaminsky
S
Maouche-Chretien
L
Vitelli
L
Vinit
MA
Blanchard
I
Yamamoto
M
Peschle
C
Romeo
PH
Chromatin immunoselection defines a TAL-1 target gene.
EMBO J
17
1998
5151
76
Grutz
GG
Bucher
K
Lavenir
I
Larson
T
Larson
R
Rabbitts
TH
The oncogenic T cell LIM-protein Imo2 forms part of a DNA-binding complex specifically in immature T cells.
EMBO J
17
1998
4594
77
Goldfarb
AN
Lewandowska
K
Inhibition of cellular differentiation by the SCL/tal oncoprotein: Transcriptional repression by an Id-like mechanism.
Blood
85
1995
465
78
Park
ST
Sun
XH
The Tal1 oncoprotein inhibits E47-mediated transcription. Mechanism of inhibition.
J Biol Chem
273
1998
7030
79
Hofmann
TJ
Cole
MD
The TAL1/Scl basic helix-loop-helix protein blocks myogenic differentiation and E-box dependent transactivation.
Oncogene
13
1996
617
80
Bain
G
Engel
I
Robanus Maandag
EC
te Riele
HP
Voland
JR
Sharp
LL
Chun
J
Huey
B
Pinkel
D
Murre
C
E2A deficiency leads to abnormalities in alphabeta T-cell development and to rapid development of T-cell lymphomas.
Mol Cell Biol
17
1997
4782
81
Johnson
SE
Wang
X
Hardy
S
Taparowsky
EJ
Konieczny
SF
Casein kinase II increases the transcriptional activities of MRF4 and MyoD independently of their direct phosphorylation.
Mol Cell Biol
16
1996
1604
82
Robb
L
Begley
CG
The SCL/TAL1 gene: roles in normal and malignant haematopoiesis.
Bioessays
19
1997
607
83
Murre
C
McCaw
PS
Baltimore
D
A new DNA binding and dimerization motif in immunoglobulin enhancer binding, daughterless, MyoD and myc proteins.
Cell
56
1989
777
84
Davis
RL
Weintraub
H
Acquisition of myogenic specificity by replacement of three amino acid residues from MyoD into E12.
Science
256
1992
1027
85
Hsu
HL
Huang
L
Tsan
JT
Funk
W
Wright
WE
Hu
JS
Kingston
RE
Baer
R
Preferred sequences for DNA recognition by the TAL1 helix-loop-helix proteins.
Mol Cell Biol
14
1994
1256
86
Drake
CJ
Brandt
SJ
Trusk
TC
Little
CD
TAL1/SCL is expressed in endothelial progenitor cells/angioblasts and defines a dorsal-to-ventral gradient of vasculogenesis.
Dev Biol
192
1997
17
87
Kallianpur
AR
Jordan
JE
Brandt
SJ
The SCL/TAL-1 gene is expressed in progenitors of both the hematopoietic and vascular systems during embryogenesis.
Blood
83
1994
1200
88
Chiba
T
Nagata
Y
Kishi
A
K.
S
Miyajima
A
Yamamoto
M
Engel
JD
Todokoro
K
Induction of erythroid-specific gene expression in lymphoid cells.
Proc Natl Acad Sci USA
90
1993
11593
89
Prasad
KS
Jordan
JE
Koury
MJ
Bondurant
MC
Brandt
SJ
Erythropoietin stimulates transcription of the TAL1/SCL gene and phosphorylation of its protein products.
J Biol Chem
270
1995
11603
90
Hoang
T
Paradis
E
Brady
G
Billia
F
Nakahara
K
Iscove
NN
Kirsch
IR
Opposing effects of the basic helix-loop-helix transcription factor SCL on erythroid and monocytic differentiation.
Blood
87
1996
102
91
Murrell
AM
Bockamp
E-O
Göttgens
B
Chan
YS
Goss
MA
Heyworth
CM
Green
AR
Discordant regulation of SCL/TAL-1 mRNA and protein during erythroid differentiation.
Oncogene
11
1995
131
92
Green
AR
Rockman
S
De Luca
E
Begley
CG
Induced myeloid differentiation of K562 cells with down-regulation of erythroid and megakaryocytic transcription factors; a novel experimental model for haemopoietic lineage restriction.
Exp Haematol
21
1993
525
93
Robb
L
Lyons
I
Li
R
Hartley
L
Kontgen
F
Harvey
RP
Metcalf
D
Begley
CG
Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene.
Proc Natl Acad Sci USA
92
1995
7075
94
Shivdasani
RA
Mayer
EL
Orkin
SH
Absence of blood formation in mice lacking the T-cell leukemia oncoprotein tal-1/SCL.
Nature
373
1995
432
95
Robb
L
Elwood
NJ
Elefanty
AG
Köntgen
F
Li
R
Barnett
LD
Begley
CG
The SCL gene product is required for the generation of all hematopoietic lineages in the adult mouse.
EMBO J
15
1996
4123
96
Porcher
C
Swat
W
Rockwell
K
Fujiwara
Y
Alt
FW
Orkin
SH
The T cell leukemia oncoprotein SCL/tal-1 is essential for development of all hematopoietic lineages.
Cell
86
1996
47
97
Elefanty
AG
Robb
L
Birner
R
Begley
CG
Hematopoietic-specific genes are not induced during in vitro differentiation of scl-null embryonic stem cells.
Blood
90
1997
1435
98
Elefanty
AG
Begley
CG
Metcalf
D
Barnett
L
Köntgen
F
Robb
L
Characterisation of hematopoietic progenitor cells that express the transcription factor SCL, using a lacZ ‘knock-in’ strategy.
Proc Natl Acad Sci
95
1998
11897
99
Green
T
Hematopoiesis—Master regulator unmasked.
Nature
383
1996
575
100
Warren
AJ
Colledge
WH
Carlton
MB
Evans
MJ
Smith
AJ
Rabbitts
TH
The oncogenic cysteine-rich LIM domain protein Rbtn2 is essential for erythroid development.
Cell
78
1994
45
101
Yamada
Y
Warren
AJ
Dobson
C
Forster
A
Pannell
R
Rabbitts
TH
The T cell leukemia LIM protein Lmo2 is necessary for adult mouse hematopoiesis.
Proc Natl Acad Sci USA
95
1998
3890
102
Sabin
FR
Studies on the origin of blood vessels and of red blood corpuscles as seen in the living blastoderm of chicks during the second day of incubation.
Contrib Embryol
36
1920
213
103
Murray
PDF
The development in vitro of the blood of the early chick embryo.
Proc R Soc B
111
1932
497
104
Wagner
RC
Endothelial cell embryology and growth.
Adv Microcirc
9
1980
45
105
Godin
IE
Garcia-Porrero
JA
Coutinho
A
Dieterlen-Lievre
F
Marcos
MAR
Para-aortic splanchnopleura from early mouse embryos contains B1a cell progenitors.
Nature
364
1993
67
106
Godin
I
Dieterlen-Lievre
F
Cumano
A
Emergence of multipotent hemopoietic cells in the yolk sac and paraaortic splanchnopleura in mouse embryos, beginiing at 8.5 days postcoitus.
Proc Natl Acad Sci USA
92
1995
773
107
Medvinsky
AL
Samoylina
NL
Muller
AM
Dzierzak
EA
An early pre-liver intra-embryonic source of CFU-S in the developing mouse.
Nature
364
1993
64
108
Cumano
A
Dieterlen-Lievre
F
Godin
I
Lymphoid potential, probed before circulation in mouse, is restricted to caudal intraembryonic splanchnopleura.
Cell
86
1996
907
109
Medvinsky
A
Dzierzak
E
Definitive hematopoiesis is autonomously initiated by the AGM region.
Cell
86
1996
897
110
Tavian
M
Coulombel
L
Luton
D
San Clemente
H
Dieterlen-Lievre
F
Péault
B
Aorta-associated CD34+ hematopoietic cells in the early human embryo.
Blood
87
1996
67
111
Wood
HB
May
G
Healy
L
Enver
T
Morriss-Kay
GM
CD34 expression patterns during early mouse development are related to modes of blood vessel formation and reveal additional sites of hematopoiesis.
Blood
90
1997
2300
112
Pardanaud
L
Luton
D
Prigent
M
L-M
B
Catala
M
Dieterlen-Lievre
F
Two distinct endothelial lineages in ontogeny, one of them related to hemopoiesis.
Development
122
1996
1363
113
Fina
L
Molgaard
HV
Robertson
D
Bradley
NJ
Monaghan
P
Delia
D
Sutherland
DR
Baker
MA
Greaves
MF
Expression of the CD34 gene in vascular endothelial cells.
Blood
75
1990
2417
114
Shalaby
F
Rossant
J
Yamaguchi
TP
Gertsenstein
M
Wu
XF
Breitman
ML
Schuh
AC
Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice.
Nature
376
1995
62
115
Shalaby
F
Ho
J
Stanford
WL
Fischer
K-D
Schuh
AC
Schwartz
L
Bernstein
A
Rossant
J
A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis.
Cell
89
1997
981
116
Kennedy
M
Firpo
M
Choi
K
Wall
C
Robertson
S
Kabrun
N
Keller
G
A common precursor for primitive erythropoiesis and definitive haematopoiesis.
Nature
386
1997
488
117
Eichmann
A
Corbel
C
Nataf
V
Vaigot
P
Bréant
C
Le Douarin
NM
Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic mesodermal cells expressing vascular endothelial growth factor receptor 2.
Proc Natl Acad Sci USA
94
1997
5141
118
Liao
W
Bisgrove
BW
Sawyer
H
Hug
B
Bell
B
Peters
K
Grunwald
DJ
Stainier
DYR
The zebrafish gene cloche acts upstream of a flk-1 homologue to regulate endothelial cell differentiation.
Development
124
1997
381
119
Hwang
L-Y
Siegelman
M
Davis
L
Oppenheimer-Marks
N
Baer
R
Expression of the TAL-1 proto-oncogene in cultured endothelial cells and blood vessels of the spleen.
Oncogene
8
1993
3043
120
Visvader
JE
Fujiwara
Y
Orkin
SH
Unsuspected role for the T-cell leukemia protein SCL/tal-1 in vascular development.
Genes Dev
12
1998
473
121
Liao
EC
Paw
BH
Oates
AC
Pratt
SJ
Postlethwait
JH
Zon
LI
SCL/Tal-1 transcription factor acts downstrem of cloche to specify hematopoietic and vascular progenitors in zebrafish.
Genes Dev
12
1998
621
122
Gering
M
Rodaway
ARF
Göttgens
B
Patient
RK
Green
AR
The SCL gene specifies haemangioblast development from early mesoderm.
EMBO J
17
1998
4029
123
Jan
YH
Jan
LY
HLH proteins, fly neurogenesis, and vertebrate myogenesis.
Cell
75
1993
827
124
Weintraub
H
The Myo D family and myogenesis: Redundancy, networks and thresholds.
Cell
75
1993
1241
125
Jen
Y
Weintraub
H
Benezra
R
Overexpression of Id protein inhibits the muscle differentiation program: In vivo association of Id with E2A proteins.
Genes Dev
6
1992
1466
126
Goldfarb
AN
Goueli
S
Mickelson
D
Greenberg
JM
T-cell acute lymphoblastic leukemia—The associated gene SCL/tal codes for a 42-kD nuclear phosphoprotein.
Blood
58
1992
537
127
Cheng
J-T
Hsu
H-L
Hwang
L-Y
Baer
R
Products of the TAL-1 oncogene: Basic helix-loop-helix proteins phosphorylated at serine residues.
Oncogene
8
1993
677
128
Elwood
NJ
Green
AR
Melder
A
Visvader
J
Begley
CG
Nicola
N
The SCL protein displays cell-specific heterogeneity in size.
Leukemia
8
1994
106
129
Mead
PE
Kelley
CM
Hahn
PS
Piedad
O
Zon
L
SCL specifies hematopoietic mesoderm in Xenopus embryos.
Development
125
1998
2611
130
Miller
BA
Floros
J
Cheung
JY
Wojchowski
DM
Bell
L
Begley
CG
Elwood
NJ
Kreider
J
Christian
C
Steel factor affects SCL expression during normal erythroid differentiation.
Blood
84
1994
2971
131
Zhang
MY
Clawson
GA
Olivieri
NF
Bell
LL
Begley
CG
Miller
BA
Expression of SCL is normal in transfusion-dependent Diamond-Blackfan anemia but other bHLH proteins are deficient.
Blood
90
1997
2068
132
Cheng
J-T
Cobb
MH
Baer
R
Phosphorylation of the TAL-1 oncoprotein by the extracellular-signal-regulated protein kinase ERK1.
Mol Cell Biol
13
1993
801
133
Bernard
O
Azogui
O
Lecointe
N
Mugneret
F
Berger
R
Larsen
CJ
Mathieu-Mahul
D
A third tal-1 promoter is specifically used in human T cell leukemias.
J Exp Med
176
1992
919
134
Krosl
G
He
G
Lefrancois
M
Charron
F
Romeo
PH
Jolicoeur
P
Kirsch
IR
Nemer
M
Hoang
T
Transcription factor SCL is required for c-kit expression and c-Kit function in hemopoietic cells.
J Exp Med
188
1998
439
135
Enver
T
Greaves
M
Loops, lineage, and leukemia.
Cell
94
1998
9
136
Lecointe
N
Bernard
O
Naert
K
Joulin
V
Larsen
CJ
Romeo
PH
Mathieu-Mahul
D
GATA- and SP1-binding sites are required for the full activity of the tissue-specific promoter of the tal-1 gene.
Oncogene
9
1994
2623
137
Bockamp
E-O
McLaughlin
F
Murrell
AM
Göttgens
B
Robb
L
Begley
CG
Green
AR
Lineage-restricted regulation of the murine SCL/TAL-1 promoter.
Blood
86
1995
1502
138
Bockamp
EO
McLaughlin
F
Göttgens
B
Murrell
AM
Elefanty
AG
Green
AR
Distinct mechanisms direct SCL/tal-1 expression in erythroid cells and CD34 positive primitive myeloid cells.
J Biol Chem
272
1997
8781
139
Bockamp
E-O
Fordham
JL
Göttgens
B
Murrell
AM
Green
AR
Transcriptional regulation of the stem cell leukaemia gene by PU.1 and ELF-1.
J Biol Chem
273
1998
29032
140
Leroy-Viard
K
Vinit
M-A
Lecointe
N
Mathieu-Mahul
D
Romeo
P-H
Distinct DNase-I hypersensitive sites are assocaited with TAL-1 transcription in erythroid and T-cell lines.
Blood
84
1994
3619
141
Göttgens
B
McLaughlin
F
Bockamp
EO
Fordham
JL
Begley
CG
Kosmopoulos
K
Elefanty
AG
Green
AR
Transcription of the SCL gene in erythroid and CD34 positive primitive myeloid cells is controlled by a complex network of lineage-restricted chromatin-dependent and chromatin-independent regulatory elements.
Oncogene
15
1997
2419

Author notes

Address reprint requests to C. Glenn Begley, MD, PhD, the Walter and Eliza Hall Institute of Medical Research and the Rotary Bone Marrow Research Laboratories, PO Royal Melbourne Hospital, Parkville, Victoria, Australia 3050.

Sign in via your Institution