Hematopoiesis is a remarkable cell-renewal process that leads to the continuous generation of large numbers of multiple mature cell types, starting from a relatively small stem cell compartment. A highly complex but efficient regulatory network is necessary to tightly control this production and to maintain the hematopoietic tissue in homeostasis. During the last 3 decades, constantly growing numbers of molecules involved in this regulation have been identified. They include soluble cytokines and growth factors, cell–cell interaction molecules, and extracellular matrix components, which provide a multifunctional scaffolding specific for each tissue. The cloning of numerous growth factors and their mass production have led to their possible use for both fundamental research and clinical application.

The regulation of hematopoiesis is a complex process that has received much attention (for reviews see Moore et al,1 1990; Metcalf,2 1993; Ogawa,3 1993). Research continues to identify the various components involved in this regulation (for review see Levesque et al,4 1991). Many of the growth factors can now be cloned for research and clinical purposes (for review see Simmons and Haylock,5 1995).

Type β transforming growth factors (TGF-βs) were discovered by De Larco and Todaro6 in 1978. Originally called “sarcoma growth factors,” they were first isolated from the supernatant fluids of Moloney MuSV-transformed mouse 3T3 fibroblasts and described as a family of growth-stimulating polypeptides. The further nomenclature “transforming growth factor” was adopted because of the ability of these molecules to confer on untransformed indicator fibroblasts functional properties associated with neoplastic transformation.6,7 

At present, TGF-βs are considered pleiotropic factors because they have been shown to play a regulatory role in most processes linked to the control of somatic tissue development and renewal. As pointed out by Sporn and Roberts,8 TGF-βs may be considered as “prototypic multifunctional signaling molecules.” Indeed, these factors can exert either a positive or a negative effect on proliferation, differentiation, or cell death, depending on the developmental stage of the target cell, its in vivo environment, or the medium used for in vitro studies. As will be described in this review, this is particularly true in the hematopoietic system, where TGF-βs play a pivotal role.

Latent and active forms

Three highly similar isoforms of TGF-β, called TGF-β1, -β2, and -β3, were identified and cloned from mammals between 1985 and 1988.9-11 Although the regulatory role of these 3 isoforms may differ, it has been established that all 3 are involved in the regulation of hematopoiesis. Two other isoforms, called TGF-β4 and -β5, have been cloned, respectively, in the chicken12 and in xenopus.13 More recently, an mRNA encoding a new member of the TGF-β family, called endometrial bleeding associated factor (ebaf), has been identified in mammals.14 

TGF-βs are synthesized as precursor proteins, which are biologically inactive. They consist of pre–pro-peptides, which require a 2-step process to give rise to active TGF-βs15 (for reviews see Lawrence,16 1991; Gleizes et al,17 1997). A first proteolytic cleavage leads to the elimination of a hydrophobic signal peptide, in the N-terminal region of the precursor protein, yielding pro–TGF-β. A second cleavage leads to the separation of the pro-region of the protein from the TGF-β mature peptide (Figure 1). In the case of TGF-β1, the entire precursor (pre–pro-peptide) is a 390–amino acid chain. The signal peptide corresponds to amino acids 1 to 29, the pro-region of the precursor to amino acids 30 to 278, and the mature peptide to amino acids 279 to 390.15 The bioactive forms of TGF-βs (25 kd) are composed of 2 mature peptide chains linked by disulfide bonds. TGF-βs are usually produced as homodimers (TGF-β1.1, -β2.2, -β3.3), but natural heterodimeric molecules have also been identified (TGF-β1.2 and -β2.3).18 

Fig. 1.

TGF-β: structure, latency, activation, and receptors.17,67 

LAP indicates latency-associated peptide; LTBP, latent TGF-β binding protein; M6P/IGFII-R, mannose-6-phosphate/type II insulin-like growth factor receptor; PLg, plasminogen; PL, plasmin; Smad, TGF-β signal transduction proteins; Anti-Smad, antagonistic Smad; Co-Smad, common-partner Smad; R-Smad, receptor-regulated Smad; TGase, transglutaminase; TβR-I, -II, -III, TGF-β receptor type I, II, III; TSP, thrombospondin; uPA, urokinase plasminogen activator; and uPA-R, uPA receptor.

Fig. 1.

TGF-β: structure, latency, activation, and receptors.17,67 

LAP indicates latency-associated peptide; LTBP, latent TGF-β binding protein; M6P/IGFII-R, mannose-6-phosphate/type II insulin-like growth factor receptor; PLg, plasminogen; PL, plasmin; Smad, TGF-β signal transduction proteins; Anti-Smad, antagonistic Smad; Co-Smad, common-partner Smad; R-Smad, receptor-regulated Smad; TGase, transglutaminase; TβR-I, -II, -III, TGF-β receptor type I, II, III; TSP, thrombospondin; uPA, urokinase plasminogen activator; and uPA-R, uPA receptor.

Close modal

Once synthesized and processed, TGF-βs are released by cells as latent complexes, which are biologically inactive. Two forms of latent complexes have been described, the “small” and “large” latent complexes, as shown in Figure 1. In the small latent complex, one molecule of mature, active TGF-β is noncovalently associated with one disulfide-bonded pro-peptide dimer, called latency-associated protein or LAP (74 kd in the case of TGF-β1). In the large latent complex, LAP is linked by disulfide bonds to one member of a family of high-molecular-weight proteins (125-160 kd), called latent TGF-β–binding proteins or LTBPs.19,20 The cDNAs of various related LTBPs have been cloned.21-23 In the erythroleukemic cell line HEL, the synthesis of LTBPs has been found to be coordinated with that of TGF-β small latent complex to form the large latent complex, which is then secreted by cells.24The LTBPs confer to this complex the ability to associate with the extracellular matrix, permitting the storage of TGF-β (for reviews see Munger et al,25 1997; Taipale and Keski-Oja,26 1997). Because LTBPs exist in several isoforms, the bioavailability of TGF-β and its specific targeting to different organs may be regulated in part by the formation of different types of large latent complexes. It has also been suggested that LTBPs participate in bone formation as structural matrix proteins.27 The release of latent TGF-β from the extracellular matrix is triggered by proteolytic enzymes such as chymase, elastase, and plasmin, which are able to cleave LTBPs.28-30 

Activation of latent TGF-βs

Extracellular activation of the TGF-β latent complexes is a critical process in the regulation of TGF-β functions in vivo. The interaction between TGF-β and LAP is not covalent and can be disrupted in vitro by heat treatment or acidification.31Although physicochemical variables such as local acidification32 or exposure to active oxygen species33 may participate in the regulation of TGF-β activation, mechanisms involving proteolytic cleavage or conformational modification of LAP are more likely to operate in vivo.

Different mechanisms of activation are presented in Figure 1. Plasmin has been shown to promote the activation of latent TGF-β by proteolytic nicking within the N-terminal region of the LAP.34,35 This disrupts noncovalent bonds and results in the release of active TGF-β.35 In monocytes, macrophages, and endothelial cells, cellular activation of latent TGF-β has been reported to involve the mannose-6-phosphate/type II insulinlike growth factor receptor (M6P/IGFII-R) and the urokinase plasminogen activator receptor (uPA-R).36-38 One proposed mechanism is that M6P/IGFII-R, which binds latent TGF-β, complexes with uPA-R. Plasmin would be generated locally from plasminogen through the action of uPA and would allow the production of active TGF-β. Another enzyme, transglutaminase, has been identified as an effector controlling both the deposition rate of LTBPs in the matrix39 and the cell-surface activation of latent TGF-β.37,40 Transglutaminase-mediated activation of latent TGF-β depends on interactions with specific residues of LTBP.41 Thrombospondin (TSP), a platelet α-granule and extracellular matrix protein, has also been shown to promote activation of latent forms of TGF-β. In contrast to what has been described for plasmin and transglutaminase, TSP-mediated activation of latent TGF-β occurs through a cell- and protease-independent mechanism, as demonstrated by in vitro studies. This effector induces a conformational change of LAP, which then results in the release of active TGF-β.42,43 The role of TSP in the activation of latent TGF-β in vivo has been demonstrated by the generation ofTSP-null mice.44 In this model, major histologic abnormalities have been observed and correlated with a lack of active TGF-β. These defects could be reversed by a treatment that activates TGF-β. Regulation of the glycosylation of LAP has also been proposed to participate in the control of TGF-β latency.45 Recently, activation of latent TGF-β via an interaction with the integrin α5 β6 has been reported, providing a novel possible mechanism regulating the function of TGF-β.46 

Two families of serine/threonine kinase receptors form heteromeric complexes

Among the several transmembrane or membrane-bound proteins known to interact with TGF-βs, the type I and type II TGF-β receptors (TβR-I or ALK, and TβR-II) are directly involved in signal transduction. TβR-I and TβR-II represent 2 families of transmembrane serine/threonine kinase receptors of 53 to 65 kd47-49 and 80 to 95 kd,50 respectively, that interact and form heterotetrameric complexes. The mechanism by which signaling by these 2 receptors occurs is now well established.51 TGF-β first binds to TβR-II, which is a constitutively active kinase. TβR-I is then recognized and recruited into the TGF-β/TβR-II complex and phosphorylated by TβR-II. Phosphorylation allows TβR-I to propagate the signal to downstream intracellular substrates.

Because TβR-I and TβR-II exist in multiple forms, it has been proposed that homodimeric and heterodimeric forms of TGF-β may induce a specific response by interacting with different heterotetrameric receptor complexes of specific signaling capacities.52 

Accessory receptors

In addition to TβR-I and TβR-II, accessory TGF-β receptors, not necessarily required for signal transduction, can be expressed at the surface of cells responsive to TGF-β (for review see Piek et al,52 1999). The type III TGF-β receptor (TβR-III or βglycan), a 300- to 400-kd membrane-anchored proteoglycan,53,54 and the 180-kd glycoprotein endoglin could function as regulators of ligand access to the signaling receptors. Although the precise roles of endoglin and βglycan are not fully understood, some of their properties suggest distinct functions for these 2 TGF-β receptors. First, βglycan is able to interact with TGF-β1, -β2, and -β3,55 whereas endoglin interacts with TGF-β1 and -β3 but not efficiently with TGF-β2.56 Second, the role of βglycan could be to present TGF-βs to TβR-II and facilitate their binding,57,58 whereas endoglin appears to diminish rather than enhance TGF-β responses in certain cell types.59Third, endoglin and βglycan possess a specific cell-distribution pattern, which may confer the ability of different cell types to respond differentially to TGF-β1, -β2, and -β3. For example, endoglin is coexpressed with TβR-I and TβR-II on vascular endothelial cells60,61 and on hematopoietic cells including macrophages,62 erythroid cell subsets,63 and B-cell precursors,64 whereas these cells express little or no βglycan. Marrow stromal cells may express both endoglin and βglycan,65 whereas none of these TGF-β receptors appear to be present on the cell surface of hematopoietic progenitors including early colony-forming units (CFU)–granulocyte/erythrocyte/monocyte/megakaryocyte (GEMM), CFU–granulocyte/monocyte (GM), and burst-forming units-erythrocyte (BFU-E).63 

Other cell-surface receptors have been identified for their ability to bind TGF-β and are classified as TβR-IV to TβR-VI (for review see Massagué,66 1992). However, the function of these other receptor families in TGF-β signaling remains to be clarified.

The Smad intracellular proteins

The intracellular TGF-β signaling pathway involves the Smad protein family as substrates for the signaling receptors (for review see Massagué,67 1998; Piek et al,521999). This network involves the cooperation among 3 subclasses of Smad proteins, which can be distinguished by distinct functions in TGF-β signal transduction. Briefly, a first group of Smads called “receptor-activated”67 or “receptor-regulated”52 Smads (R-Smads) are directly phosphorylated by activated TβR-I. Upon phosphorylation, R-Smads interact with members of a second subclass of Smads called “common-partner Smads” or Co-Smads, with which they form heterodimeric complexes. R-Smad/Co-Smad complexes are translocated to the nucleus, where they associate with DNA-binding partners and then regulate the transcriptional response of the target genes. A third subclass of Smads called “antagonistic”67 or “inhibitory”52 Smads (Anti-Smads) prevents the interaction between R-Smads and Co-Smads and participates in negative feedback to repress TGF-β responses. In the case of signal transduction by TGF-β in mammalian cells, R-Smads include Smad2 and Smad3,68,69 Co-Smads include Smad4,69,70 and Anti-Smads include Smad6 and Smad7.71-73 Smad5, otherwise described as an R-Smad and involved in signal transduction by bone morphogenetic proteins (BMPs),74 has been demonstrated to mediate the inhibitory effect of TGF-β on human hematopoietic stem/progenitor cells.75 The specificity of the cellular response depends on interactions between these different possible partners.

Studies of hematopoietic pathologies involving TGF-β have provided important evidence of its key role in the regulation of human hematopoietic stem/progenitor cell quiescence, proliferation, and differentiation. These human pathologies are often more informative than knockout mice as regards the role of this pleiotropic factor. Indeed, knockout mice often exhibit secondary disorders subsequent to the accumulation of early defects during embryonic development. This situation is also often observed in the case of genetic diseases, in which more than one function or organ can be affected during development by a single mutation. In contrast, the pathologic situation resulting from a single gene mutation occurring in the adult will produce a clonal defect, allowing the precise evaluation of the role of the mutated gene in a specific function. This is particularly true in the case of various cancers.

Inactivation of the TGF-β signaling cascade leads to malignant transformation of early human hematopoietic cells

The inactivation of one of the various genes involved in the TGF-β signal transduction pathway may represent a possible mechanism by which some early hematopoietic progenitors, which are normally quiescent, escape from cell-cycling inhibition. Abnormalities in the expression of TGF-β receptors have been described in proliferative syndromes including both early myeloid76,77 and lymphocytic leukemia.78,79 In these pathologies, a selective advantage is given to the tumor cells by the loss of TβR-I or TβR-II expression and by the fact that these cells continue to produce TGF-β1 to inhibit normal cell proliferation. Active TGF-β present in the bone marrow microenvironment and autocrine/paracrine TGF-β1 secreted by normal and leukemic hematopoietic cells are able to exert a negative control on the growth of normal progenitors, but not on leukemic cells, which have overcome TGF-β regulatory signals. A loss of sensitivity to the growth-inhibitory effect of TGF-β due to an inactivation of TβR-II has also been described in the case of human cutaneous T-cell lymphoma cells.80-82 

A mutational analysis of the gene coding for the TGF-β signal transducer Smad2 has been performed on 50 primary lymphoid and myeloid leukemia cells, but no genetic defects were found in this gene.83 However, a larger panel of hematologic disorders should be analyzed before excluding the possibility of various mutations in Smad genes in some of these pathologies. Blocking of TGF-β signaling by repression of Smad3 activity has been reported in chronic myeloid leukemia. In these cases, the dysregulation was not due to mutations in the Smad3 gene, but was correlated with an abnormal expression of Evi-1, a zinc-finger oncoprotein that interacts with Smad3 and suppresses its transcriptional activity.84 In hematopoietic cells, Evi-1 expression is normally restricted to a transient stage of myeloid differentiation.85 Its constitutive expression can result from chromosomal rearrangements and may contribute to leukemogenesis by specifically blocking the growth-inhibitory signaling of TGF-β.86 

Pathologic overproduction of TGF-β induces bone marrow fibrosis and decreases stem/progenitor frequency

In physiologic conditions, the amount of TGF-β produced by bone marrow stromal cells and hematopoietic cells should be adequate to maintain homeostasis of the stem/progenitor cell compartment. In pathologic situations, excessive production of TGF-β by stromal cells has been correlated with a failure of early hematopoietic progenitors in the marrow. This situation has been described in the case of human chronic idiopathic neutropenia, in which a drastic reduction in CD34+ progenitor cell frequency is observed.87The same phenomenon has been described for B-cell lymphocytic leukemia. In this case, the pathogenesis is due to the proliferation of leukemic cells, but also to an increased inhibition of normal progenitor cell growth, in response to excessive amounts of TGF-β1 secreted by bone marrow stromal cells.88 

An abnormally elevated production of TGF-β has also been shown to contribute to the pathogenesis of leukemia by promoting the progression of bone marrow fibrosis (for review see Le Bousse-Kerdiles and Martyre,89 1999). In these pathologies, TGF-β is often secreted in excess by leukemic cells, monocytes, and megakaryocytes, which results in stimulation of collagen synthesis in bone marrow fibroblasts and deposition in the marrow.90-93 This observation is in agreement with the fact that TGF-β is able to activate the promoter of the human type VII collagen gene through the action of Smad3 and Smad4.94 

Vascular pathologies involving TGF-β

Defects in genes coding for a TGF-β receptor have also been correlated with the genetic diseases called hereditary hemorrhagic telangiectasia (HHT). These syndromes concern mainly endothelial cells and are characterized by arteriovenous malformations and recurrent hemorrhage. HHT syndromes were first found to be caused by mutations in the endoglin gene95 but have also been correlated with mutations in the TβR-Igene,96 suggesting that endoglin and TβR-I act through a common pathway to control blood vessel development and repair.

TGF-β has also been reported to be involved in another vascular disease, atherosclerosis, which has a multifactorial pathology implicating many other interacting phenomena. Atherosclerosis has been associated with the presence of lipoprotein Lp(a), a glycoprotein that has a structure similar to that of plasminogen. Lp(a) binds to the membranes of endothelial cells and monocytes and thereby inhibits plasminogen binding and the subsequent generation of plasmin by these cells. Because plasmin is a potent activator of latent TGF-β in vivo, this results in an insufficient rate of activation of TGF-β and, as a consequence, in the migration and proliferation of smooth muscle cells in the arterial intima (for review see Angles-Cano,971997).

TGF-β and the control of hematopoietic stem/progenitor cell proliferation: a model for other somatic cells?

We have reviewed above various studies showing that the inactivation of the TGF-β signaling cascade participates in malignant transformation of early hematopoietic cells, which then escape from negative cell-cycle controls. It is interesting to note that the same phenomenon has been described in cancers affecting various types of nonhematopoietic somatic cells, which suggests that TGF-β may act as a cell-cycle inhibitor in several nonhematopoietic somatic tissues in vivo. Indeed, although they probably do not represent the entire cause of the pathology, mutations or genetic defects resulting in a lack of TβR-I or TβR-II function are associated with the acquisition of a transformed phenotype in several types of murine and human cancers, including colon cancers,98,99 gastric cancers,100,101 prostate cancers,102pancreatic cancers,103 thyroid tumors,104hepatic tumors,105,106 retinoblastoma,107,108and lung adenocarcinoma.109 Moreover, the importance of TGF-β signaling for the control of normal somatic cell proliferation has been demonstrated in skin keratinocytes,110,111 cells of the mammary gland, lung,112 and exocrine pancreas113 with the use of transgenic mice expressing a dominant-negative mutant TβR-II (for review see Letterio and Bottinger,114 1998).

In addition, elements of the TGF-β signal transduction pathway downstream to the TGF-β receptors have been identified as potential targets for oncogenic transformation. Indeed, mutations or somatic alterations resulting in a disruption of the Smad signaling cascade have been observed in several tumor cells resistant to the growth-inhibitory effect of TGF-β1 (for review see Hata et al,115 1998). Briefly, in these cells, insensitivity to TGF-β1 was reported to be due either to an inactivation of the TGF-β signal transducers Smad2 and Smad4116-118 or to an enhanced expression of the TGF-β1 signaling inhibitor Smad6.119 These observations should promote a search forSmad gene mutations in the hematopoietic system, especially in leukemia.

The role of TGF-β in the regulation of hematopoiesis has also been analyzed in vivo using different murine models (for review see Bottinger et al,120 1997). However, the involvement of this pleiotropic factor in the regulation of various nonhematopoietic tissue functions renders these studies sometimes difficult to interpret because of possible indirect regulatory effects.

TGF-β protects stem/progenitor cells from agents that selectively kill cycling cells

Hematopoietic stem/progenitor cells are able to reconstitute the pool of mature blood cells in the case of severe hematopoietic failure. This implies that these cells can rapidly pass from a quiescent or slow cycling state to active proliferation. This phenomenon can be observed in mice treated with chemotherapeutic drugs, such as 5-fluorouracil (5-FU), that selectively kill the cycling cells. Following the administration of 5-FU, a dramatic decrease in hematopoietic progenitor cell frequency in the bone marrow is observed, as only the most primitive and quiescent cells remain unaffected by this treatment. A few days later, the quiescent stem/progenitor cells enter a hyperproliferative state, promoting hematopoietic reconstitution. This experimental system has been used to evaluate the ability of TGF-β to affect hematopoietic stem/progenitor cell cycling. TGF-β1 has been shown to delay hematologic recovery after a sublethal injection of 5-FU.121 Moreover, both the TGF-β1 and -β2 isoforms were able to protect hematopoietic stem/progenitor cells from a treatment by a high dose of 5-FU,122 which demonstrates the ability of these molecules to exert a negative control on the cell cycle of primitive murine hematopoietic cells in vivo. It is important to note that this effect was reversible,121,122 which suggests that TGF-β is not an inducer of cell death for primitive stem/progenitor cells in vivo. This point will be discussed more extensively later.

In vivo administration of TGF-β in mice modulates hematopoietic development in a lineage-specific manner

In vivo administration of TGF-β in mice has also been performed to study its specific effects on early and late progenitors and on the different hematopoietic lineages. One approach, which was to test in vitro the clonogenic capacity of hematopoietic progenitors after the local administration of TGF-β1 into the femur of mice, revealed a preferential growth-inhibitory effect of this factor on the earlier progenitors.123 A second approach was to perform histologic analyses of hematopoietic tissues from mice treated with TGF-β1. Such studies revealed an inhibition of erythropoiesis and thrombopoiesis in TGF-β1–treated mice,124,125 whereas granulopoiesis was stimulated.124-126 Although some of these effects may be directly mediated, the possibility that administration of exogenous TGF-β in vivo may deregulate the production of other factors involved in the control of hematopoiesis should be taken into account. For example, an increased production of tumor necrosis factor (TNF)-α has been observed after the administration of TGF-β in mice.127 

Hematopoiesis in knockout mice

An opposing strategy has been to study in vivo the development of the hematopoietic tissue in the absence of endogenous TGF-β, or in a context in which the cell responsiveness to TGF-β is abrogated. For this purpose, a variety of knockout mice have been generated, in which a targeted disruption of genes encoding a TGF-β isoform or another element of the TGF-β signaling cascade has been performed. HomozygousTGF-β1 knockout mice have a 50% intrauterine death rate because of severe developmental retardation. Defective hematopoiesis, resulting in a reduced number of erythroid cells128 as well as a lack of Langerhans dendritic cells,129 has been correlated with the absence of TGF-β1. However, TGF-β1knockout mice also present defects in liver development130and bone formation,131 as well as many other dysregulations including autoimmune manifestations.132,133The phenotype of TβR-IIknockout mice has been reported to be indistinguishable from that ofTGF-β1 knockout mice.134 Mice lacking endoglin show defective angiogenesis,135 providing a good animal model of HHT.136,TGF-β3knockout mice show abnormal lung and craniofacial development due to altered epidermal–mesenchymal interactions,137 but these mice do not provide clear information concerning the role of this gene in the development of hematopoietic tissue. Knockout mice for theTGF-β2 gene exhibit a wide range of developmental defects that do not overlap with those of the other TGF-β knockout phenotypes.138 Concerning the Smad genes,Smad3 knockout mice demonstrate defects in immune function,139 whereas Smad4 and Smad5knockout mice have multiple embryonic and extraembryonic defects.140,141 

Human and murine hematopoietic stem/progenitor cells are usually in a quiescent or slow cycling state in adults.142,143 As suggested by in vivo studies, TGF-β is a good candidate for controlling this quiescence. This possible function of TGF-β has been studied extensively in vitro using clonal semisolid colony-forming assays, stroma-supported culture systems, and single-cell liquid cultures in both the murine and human hematopoietic systems.

TGF-β exerts a preferential growth-inhibitory effect on the most primitive stem/progenitor cells

A first approach has been to study the effects of exogenous TGF-β added to clonal cultures of hematopoietic progenitors. The first TGF-β isoform, TGF-β1, has been shown to inhibit colony formation by early murine144-146 and human hematopoietic progenitors in semisolid media,147-149 but not that of late progenitors.145,147-149 

In these studies, the effects of TGF-β on colony formation have often been tested in combination with only 1 or 2 other exogenous growth factors, added to serum-containing or conditioned culture media that contain undefined combinations of other growth factors. The cytokines used were mainly interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), and/or erythropoietin (Epo). In such culture conditions, TGF-β1 was found to efficiently inhibit colony formation by early human multipotential progenitors (CFU-Mix) at concentrations between 10 and 100 pg/mL, whereas later progenitors were less affected or were, in contrast, stimulated by these low TGF-β1 concentrations.149 

An alternative approach has been to use blocking antibodies or antisense oligonucleotides to neutralize TGF-β secreted by cells and present in the culture media. A study performed at a clonal level in semisolid or in single-cell liquid assays has revealed that the quiescence of human stem/progenitor cells is controlled in part through an autocrine loop involving TGF-β1 with the retinoblastoma susceptibility gene Rb as a downstream effector.150 In this study, blocking of autocrine TGF-β1 was sufficient to release from quiescence primitive erythro-myeloid (CFU-Mix), myeloid (CFU-GM), and erythroid progenitors (BFU-E), which then gave rise to macroscopic colonies, in the presence of IL-3, IL-6, granulocyte colony-stimulating factor (G-CSF), and Epo. Using the same antisense oligonucleotide, it was observed that autocrine TGF-β1 inhibits colony formation by early murine and human hematopoietic stem/progenitor cells stimulated with KIT ligand (SF).151,152 

Since the first studies on TGF-β, many others have subsequently addressed its effects on progenitor cell growth when present in association with various other cytokines. For example, in a single-cell liquid assay, it has been demonstrated that this factor directly inhibits early human hematopoietic progenitor cell proliferation in the presence of Epo, SF, GM-CSF, and IL-3.153 Moreover, in a semisolid culture system containing a combination of 7 stimulatory growth factors (IL-3, IL-6, IL-11, Epo, SF, GM-CSF, and G-CSF), it has been shown that whereas 10 to 30 pg/mL of TGF-β1 is sufficient to inhibit 90% of primitive high proliferative potential (HPP)-Mix, 100 to 300 pg/mL of TGF-β1 is required to inhibit 70% of the bipotent HPP-GM and the HPP–BFU-E. Concentrations of up to 1000 to 3000 pg/mL of TGF-β1 had little or no effect on the development of late CFU-G and CFU-M.154 

Preferential growth inhibition of the most primitive human hematopoietic cells has also been reported in studies in which the effects of anti–TGF-β1 antisense oligonucleotides, which neutralize autocrine TGF-β1 production155 or exogenous TGF-β1,156 were investigated on CD34+CD38 cells and on CD34+CD38+ cells. It was indeed observed that primitive CD34+CD38 cells show a high sensitivity to cell-cycle inhibition by TGF-β1, whereas more mature CD34+CD38+ cells are poorly affected or are even stimulated by TGF-β1. Interestingly, the concentrations of TGF-β1 for which these inhibitory effects were observed on primitive cells correspond to those detected in human plasma in an active form (usually less than 300 pg/mL).157 

The second isoform, TGF-β2, has also been shown to inhibit early progenitor cell proliferation in both the human148,149,158and murine hematopoietic systems,144 but with a lower efficiency.149,158 The third isoform, TGF-β3, also inhibits colony formation by early human hematopoietic progenitors149 or slows their rate of proliferation159 at least as efficiently as does TGF-β1. However, only TGF-β1 and -β2 were shown to exert bidirectional effects on proliferation of early and late hematopoietic progenitors, whereas the effects of TGF-β3 were only inhibitory.149 

TGF-β in stroma-supported cultures of hematopoietic cells

Stroma-supported cultures containing both hematopoietic progenitors and nonhematopoietic accessory cells allow the proliferation and differentiation of primitive stem/progenitor cells for several weeks without the addition of exogenous factors (for reviews see Dexter,160 1979; Eaves et al,1611991). In these systems, progenitor cell development is regulated through a complex interaction between positive and negative factors that are secreted by both stromal and hematopoietic cells.

Studies performed on adherent bone marrow primary stromal cells or cell lines have shown that these cells can produce a variety of growth factors, including cytokines such as G-CSF; GM-CSF162,163; IL-1β, IL-6, and IL-7162,164; SF and M-CSF165; and thrombopoietin (TPO).166They also produce chemokines such as the monocyte chemoattractant protein-1 and the interferon-inducible protein-10,165 as well as amounts of active TGF-β sufficient to control the proliferation of hematopoietic progenitors.162,167 Two aspects render the presence of TGF-β in stroma-supported cultures critical for the development of hematopoietic cells. First, TGF-β acts directly on these cells. Second, TGF-β is able to modulate the growth factor production by stromal cells,163-166 a process that indirectly controls their development.

The term “long-term culture initiating cell” (LTC-IC) has been assigned to a subpopulation of primitive human hematopoietic stem/progenitor cells that possess the potential to sustain continuous production of progenitors for at least 8 weeks in the presence of stroma.161 Anti–TGF-β added to stroma-supported cultures of human hematopoietic stem/progenitor cells was able to prolong or reactivate the proliferation of LTC-ICs,162implicating TGF-β as an endogenous inhibitor of primitive hematopoietic cells. The proliferation of these cells was also selectively inhibited when exogenous TGF-β was added.168 

Similar observations have been reported for murine hematopoietic stem/progenitor cells cultured according to the Dexter method. Dexter-type culture systems consist of total marrow cultures in which an expansion of primitive hematopoietic stem/progenitor cells is maintained for several months because of the presence of a bone marrow–derived adherent layer consisting of different types of nonhematopoietic accessory cells including adipocytes.169The addition of antibodies neutralizing the biologic activity of TGF-β1, -β2, and -β3 in such cultures resulted in a significant increase in early hematopoietic cells, demonstrating the ability of these factors to inhibit their cell cycling.170 

These stroma-supported in vitro culture systems have provided important information concerning the role of TGF-β in the control of the cell-cycle status of primitive murine and human hematopoietic stem/progenitor cells. However, they do not permit discrimination between the effect of TGF-β secreted by stromal cells (paracrine TGF-β) and that of TGF-β produced by the hematopoietic progenitors themselves (autocrine and paracrine TGF-β).

Remarks

All of these in vitro studies converge to suggest that TGF-β inhibits preferentially the cell cycling of the most primitive hematopoietic cells. These results have also been reproduced in an in vivo model. Indeed, nonobese diabetic/severe combined immunodeficient mice have been transplanted with human bone marrow and umbilical cord blood cells and injected with TGF-β1 at 6 weeks after transplantation. Analysis of the specific responses of different progenitor cell types has revealed that TGF-β1 is active on primitive progenitor cell populations, including LTC-IC and HPP–colony-forming cells, but not on the more mature cells.171 

It should be pointed out that differences in the responsiveness to TGF-β have been observed between human stem/progenitor cells of distinct ontogenic origins. Indeed, fetal liver stem/progenitor cells, which are generally in a more active cycling status than cells from adult bone marrow, appear to be less sensitive to growth inhibition by TGF-β.172 

TGF-β modulates growth factor receptor expression

The ability of TGF-β to modulate the expression of cytokine receptors has been observed both in transformed cell lines and in normal hematopoietic progenitors. In murine progenitor cell lines, TGF-β has been shown to down-modulate the expression of the receptors for IL-1,173 IL-3, G-CSF, and GM-CSF.174 In normal hematopoietic bone marrow cells, similar observations were made in humans for the IL-1 receptor173 and in mice for the IL-1, IL-3, and the M-CSF receptors, but not for the GM-CSF receptor which, on the contrary, is up-modulated by TGF-β on murine progenitor cells.175,176 G-CSF receptor expression was not affected.176 These effects appear to be isoform dependent. For example, TGF-β3 may be a more potent inhibitor of IL-3 receptor expression than TGF-β1, whereas TGF-β3 is less efficient than TGF-β1 in stimulating the expression of the GM-CSF receptor.176 

More recently, TGF-β1 has been reported to down-modulate the c-KIT receptor on murine177,178 and human hematopoietic progenitors,179 explaining its ability to inhibit the action of SF.152,180 A similar modulation of the FLT3 receptor has been described on early human hematopoietic progenitors,181 which is consistent with previous data showing an inhibition of FL-stimulated cell cycling of murine progenitors by TGF-β1.182,183 TGF-β1 has also been shown to down-modulate the IL-6 receptor (IL-6R) on human hematopoietic progenitors.181 Finally, the cell-surface expression of another important receptor involved in the control of primitive hematopoietic progenitor cell proliferation, the TPO receptor c-MPL, has been found to be down-modulated by TGF-β1 (Fortunel et al, unpublished results). This explains the ability of TGF-β1 to abrogate TPO-induced progenitor cell growth.184 

The HPP-Q working model

The concept of the high-proliferative-potential quiescent cell (HPP-Q) has been introduced to refer to a primitive subpopulation of human hematopoietic stem/progenitor cells that are highly sensitive to growth inhibition by TGF-β1.181 TGF-β1 maintains these cells in a quiescent or slow cycling state by down-modulating various cytokine receptors, preventing them from responding rapidly to mitogenic stimulation (Figure 2). In the in vitro HPP-Q assay, which can be performed both in semisolid conditions and in single-cell liquid cultures,150,181the use of antisense oligonucleotides or anti–TGF-β1 blocking antibodies allows these primitive cells to escape from cell-cycle inhibition. A short treatment with anti–TGF-β1 is sufficient to activate HPP-Q cells and render them competent and responsive to cytokines (Figure 2). Indeed, similar effects were obtained when cells were pretreated for only 6 to 12 hours before plating and when anti–TGF-β1 was maintained or added repeatedly throughout the culture period (Hatzfeld A et al, unpublished results).

Fig. 2.

A working model to explain the inhibition of primitive hematopoietic stem/progenitor cell cycling by TGF-β1.

Fig. 2.

A working model to explain the inhibition of primitive hematopoietic stem/progenitor cell cycling by TGF-β1.

Close modal

The HPP-Q population presents at least 3 characteristics of a primitive stem/progenitor cell compartment: (1) HPP-Q cells possess a high proliferative potential, as they are able to generate, within 18 days, clones containing more than 105 cells when cultured in a liquid medium in single-cell conditions; (2) they give rise to multilineage hematopoietic clones or colonies when cultured in the presence of the appropriate cytokine combination; and (3) they are in a quiescent or slow cycling state in spite of their high proliferative potential. A fourth characteristic of the HPP-Q cell population is under study. It concerns the ability of HPP-Q cells to promote long-term engraftment.

One main interest of this working model is the possibility of further phenotyping HPP-Q stem/progenitor cells on the basis of cytokine receptor expression. These cells express low levels of various cytokine receptors such as KIT, FLT3, IL-6R, and MPL. This renders possible the sorting of cell subpopulations enriched in HPP-Q stem/progenitor cells. The long-term engraftment capacity of CD34+/ KITlow/ FLT3low/ IL-6Rlow/ MPLlow cells should be studied in xenograft animal models such as preimmune sheep fetuses.185 Up to now, in this sheep model, the CD34+/ KITlow population, which includes HPP-Q cells, has been reported to contain the marrow-engrafting stem/progenitor cells. This was not the case for KIThigh and KITneg cells.186 

The identification of growth inhibitors able to control the proliferation of leukemic cells is of particular importance in the development of therapeutic approaches. As stated earlier, the acquisition of resistance to growth inhibition by TGF-β is often associated with malignant transformation. As discussed later, the selective advantage of cancer cells that are insensitive to TGF-β may be due not only to the inactivation of mechanisms involved in cell-cycle control, but also could result from resistance to apoptotic cell death. Indeed, TGF-β1 has been reported to induce cell death in several leukemic cell types, including myeloid leukemia–derived cell lines187 and B-lymphoma cell lines,188,189 as well as in primary leukemia cells such as acute myelogenous cells190 and lymphoblastic leukemia cells.191This suggests that the growth-inhibitory effect exerted by TGF-β on some leukemic blasts could occur in part through programmed cell death and, as a consequence of this, that abrogation of TGF-β1–mediated apoptosis could contribute to the pathogenesis of leukemia in the case of cells that have lost their sensitivity to TGF-β by mutation ofTβR-I or TβR-II. On the contrary, TGF-β1 has been reported to protect other types of leukemia-derived cells, such as HL-60 cells, from apoptosis.192,193 

The relations between TGF-β and the control of apoptosis have also been studied in various types of normal hematopoietic cells and appear to be strictly dependent on the differentiation status of the cells and on the culture variables such as the concentrations of cytokines. For example, TGF-β has been found to induce apoptosis in human eosinophils,194 whereas this factor has been found to sustain viability of rat195 and human T cells196 and also of human dendritic cell precursors.197 Whether TGF-β is an inducer of apoptosis for hematopoietic stem/progenitor cells is a question frequently raised. In a previous paragraph, we stressed the fact that TGF-β injected in mice only transiently inhibited the proliferation of primitive hematopoietic stem/progenitor cells. This suggests a reversible mechanism, which is not compatible with induction of cell death. Reversibility also has been suggested by in vitro studies in which TGF-β1 was found to delay proliferation of primitive murine198 and human hematopoietic stem/progenitor cells199 in batch cultures. However, some properties of TGF-β are consistent with both apoptotic and antiapoptotic effects exerted on stem/progenitor cells, which may lead to confusion. Indeed, TGF-β1 has been found to abrogate Fas-induced apoptosis of murine bone marrow progenitors,200 while counteracting the ability of growth factors such as SF201 and FL202 to sustain their viability. The reversibility of the growth-inhibitory effect exerted by TGF-β1 on hematopoietic stem/progenitor cells in vitro has been verified recently at a clonal level.203 In this study, the proliferative potential of single human umbilical cord blood CD34+ cells was analyzed after a 3-day culture in the presence of TGF-β1. A reversible cell-cycle inhibition was observed on primitive stem/progenitors, without detectable cell-death induction. In another study, it was demonstrated that the autocrine production of TGF-β1 by human peripheral blood cycling stem/progenitor cells contributed to sustaining their survival throughout successive cell divisions, in part by maintaining a high expression of the antiapoptotic protein bcl-2.204 

TGF-β controls multiple phases of erythropoiesis

TGF-β is one of the key regulators of erythropoiesis because it is involved in the control of both early and later stages of erythroid progenitor cell development (Figure 3). The first isoform, TGF-β1, has been identified as a cell-cycle inhibitor for early human and mouse BFU-E, but not for later erythroid progenitors, which are poorly affected by its growth-inhibitory effect.150,154,155,205 This negative regulation occurs in part through an autocrine negative-control loop.150 It has been reported that TGF-β2 and TGF-β3 could also inhibit the development of human BFU-E,149 but the effects of these 2 isoforms on erythropoiesis have not been investigated in detail.

Fig. 3.

Regulatory effects of TGF-β on the growth/proliferation and maturation/differentiation of developmentally distinct hematopoietic cells.

BFU indicates burst-forming unit; CFU, colony-forming unit; HPP, high proliferative potential; DC, dendritic cell; E, erythroid cell; G, granulocyte; M, monocyte/macrophage; and Mk, megakaryocyte.

Fig. 3.

Regulatory effects of TGF-β on the growth/proliferation and maturation/differentiation of developmentally distinct hematopoietic cells.

BFU indicates burst-forming unit; CFU, colony-forming unit; HPP, high proliferative potential; DC, dendritic cell; E, erythroid cell; G, granulocyte; M, monocyte/macrophage; and Mk, megakaryocyte.

Close modal

In contrast, TGF-β1 has been reported to exert a positive effect on the growth of a subset of later human erythroid progenitors. Interestingly, this effect has been observed in the presence of very low concentrations of TGF-β1 (less than 30 pg/mL), which otherwise selectively inhibit the growth of the most primitive progenitors.154 A growth-inducing effect of TGF-β1 has also been observed in an avian model system, in which TGF-β1 sustains erythroid progenitor cell proliferation and self-renewal. In the case of these avian cells, this effect occurs through a cooperation between the TGF-β and the TGF-α receptors via the Mek-Map kinase pathway,206 but this mechanism has not yet been investigated in a mammalian erythroid system.

In addition to its role in the control of erythroid progenitor cell proliferation, TGF-β1 is involved in the regulation of erythroid differentiation. Indeed, studies performed using the erythroleukemic cell lines K-562 and UT-7 as models have shown that TGF-β1 promotes late erythroid maturation processes such as the synthesis and accumulation of hemoglobin.207,208 A differentiating effect of TGF-β1 on erythroid cells has also been reported in nontransformed human cells by Krystal et al,209 who demonstrated that TGF-β1 triggers the conversion of early BFU-E into later BFU-E or CFU-E. These authors also showed that TGF-β1 up-modulates the expression of the differentiation marker glycophorin A and induces the synthesis of hemoglobin in mature cells.209 

As already described, TGF-β1 inhibits the effects of SF on early human and mouse hematopoietic progenitors by a functional down-modulation of its cell-surface receptor c-KIT.177-180This is theoretically sufficient to explain the inhibitory effect of TGF-β1 on early events of erythrocytic progenitor development. Indeed, among the various cytokines involved in controlling this process, SF may be one of the most potent factors triggering proliferation of early BFU-E. In the human hematopoietic system, the burst-promoting activity of SF is required for the growth of at least some early BFU-E.210 If this interaction does not occur, further development is prevented and the colony formation by BFU-E in semisolid culture medium is inhibited. Furthermore, it has been shown that retroviral-mediated gene transfection of c-kitinto human umbilical cord blood progenitors enhances erythroid colony formation and decreases their sensitivity to growth inhibition by TGF-β1.211 The ability of TGF-β1 to counteract the activity of SF also explains its positive effect on the maturation and differentiation of both K-562 cells and normal human erythrocytic progenitors. Indeed, in addition to its mitogenic activity, SF has been shown to retard the erythroid differentiation program.212,213 

Another key regulator of erythropoiesis is Epo. In addition to being the principal factor promoting the terminal differentiation of erythrocytes, this cytokine is able to stimulate earlier stages of erythropoiesis. Indeed, Epo is well known to stimulate the growth and development of CFU-E, and has been shown to sustain the proliferation of some BFU-E, but only if anti–TGF-β is added to the culture medium.214 Studies performed on normal mouse and rat erythroid progenitors have revealed that the potent growth-inducing activity of Epo on BFU-E is counteracted by TGF-β1,214,215 the effect of which could be explained by a down-modulation of the Epo receptor by TGF-β1. This possibility has been verified in the human leukemic cell line UT-7,215 but not yet for normal erythroid progenitors.

Other reports provide convincing evidence that TGF-β1 and the retinoblastoma gene product pRb could function through a common pathway to inhibit the cell cycling of early BFU-E and to promote erythroid differentiation. Indeed, TGF-β1–induced cell-cycle arrest has been found to be mediated through the control of pRb function in human hematopoietic stem/progenitor cells, including primitive CFU-Mix and BFU-E.150 Moreover, several studies performed on human and murine erythroleukemic cell lines,216-218 as well as on normal human erythroid progenitors,219 have clearly demonstrated the involvement of pRb in the regulation of later events in erythroid differentiation. Furthermore, analyses of hematopoietic tissues derived from Rb−/− knockout mice showed lineage-specific abnormalities in the development and maturation of erythroid cells.220-222 This confirms the role of pRb in the regulation of cellular events associated with the maturation and differentiation of normal erythroid cells, processes that are also controlled by TGF-β1.

Bidirectional effects of TGF-β1 on granulocytic and monocytic/macrophagic cell development

Mature granulocytic (G) and monocytic/macrophagic (M) cells develop from a common bipotent myeloid progenitor (CFU-GM). Both positive and negative effects of TGF-β on myelopoiesis have been described in vitro (Figure 3). Inhibitory effects were mainly observed on the earlier bipotent myeloid progenitors, whereas cells in later stages were either slightly inhibited or even stimulated, depending on the other factors present. TGF-β1 added to a semisolid medium containing IL-3, IL-6, IL-11, Epo, SF, GM-CSF, and G-CSF has been found to inhibit the growth of early human HPP-GM, but not that of late CFU-G and CFU-M.154 In contrast, when the TGF-β1 regulatory effects were studied in the presence of individual cytokines, clear stimulating effects were observed on the middle and late stages of myeloid cell development. As an example, in culture media containing only GM-CSF and TGF-β1, these 2 factors were found to synergize and then promote the development of a population of human mid-stage CFU-GM (day-7 CFU-GM), whereas earlier CFU-GM (day-14 CFU-GM) were not stimulated.149 

The effects of TGF-β1 on the development of the G and M cell lineages have also been analyzed separately. TGF-β1 was found to synergize with GM-CSF to stimulate the growth and terminal differentiation of murine granulocytic progenitors.175 Both positive and negative effects of TGF-β1 on the proliferation of macrophagic cells have been described. Indeed, TGF-β1 has been shown to inhibit GM-CSF– and M-CSF–dependent proliferation of early murine marrow-derived macrophages, whereas proliferation of later ones was enhanced.223 Another study performed on murine cells showed a positive effect of TGF-β1 on GM-CSF–induced macrophage proliferation but an inhibitory effect on the proliferation of macrophages stimulated by M-CSF.224 As shown for granulocytes, TGF-β1 can also promote the maturation and differentiation of monocytic cells.225 The roles of TGF-β2 and -β3 in the control of myelopoiesis have been less well studied than that of TGF-β1. However, it appears that TGF-β2 can exert bidirectional proliferative effects on myeloid cells as does TGF-β1, whereas the effect of TGF-β3 is only inhibitory.149 

In contrast to what has been observed for many cytokine receptors controlling the cell cycling of hematopoietic stem/progenitor cells, TGF-β1 has been shown to up-modulate the GM-CSF receptor on murine myeloid progenitors.175,176 This is in accordance with its ability to enhance GM-CSF–induced growth and differentiation of granulocytic and monocytic/macrophagic cells. TGF-β3 was less potent than TGF-β1 in up-modulating this receptor, which is in agreement with its inability to stimulate late myelopoiesis efficiently.176 TGF-β1 has also been shown to increase the cell-surface expression of the M-CSF receptor in human fetal liver monocytic progenitors,225 suggesting a possible positive cooperation between these factors. Thus, because TGF-β1 is secreted by myeloid cells at various stages of their development, this factor could regulate myelopoiesis through both negative and positive autocrine and paracrine loops.

Control of early and late megakaryopoiesis by TGF-β

Megakaryopoiesis includes 2 major steps that lead to the production of platelets. The first step of this process consists of progenitor expansion, which produces large numbers of megakaryocytic precursors. These cells subsequently differentiate into mature megakaryocytes (Mks), which produce platelets.

Early megakaryocytic progenitors first enter into a proliferating state when stimulated by appropriate growth factors. For instance, IL-3, IL-6, IL-11, SF, and TPO have been identified as potent “early-acting” Mk-stimulating factors.226-230 In contrast, several studies have shown TGF-β to inhibit early stages of megakaryopoiesis,227,231,232 as reported for other hematopoietic cell lineages (Figure 3). Addition of exogenous TGF-β1 to clonal semisolid cultures of murine bone marrow cells has been shown to inhibit the growth of primitive HPP-Mk stimulated by IL-3, IL-6, and IL-11.227 A similar effect of TGF-β1 was reported for early human Mk progenitors cultured in the presence of IL-3 and IL-6.232 In contrast, when anti–TGF-β blocking antibodies were added to the cultures, an increased proliferation of rat megakaryocytes was observed.231 

TGF-β has been reported to promote maturation and late differentiation processes in several hematopoietic cell lineages. However, this is not the case for the megakaryocytic lineage (Figure3). The second step of megakaryocyte development consists of endomitosis, leading to the generation of mature polyploid cells, which then differentiate into platelets. This process is promoted by “late-acting” Mk potentiators including IL-6, IL-11, and TPO.229,233,234 TGF-β has also been shown to exert an inhibitory effect on these late events of megakaryopoiesis. Inhibition of endomitosis by TGF-β1 was first observed using the human megakaryocytic cell line Dami as a model,235,236 and this effect has since been reproduced using rat megakaryocytes.231 

TGF-β1 is produced in large amounts by megakaryocytes and platelets. Once synthesized, it is sequestered within the intracytoplasmic α-granules237,238 and is released by cells spontaneously or in response to stimulation by growth factors such as IL-3 and, to a lesser extent, IL-1 or IL-11.239 Because TGF-β1 inhibits efficiently both early and late stages of megakaryocytic development, this factor could be considered as a possible feedback regulator of megakaryopoiesis. The physiologic relevance of this regulatory mechanism has been clearly demonstrated by studies of human thrombocythemia, although TGF-β is obviously not the only factor responsible for this complex process. In these pathologies, abnormally increased platelet production has been reported to be due to a decreased sensitivity of Mk progenitors to TGF-β1.240,241 

We have described the ability of TGF-β1 to down-modulate the expression of several cytokine receptors, including the MPL receptor, on primitive hematopoietic stem/progenitor cells (Fortunel et al, unpublished results). Because TPO is probably one of the main factors promoting both megakaryocyte proliferation and differentiation,233,242 such an effect could explain at least in part the inhibitory effects of TGF-β1 on megakaryocytic cell development. Receptor modulation studies performed at distinct developmental stages of megakaryopoiesis would help to elucidate this point. Other studies have provided evidence that the control of late megakaryocytic differentiation by TGF-β1 could involve the transcription factors c-jun and c-fos as downstream effectors.243,244 

TGF-β and the development of dendritic antigen-presenting cells

To discuss the effects of TGF-β on lymphoid cell lineages would require a specific review. Here we will just mention the role of TGF-β in the regulation of the development of cells of the immune system by introducing the case of dendritic cells (DCs) (Figure 3). TGF-β contributes to the generation of DCs, which correspond to a particular population of leukocytes specialized in antigen presentation for T-cell responses. DCs can be obtained in vitro from human CD34+ cells,197,245 murine lineage-negative (Lin)KIT+ cells,246 human monocytes,247-249 and from their precursors.250 

In vitro studies have demonstrated that the growth-factor requirement for the development of DCs includes TGF-β1, which is in agreement with the in vivo observation that TGF-β1 knockout mice lack DCs.129 This factor has been shown to promote efficiently the generation of DCs from CD34+ progenitors, in combination with GM-CSF, SF, TNF-α,245 and FL,251 in part by protecting progenitors from apoptosis.197 

Generation of human and mouse DCs in response to TGF-β1 may occur via a monocyte/macrophage differentiation pathway.246-250 Indeed, TGF-β1 has been found to induce differentiation of human peripheral blood monocytes into DCs, in cooperation with cytokines such as GM-CSF and IL-4,247 and to prevent their noncognate maturation.248 Similarly, TGF-β1 has been reported to promote the generation of DCs from human CD14+ precursor cells, when present in association with GM-CSF and TNF-α.250 This was also the case for murine monocytic/macrophagic cells cultured with the same cytokine combination.246 In contrast, generation of DCs independently of the monocytic/macrophagic cell lineage does not require the presence of TGF-β1.250 

Efficient retroviral-mediated gene transfer into stem/progenitor cells released from quiescence by anti–TGF-β

Progress in the identification and cloning of genes involved in human genetic diseases has led to important efforts to improve gene-transfer technology. Various types of viral vectors have been developed, each presenting different advantages. Among these, retroviral vectors ensure the insertion of a transgene into the host genome and its transmission to cell progeny. However, efficient retroviral-mediated gene transfection implies that the host cells are in a cycling state, which is not normally the case for primitive hematopoietic cells.252,253 Because autocrine and paracrine TGF-β1 produced by hematopoietic progenitors is partly responsible for their maintenance in a quiescent or slow cycling state,150,151 the use of anti–TGF-β1 blocking antibodies to neutralize the bioactivity of endogenous TGF-β1 during the retroviral transfection procedure was attempted to increase gene transfer into primitive stem/progenitor cells. This strategy has been tested with success on hematopoietic cells of several origins, including human umbilical cord blood,254 bone marrow,255 peripheral blood,256 and murine bone marrow.257 The efficiency of this method was shown to be improved by the coupled use of anti–TGF-β1 blocking antibodies with antisense oligonucleotides against the cyclin-dependent kinase inhibitor (CDKI) p27kip1.255 This approach could be extended to other effectors involved in the negative regulation of hematopoietic stem/progenitor cell cycling by TGF-β1, such as pRb150 or the CDKI p21cip1.258 

Moreover, it has been observed that CD34+ cells produce TGF-β1 during retroviral transfection protocols, promoting a return to quiescence of the most primitive stem/progenitor cells after gene transfer. The HPP-Q assay, which allows activation of these quiescent progenitors in vitro, permits an optimal evaluation of gene-transfer efficiency in these primitive hematopoietic stem/progenitor cells (Ducos et al, unpublished results).

In vitro amplification of stem/progenitor cells: key role of TGF-β?

The development of methods for in vitro expansion of hematopoietic stem/progenitor cells is an important goal for clinical research, given the therapeutic potential of these cells. The challenge is to obtain defined culture systems in which the cell cycling of primitive quiescent stem/progenitor cells is stimulated, while their maturation and senescence are prevented.

TGF-β1 present in a culture medium at concentrations from 30 to 300 pg/mL has been reported to up-modulate the expression of the immaturity marker CD34 on slowly cycling human progenitors,202promoting the maintenance of an immature stem/progenitor cell compartment (Figure 2). In this study, the expression of the mucinlike protein CD34 was rapidly lost on umbilical cord blood progenitors grown in a differentiating medium containing IL-3, IL-6, IL-11, Epo, SF, GM-CSF, and G-CSF. This was particularly the case when anti–TGF-β1 was added. In contrast, low TGF-β1 concentrations decreased cell proliferation but promoted the maintenance of a stem/progenitor cell subpopulation that still expressed a high level of CD34 after 4 divisions. These results suggest the ability of TGF-β1 to up-modulate the expression of a membrane antigen associated with the hematopoietic stem/progenitor cell undifferentiated phenotype. This stresses the importance of research into the potential use of TGF-β for the in vitro maintenance and production of stem cells for therapeutic purposes.

The pleiotropic effects of TGF-βs already observed in various tissues are particularly well illustrated in the hematopoietic system. Depending on the degree of maturation of the hematopoietic cells, these factors are able to control either positively or negatively cell proliferation, differentiation, or apoptosis. The importance of TGF-βs in the development of multicellular organisms is stressed by their high degree of conservation during evolution and by the fact that they are present in most tissues where they are stored in a latent form, ready for rapid local activation. Moreover, TGF-βs are often involved in autocrine control loops, which allow precise tuning at the cell level.

Despite the considerable numbers of studies performed to understand the complex regulatory role of TGF-βs in hematopoiesis—many of which could not be cited here because of space limitations—many questions remain. For example, the physiologic concentrations of active TGF-β that control each particular function in vivo remain unknown. This is of particular importance because different specific responses can be observed in vitro depending on the concentration of TGF-β used, which in the literature ranges from 10 pg/mL to more than 10 ng/mL. Moreover, it remains unclear whether TGF-βs, which block the cell cycle in G1,259 can promote a return to G0. A progressive disappearance of G1 mRNAs could explain the reversion to quiescence induced by TGF-β. Different degrees of cell-cycle inhibition by TGF-βs may exist, ranging from profound quiescence to a slow cycling state. Whether hematopoietic stem/progenitor cells maintained in a slow cycling state by TGF-βs could be amplified in vitro more efficiently, without differentiation and senescence, must be studied further to define optimal in vitro culture systems for cell therapy.

In this review, we have focused on studies performed on TGF-β1, -β2, and -β3. Other members of the TGF-β superfamily, such as the BMPs, should also be considered as important regulators of hematopoiesis. Indeed, in addition to being involved in the control of bone formation, BMPs could regulate the developmental program of human hematopoietic stem cells, as do TGF-βs, including cell proliferation, differentiation, and stem cell survival.260 

Signal transduction by the members of the TGF-β family involves interactions among multiple possible receptors and intracellular signaling proteins, allowing the diversity of specific biologic responses. Many postgenomic studies will certainly be required to unravel the complexity of this key regulatory system. These studies will probably lead to the understanding of many different pathologies. In a few cases, gene therapy will perhaps enable the correction of a single gene mutation responsible for a disease. In most cases, it will not be easy to develop a drug sufficiently specific for one single combination of regulatory proteins in a specific cell type, without affecting other cells. Antisense oligonucleotides have been used successfully in vitro to neutralize the activity of genes involved in TGF-β signal transduction. This strategy has been efficient for short time effects, such as a rapid activation of quiescent stem/progenitor cells. However, these experiments were performed using purified cells, which is far from being representative of the in vivo situation, where it is difficult to target the treatment to a specific cell population. Promoters controlled by TGF-β, such as the promoter of collagen VII, which is involved in the pathogenesis of myelofibrosis, or oncogenes involved in leukemogenesis, such as Evi-1, could be potential targets to devise more specific drugs to treat diseases caused by dysregulation of TGF-β production or activity.

In conclusion, the development of therapeutic agents to correct, in vivo, dysfunctions of the TGF-β signaling pathway represents an important challenge for the future. However, a main difficulty will come from the pleiotropism of its regulatory role.

We are indebted to Dr Mary Osborne for her critical reading of the manuscript.

Supported by European Contract no. BIO4-CT96-0646, the Centre National de la Recherche Scientifique (CNRS), and the Association pour la Recherche sur le Cancer (ARC).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Moore
MA
Muench
MO
Warren
DJ
Laver
J
Cytokine network involved in the regulation of haemopoietic stem cell proliferation and differentiation.
Ciba Found Symp.
148
1990
43
61
2
Metcalf
D
Hematopoietic regulators: redundancy or subtlety?
Blood.
82
1993
3515
3523
3
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood.
81
1993
2844
2853
4
Levesque
JP
Hatzfeld
A
Hatzfeld
J
Mitogenic properties of major extracellular proteins.
Immunol Today.
12
1991
258
262
5
Simmons
PJ
Haylock
DN
Use of hematopoietic growth factors for in vitro expansion of precursor cell populations.
Curr Opin Hematol.
2
1995
189
195
6
De Larco
JE
Todaro
GJ
Growth factors from murine sarcoma virus-transformed cells.
Proc Natl Acad Sci U S A.
75
1978
4001
4005
7
Roberts
AB
Lamb
LC
Newton
DL
Sporn
MB
De Larco
JE
Todaro
GJ
Transforming growth factors: isolation of polypeptides from virally and chemically transformed cells by acid/ethanol extraction.
Proc Natl Acad Sci U S A.
77
1980
3494
3498
8
Sporn
MB
Roberts
AB
Introduction: what is TGF-β? In clinical applications of TGF-β.
Ciba Found Symp.
157
1991
1
6
9
Derynck
R
Jarrett
JA
Chen
EY
et al
Human transforming growth factor β complementary DNA sequence and expression in normal and transformed cells.
Nature.
316
1985
701
705
10
de Martin
R
Haendler
B
Hofer-Warbinek
R
et al
Complementary DNA for human glioblastoma-derived T cell suppressor factor, a novel member of the transforming growth factor-β gene family.
EMBO J.
6
1987
3673
3677
11
ten Dijke
P
Hansen
P
Iwata
KK
Pieler
C
Foulkes
JG
Identification of another member of the transforming growth factor type β gene family.
Proc Natl Acad Sci U S A.
85
1988
4715
4719
12
Jakowlew
SB
Dillard
PJ
Sporn
MB
Roberts
AB
Complementary deoxyribonucleic acid cloning of a messenger ribonucleic acid encoding transforming growth factor β4 from chicken embryo chondrocytes.
Mol Endocrinol.
2
1988
1186
1195
13
Kondaiah
P
Sands
MJ
Smith
JM
et al
Identification of a novel transforming growth factor β (TGF-β5) mRNA in Xenopus laevis.
J Biol Chem.
256
1990
1089
1093
14
Kothapalli
R
Buyuksal
I
Wu
SQ
Chegini
N
Tabibzadeh
S
Detection of ebaf, a novel human gene of the transforming growth factor β superfamily: association of gene expression with endometrial bleeding.
J Clin Invest.
99
1997
2342
2350
15
Gentry
LE
Lioubin
MN
Purchio
AF
Marquardt
H
Molecular events in the processing of recombinant type 1 pre-pro-transforming growth factor-β to the mature polypeptide.
Mol Cell Biol.
8
1988
4162
4168
16
Lawrence
DA
Identification and activation of latent transforming growth factor-β.
Methods Enzymol.
198
1991
327
336
17
Gleizes
PE
Munger
JS
Nunes
I
et al
TGF-β latency: biological significance and mechanisms of activation.
Stem Cells.
15
1997
190
197
18
Ogawa
Y
Schmidt
DK
Dasch
JR
Chang
RJ
Glaser
CB
Purification and characterization of transforming growth factor-β 2.3 and -β1.2 heterodimers from bovine bone.
J Biol Chem.
267
1992
2325
2328
19
Miyazono
K
Hellman
U
Wernstedt
C
Heldin
CH
Latent high molecular weight complex of transforming growth factor β1. Purification from human platelets and structural characterization.
J Biol Chem.
263
1988
6407
6415
20
Wakefield
LM
Smith
DM
Flanders
KC
Sporn
MB
Latent transforming growth factor-β from human platelets. A high molecular weight complex containing precursor sequences.
J Biol Chem.
263
1988
7646
7654
21
Yin
W
Smiley
E
Germiller
J
et al
Isolation of a novel latent transforming growth factor-β binding protein gene (LTBP-3).
J Biol Chem.
270
1995
10147
10160
22
Moren
A
Olofsson
A
Stenman
G
et al
Identification and characterization of LTBP-2, a novel latent transforming growth factor-β-binding protein.
J Biol Chem.
269
1994
32469
32478
23
Saharinen
J
Taipale
J
Monni
O
Keski-Oja
J
Identification and characterization of a new latent transforming growth factor-β-binding protein, LTBP-4.
J Biol Chem.
273
1998
18459
18469
24
Miyazono
K
Olofsson
A
Colosetti
P
Heldin
CH
A role of the latent TGF-β1-binding protein in the assembly and secretion of TGF-β1.
EMBO J.
10
1991
1091
1101
25
Munger
JS
Harpel
JG
Gleizes
PE
Mazzieri
R
Nunes
I
Rifkin
DB
Latent transforming growth factor-β: structural features and mechanisms of activation.
Kidney Int.
51
1997
1376
1382
26
Taipale
J
Keski-Oja
J
Growth factors in the extracellular matrix.
FASEB J.
11
1997
51
59
27
Dallas
SL
Miyazono
K
Skerry
TM
Mundy
GR
Bonewald
LF
Dual role for the latent transforming growth factor-β binding protein in storage of latent TGF-β in the extracellular matrix and as a structural matrix protein.
J Cell Biol.
131
1995
539
549
28
Taipale
J
Miyazono
K
Heldin
CH
Keski-Oja
J
Latent transforming growth factor-β1 associates to fibroblast extracellular matrix via latent TGF-β binding protein.
J Cell Biol.
124
1994
171
181
29
Taipale
J
Lohi
J
Saarinen
J
Kovanen
PT
Keski-Oja
J
Human mast cell chymase and leukocyte elastase release latent transforming growth factor type β1 from the extracellular matrix of cultured human epithelial and endothelial cells.
J Biol Chem.
270
1995
4689
4696
30
Olofsson
A
Ichijo
H
Moren
A
ten Dijke
P
Miyazono
K
Heldin
CH
Efficient association of an amino-terminally extended form of human latent transforming growth factor-β binding protein with the extracellular matrix.
J Biol Chem.
270
1995
31294
31297
31
Brown
PD
Wakefield
LM
Levinson
AD
Sporn
MB
Physicochemical activation of recombinant latent transforming growth factor-β's 1, 2, and 3.
Growth Factors.
3
1990
35
43
32
Jullien
P
Berg
TM
Lawrence
DA
Acidic cellular environments: activation of latent TGF-β and sensitization of cellular responses to TGF-β and EGF.
Int J Cancer.
43
1989
886
891
33
Barcellos-Hoff
MH
Dix
TA
Redox-mediated activation of latent transforming growth factor-β1.
Mol Endocrinol.
10
1996
1077
1083
34
Lyons
RM
Keski-Oja
J
Moses
HL
Proteolytic activation of latent transforming growth factor β from fibroblast-conditioned medium.
J Cell Biol.
106
1988
1659
1665
35
Lyons
RM
Gentry
LE
Purchio
AF
Moses
HL
Mechanism of activation of latent transforming growth factor β1 by plasmin.
J Cell Biol.
110
1990
1361
1367
36
Dennis
PA
Rifkin
DB
Cellular activation of latent transforming growth factor β requires binding to the cation-independent mannose 6-phosphate/insulin-like growth factor type II receptor.
Proc Natl Acad Sci U S A.
88
1991
580
584
37
Nunes
I
Shapiro
RL
Rifkin
DB
Characterization of latent TGF-β activation by murine peritoneal macrophages.
J Immunol.
155
1995
1450
1459
38
Godar
S
Horejsi
V
Weidle
UH
Binder
BR
Hansmann
C
Stockinger
H
M6P/IGFII-receptor complexes urokinase receptor and plasminogen for activation of transforming growth factor-β1.
Eur J Immunol.
29
1999
1004
1013
39
Verderio
E
Gaudry
C
Gross
S
Smith
C
Downes
S
Griffin
M
Regulation of cell surface tissue transglutaminase. Effects on matrix storage of latent transforming growth factor-β binding protein-1.
J Histochem Cytochem.
47
1999
1417
1432
40
Kojima
S
Nara
K
Rifkin
DB
Requirement for transglutaminase in the activation of latent transforming growth factor β in bovine endothelial cells.
J Cell Biol.
121
1993
439
448
41
Nunes
I
Gleizes
PE
Metz
CN
Rifkin
DB
Latent transforming growth factor-β binding protein domains involved in activation and transglutaminase-dependent cross-linking of latent transforming growth factor-β.
J Cell Biol.
136
1997
1151
1163
42
Schultz-Cherry
S
Ribeiro
S
Gentry
L
Murphy-Ullrich
JE
Thrombospondin binds and activates the small and large forms of latent transforming growth factor-β1 in a chemically defined system.
J Biol Chem.
269
1994
26775
26782
43
Schultz-Cherry
S
Lawler
J
Joanne
E
Murphy-Ullrich
JE
The type I repeats of thrombospondin 1 activate latent transforming growth factor-β.
J Biol Chem.
269
1994
26783
26788
44
Crawford
SE
Stellmach
V
Murphy-Ullrich
JE
et al
Thrombospondin-1 is a major activator of TGF-β1 in vivo.
Cell.
93
1998
1159
1170
45
Miyazono
K
Heldin
CH
Role for carbohydrate structures in TGF-β1 latency.
Nature.
338
1989
158
160
46
Munger
JS
Huang
X
Kawakatsu
H
et al
The integrin α5 β6 binds and activates latent TGF β1: a mechanism for regulating pulmonary inflammation and fibrosis.
Cell.
96
1999
319
328
47
Attisano
L
Carcamo
J
Ventura
F
Weis
FM
Massagué
J
Wrana
JL
Identification of human activin and TGF β type I receptors that form heteromeric kinase complexes with type II receptors.
Cell.
75
1993
671
680
48
Ebner
R
Chen
RH
Shum
L
et al
Cloning of a type I TGF-β receptor and its effect on TGF-β binding to the type II receptor.
Science.
260
1993
1344
1348
49
Franzen
P
ten Dijke
P
Ichijo
H
et al
Cloning of a TGF β type I receptor that forms a heteromeric complex with the TGF β type II receptor.
Cell.
75
1993
681
692
50
Lin
HY
Wang
XF
Ng-Eaton
E
Weinberg
RA
Lodish
HF
Expression cloning of the TGF-β type II receptor, a transmembrane serine/threonine kinase.
Cell.
68
1992
775
785
51
Wrana
JL
Attisano
L
Wieser
R
Ventura
F
Massagué
J
Mechanism of activation of the TGF-β receptor.
Nature.
370
1994
341
347
52
Piek
E
Heldin
CH
ten Dijke
P
Specificity, diversity, and regulation in TGF-β superfamily signaling.
FASEB J.
13
1999
2105
2124
53
Segarini
PR
Seyedin
SM
The high molecular weight receptor to transforming growth factor-β contains glycosaminoglycan chains.
J Biol Chem.
263
1988
8366
8370
54
Lopez-Casillas
F
Cheifetz
S
Doody
J
Andres
JL
Lane
WS
Massagué
J
Structure and expression of the membrane proteoglycan βglycan, a component of the TGF-β receptor system.
Cell.
67
1991
785
795
55
Kaname
S
Ruoslahti
E
Betaglycan has multiple binding sites for transforming growth factor-β1.
Biochem J.
315
1996
815
820
56
Barbara
NP
Wrana
JL
Letarte
M
Endoglin is an accessory protein that interacts with the signaling receptor complex of multiple members of the transforming growth factor-β superfamily.
J Biol Chem.
274
1999
584
594
57
Moustakas
A
Lin
HY
Henis
YI
Plamondon
J
O'Connor-McCourt
MD
Lodish
HF
The transforming growth factor β receptors types I, II, and III form hetero-oligomeric complexes in the presence of ligand.
J Biol Chem.
268
1993
22215
22218
58
Lopez-Casillas
F
Wrana
JL
Massagué
J
Betaglycan presents ligand to the TGF-β receptors.
Cell.
73
1993
1435
1444
59
Lastres
P
Letamendia
A
Zhang
H
et al
Endoglin modulates cellular response to TGF-β1.
J Cell Biol.
133
1996
1109
1121
60
Cheifetz
S
Bellon
T
Cales
C
et al
Endoglin is a component of the transforming growth factor-β receptor system in human endothelial cells.
J Biol Chem.
267
1992
19027
19030
61
St-Jacques
S
Cymerman
U
Pece
N
Letarte
M
Molecular characterization and in situ localization of murine endoglin reveal that it is a transforming growth factor β binding protein of endothelial and stromal cells.
Endocrinology.
134
1994
2645
2657
62
O'Connell
PJ
McKenzie
A
Fisicaro
N
Rockman
SP
Pearse
MJ
D'Apice
AJ
Endoglin: a 180-kD endothelial cell and macrophage restricted differentiation molecule.
Clin Exp Immunol.
90
1992
154
159
63
Buhring
HJ
Muller
CA
Letarte
M
et al
Endoglin is expressed on a subpopulation of immature erythroid cells of normal human bone marrow.
Leukemia.
5
1991
841
847
64
Rokhlin
OW
Cohen
MB
Kubagawa
H
Letarte
M
Cooper
MD
Differential expression of endoglin on fetal and adult hematopoietic cells in human bone marrow.
J Immunol.
154
1995
4456
4465
65
Robledo
MM
Hidalgo
A
Lastres
P
et al
Characterization of TGF-β-binding proteins in human marrow stromal cells.
Br J Haematol.
93
1996
507
514
66
Massagué
J
Receptors for the TGF-β family.
Cell.
69
1992
1067
1070
67
Massagué
J
TGF-β signal transduction.
Annu Rev Biochem.
67
1998
753
791
68
Abdollah
S
Macias-Silva
M
Tsukazaki
T
Hayashi
H
Attisano
L
Wrana
JL
TβRI phosphorylation of Smad2 on Ser465 and Ser467 is required for Smad2-Smad4 complex formation and signaling.
J Biol Chem.
272
1997
27678
27685
69
Nakao
A
Imamura
T
Souchelnytskyi
S
et al
TGF-β1 receptor-mediated signalling through Smad2, Smad3 and Smad4.
EMBO J.
16
1997
5353
5362
70
Zhang
Y
Feng
X
We
R
Derynck
R
Receptor-associated Mad homologues synergize as effectors of the TGF-β response.
Nature.
383
1996
168
172
71
Hayashi
H
Abdollah
S
Qiu
Y
et al
The MAD-related protein Smad7 associates with the TGFβ receptor and functions as an antagonist of TGFβ signaling.
Cell.
89
1997
1165
1173
72
Imamura
T
Takase
M
Nishihara
A
et al
Smad6 inhibits signalling by the type β superfamily.
Nature.
389
1997
622
626
73
Nakao
A
Afrakhte
M
Moren
A
et al
Identification of Smad7, a TGF-β-inducible antagonist of TGF-β signalling.
Nature.
389
1997
631
635
74
Nishimura
R
Kato
Y
Chen
D
Harruis
SE
Mundy
GR
Yoneda
T
Smad5 and DPC4 are key molecules in mediating BMP2-induced osteoblastic differentiation of the pluripotent mesenchymal precursor cell line C2C12.
J Biol Chem.
273
1998
1872
1879
75
Bruno
E
Horrigan
SK
Van Den Berg
D
et al
The Smad5 gene is involved in the intracellular signaling pathways that mediate the inhibitory effects of transforming growth factor β on human hematopoiesis.
Blood.
91
1998
1917
1923
76
Le Bousse-Kerdiles
MC
Chevillard
S
Charpentier
A
et al
Differential expression of transforming growth factor-β, basic fibroblast growth factor, and their receptors in CD34+ hematopoietic progenitor cells from patients with myelofibrosis and myeloid metaplasia.
Blood.
88
1996
4534
4546
77
Rooke
HM
Vitas
MR
Crosier
PS
Crosier
KE
The TGF-β type II receptor in chronic myeloid leukemia: analysis of microsatellite regions and gene expression.
Leukemia.
13
1999
535
541
78
DeCoteau
JF
Knaus
PI
Yankelev
H
et al
Loss of functional cell surface transforming growth factor β (TGF-β) type I receptor correlates with insensitivity to TGF-β in chronic lymphocytic leukemia.
Proc Natl Acad Sci U S A.
94
1997
5877
5881
79
Lagneaux
L
Delforge
A
Bron
D
Massy
M
Bernier
M
Stryckmans
P
Heterogenous response of B lymphocytes to transforming growth factor-β in B-cell chronic lymphocytic leukaemia: correlation with the expression of TGF-β receptors.
Br J Haematol.
97
1997
612
620
80
Kadin
ME
Cavaille-Coll
MW
Gertz
R
Massagué
J
Cheifetz
S
George
D
Loss of receptors for transforming growth factor-β in human T-cell malignancies.
Proc Natl Acad Sci U S A.
91
1994
6002
6006
81
Knaus
PI
Lindemann
D
DeCoteau
JF
et al
A dominant inhibitory mutant of the type II transforming growth factor-β receptor in the malignant progression of a cutaneous T-cell lymphoma.
Mol Cell Biol.
16
1996
3480
3489
82
Schiemann
WP
Pfeifer
WM
Levi
E
Kadin
ME
Lodish
HF
A deletion in the gene for transforming growth factors β type I receptor abolishes growth regulation by transforming growth factors β in a cutaneous T-cell lymphoma.
Blood.
94
1999
2854
2861
83
Wieser
R
Gruber
B
Rieder
H
Fonatsch
C
Mutational analysis of the tumor suppressor Smad2 in acute lymphoid and myeloid leukemia.
Leukemia.
12
1998
1114
1118
84
Kurokawa
M
Mitani
K
Irie
K
et al
The oncoprotein Evi-1 represses TGF-β signalling by inhibiting Smad3.
Nature.
394
1998
92
96
85
Hirai
H
The transcription factor Evi-1.
Int J Biochem Cell Biol.
31
1999
1367
1371
86
Kurokawa
M
Mitani
K
Imai
Y
Ogawa
S
Yzaki
Y
Hirai
H
The t(3;21) fusion product, AML1/Evi-1, interacts with Smad3 and blocks transforming growth factor-β-mediated growth inhibition of myeloid cells.
Blood.
92
1998
4003
4012
87
Papadaki
HA
Giouremou
K
Eliopoulos
GD
Low frequency of myeloid progenitor cells in chronic idiopathic neutropenia of adults may be related to increased production of TGF-β1 by bone marrow stromal cells.
Eur J Haematol.
63
1999
154
162
88
Lagneaux
L
Delforge
A
Dorval
C
Bron
D
Stryckmans
P
Excessive production of transforming growth factor-β by bone marrow stromal cells in B-cell chronic lymphocytic leukemia inhibits growth of hematopoietic precursors and interleukin-6 production.
Blood.
82
1993
2379
2385
89
Le Bousse-Kerdiles
MC
Martyre
MC
Dual implication of fibrogenic cytokines in the pathogenesis of fibrosis and myeloproliferation in myeloid metaplasia with myelofibrosis.
Ann Hematol.
78
1999
437
444
90
Terui
T
Niitsu
Y
Mahara
K
et al
The production of transforming growth factor-β in acute megakaryoblastic leukemia and its possible implications in myelofibrosis.
Blood.
75
1990
1540
1548
91
Martyre
MC
Romquin
N
Le Bousse-Kerdiles
MC
et al
Transforming growth factor β and megakaryocytes in the pathogenesis of idiopathic myelofibrosis.
Br J Haematol.
88
1994
9
16
92
Rameshwar
P
Denny
TN
Stein
D
Gascon
P
Monocyte adhesion in patients with bone marrow fibrosis is required for the production of fibrogenic cytokines. Potential role for interleukin-1 and TGF-β.
J Immunol.
153
1994
2819
2830
93
Johnston
JB
Dalal
BI
Israels
SJ
et al
Deposition of transforming growth factor-β in the marrow in myelofibrosis, and the intracellular localization and secretion of TGF-β by leukemic cells.
Am J Clin Pathol.
103
1995
574
582
94
Vindevoghel
L
Lechleider
RJ
Kon
A
et al
SMAD3/4-dependent transcriptional activation of the human type VII collagen gene (COL7A1) promoter by transforming growth factor β.
Proc Natl Acad Sci U S A.
95
1998
14769
14774
95
McAllister
KA
Grogg
KM
Johnson
DW
et al
Endoglin, a TGF-β binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1.
Nat Genet.
8
1994
345
351
96
Johnson
DW
Berg
JN
Baldwin
MA
et al
Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2.
Nat Genet.
13
1996
189
195
97
Angles-Cano
E
Structural basis for the pathophysiology of lipoprotein (a) in the athero-thrombotic process.
Braz J Med Biol Res.
30
1997
1271
1280
98
Markowitz
S
Wang
J
Myeroff
L
et al
Inactivation of the type II TGF-β receptor in colon cancer cells with microsatellite instability.
Science.
268
1995
1336
1338
99
Vincent
F
Nagashima
M
Takenoshita
S
et al
Mutation analysis of the transforming growth factor-β type II receptor in human cell lines resistant to growth inhibition by transforming growth factor-β.
Oncogene.
15
1997
117
122
100
Park
K
Kim
SJ
Bang
YJ
et al
Genetic changes in the transforming growth factor β (TGF-β) type II receptor gene in human gastric cancer cells: correlation with sensitivity to growth inhibition by TGF-β.
Proc Natl Acad Sci U S A.
91
1994
8772
8776
101
Yang
HK
Kang
SH
Kim
YS
Won
K
Bang
YJ
Kim
SJ
Truncation of the TGF-β type II receptor gene results in insensitivity to TGF-β in human gastric cancer cells.
Oncogene.
18
1999
2213
2219
102
Kim
IY
Ahn
HJ
Zelner
DJ
et al
Loss of expression of transforming growth factor β type I and type II receptors correlates with tumor grade in human prostate cancer tissues.
Clin Cancer Res.
2
1996
1255
1261
103
Baldwin
RL
Friess
H
Yokoyama
M
et al
Attenuated ALK5 receptor expression in human pancreatic cancer: correlation with resistance to growth inhibition.
Int J Cancer.
67
1996
283
288
104
Lazzereschi
D
Ranieri
A
Mincione
G
Taccogna
S
Nardi
F
Colletta
G
Human malignant thyroid tumors displayed reduced levels of transforming growth factor β receptor type II messenger RNA and protein.
Cancer Res.
57
1997
2071
2076
105
Reisenbichler
H
Chari
RS
Boyer
IJ
Jirtle
RL
Transforming growth factor-β receptors type I, II and III in phenobarbital-promoted rat liver tumors.
Carcinogenesis.
15
1994
2763
2767
106
Bedossa
P
Peltier
E
Terris
B
Franco
D
Poynard
T
Transforming growth factor-β1 (TGF-β1) and TGF-β1 receptors in normal, cirrhotic, and neoplastic human livers.
Hepatology.
21
1995
760
766
107
Kimchi
A
Wang
XF
Weinberg
RA
Cheifetz
S
Massagué
J
Absence of TGF-β receptors and growth inhibitory responses in retinoblastoma cells.
Science.
240
1988
196
199
108
Horie
K
Yamashita
H
Mogi
A
Takenoshita
S
Miyazono
K
Lack of transforming growth factor-β type II receptor expression in human retinoblastoma cells.
J Cell Physiol.
175
1998
305
313
109
Kim
WS
Park
C
Jung
YS
et al
Reduced transforming growth factor-β type II receptor (TGF-βRII) expression in adenocarcinoma of the lung.
Anticancer Res.
19
1999
301
306
110
Wang
XJ
Greenhalgh
DA
Bickenbach
JR
et al
Expression of a dominant-negative type II transforming growth factor β (TGF-β) receptor in the epidermis of transgenic mice blocks TGF-β-mediated growth inhibition.
Proc Natl Acad Sci U S A.
94
1997
2386
2391
111
Amendt
C
Schirmacher
P
Weber
H
Blessing
M
Expression of a dominant negative type II TGF-β receptor in mouse skin results in an increase in carcinoma incidence and an acceleration of carcinoma development.
Oncogene.
17
1998
25
34
112
Bottinger
EP
Jakubczak
JL
Haines
DC
Bagnall
K
Wakefield
LM
Transgenic mice overexpressing a dominant-negative mutant type II transforming growth factor β receptor show enhanced tumorigenesis in the mammary gland and lung in response to the carcinogen 7,12 dimethylbene-α-anthracene.
Cancer Res.
57
1997
5564
5570
113
Bottinger
EP
Jakubczak
JL
Roberts
IS
et al
Expression of a dominant-negative mutant TGF-β type II receptor in transgenic mice reveals essential roles for TGF-β in regulation of growth and differentiation in the exocrine pancreas.
EMBO J.
16
1997
2621
2633
114
Letterio
JJ
Bottinger
EP
TGF-β knockout and dominant-negative receptor transgenic mice.
Miner Electrolyte Metab.
24
1998
161
167
115
Hata
A
Shi
Y
Massagué
J
TGF-β signaling and cancer: structural and functional consequences of mutations in Smads.
Mol Med Today.
4
1998
257
262
116
Hata
A
Lo
RS
Wotton
D
Lagna
G
Massagué
J
Mutations increasing autoinhibition inactivate tumour suppressors Smad2 and Smad4.
Nature.
388
1997
82
87
117
Takagi
Y
Koumura
H
Futamura
M
et al
Somatic alterations of the SMAD-2 gene in human colorectal cancers.
Br J Cancer.
78
1998
1152
1155
118
Miyaki
M
Iijima
T
Konishi
M
et al
Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis.
Oncogene.
18
1999
3098
3103
119
Kleeff
J
Maruyama
H
Friess
H
Buchler
MW
Falb
D
Korc
M
Smad6 suppresses TGF-β-induced growth inhibition in COLO-357 pancreatic cancer cells and is overexpressed in pancreatic cancer.
Biochem Biophys Res Commun.
255
1999
268
273
120
Bottinger
EP
Letterio
JJ
Roberts
AB
Biology of TGF-β knockout and transgenic mouse models.
Kidney Int.
51
1997
1355
1360
121
Jansen
R
Damia
G
Usui
N
et al
Effects of recombinant transforming growth factor-β1 on hematologic recovery after treatment of mice with 5-fluorouracil.
J Immunol.
147
1991
3342
3347
122
Grzegorzewski
K
Ruscetti
FW
Usui
N
et al
Recombinant transforming growth factor β1 and β2 protect mice from acutely lethal doses of 5-fluorouracil and doxorubicin.
J Exp Med.
180
1994
1047
1057
123
Goey
H
Keller
JR
Back
T
Longo
DL
Ruscetti
FW
Wiltrout
RH
Inhibition of early murine hemopoietic progenitor cell proliferation after in vivo locoregional administration of transforming growth factor-β1.
J Immunol.
143
1989
877
880
124
Carlino
JA
Higley
HR
Creson
JR
Avis
PD
Ogawa
Y
Ellingsworth
LR
Transforming growth factor-β1 systemically modulates granuloid, erythroid, lymphoid, and thrombocytic cells in mice.
Exp Hematol.
20
1992
943
950
125
Miller
KL
Carlino
JA
Ogawa
Y
Avis
PD
Carroll
KG
Alteration in erythropoiesis in TGF-β-treated mice.
Exp Hematol.
20
1992
951
956
126
Hestdal
K
Jacobsen
SE
Ruscetti
FW
Longo
DL
Boone
TC
Keller
JR
Increased granulopoiesis after sequential administration of transforming growth factor-β1 and granulocyte-macrophage colony-stimulating factor.
Exp Hematol.
21
1993
799
805
127
Chuncharunee
S
Carter
CD
Studtmann
KE
Caro
J
Coffey
RJ
Dessypris
EN
Chronic administration of transforming growth factor-β suppresses erythropoietin-dependent erythropoiesis and induces tumour necrosis factor in vivo.
Br J Haematol.
84
1993
374
380
128
Dickson
MC
Martin
JS
Cousins
FM
Kulkarni
AB
Karlsson
S
Akhurst
RJ
Defective haematopoiesis and vasculogenesis in transforming growth factor-β1 knockout mice.
Development.
121
1995
1845
1854
129
Borkowski
TA
Letterio
JJ
Mackall
CL
et al
A role for TGF-β1 in Langerhans cell biology. Further characterization of the epidermal Langerhans cell defect in TGF-β1 null mice.
J Clin Invest.
100
1997
575
581
130
Williams
AO
Knapton
AD
Geiser
A
Letterio
JJ
Roberts
AB
The liver in transforming growth factor β1 (TGF-β1) null mutant mice.
Ultrastruct Pathol.
20
1996
477
490
131
Geiser
AG
Zeng
QQ
Sato
M
Helvering
LM
Hirano
T
Turner
CH
Decreased bone mass and bone elasticity in mice lacking the transforming growth factor-β1 gene.
Bone.
23
1998
87
93
132
Christ
M
McCartney-Francis
NL
Kulkarni
AB
et al
Immune dysregulation in TGF-β1-deficient mice.
J Immunol.
153
1994
1936
1946
133
Yaswen
L
Kulkarni
AB
Fredrickson
T
et al
Autoimmune manifestations in the transforming growth factor-β1 knockout mouse.
Blood.
87
1996
1439
1445
134
Oshima
M
Oshima
H
Taketo
MM
TGF-β receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis.
Dev Biol.
179
1996
297
302
135
Li
DY
Sorensen
LK
Brooke
BS
et al
Defective angiogenesis in mice lacking endoglin.
Science.
284
1999
1534
1537
136
Bourdeau
A
Dumont
DJ
Letarte
M
A murine model for hereditary hemorrhagic telangiectasia.
J Clin Invest.
104
1999
1343
1351
137
Kaartinen
V
Voncken
JW
Shuler
C
et al
Abnormal lung development and cleft palate in mice lacking TGF-β3 indicates defects of epithelial-mesenchymal interaction.
Nat Genet.
11
1995
415
421
138
Sanford
LP
Ormsby
I
Gittenberger-de Groot
AC
et al
TGF-β2 knockout mice have multiple developmental defects that are non-overlapping with other TGF-β knockout phenotypes.
Development.
124
1997
2659
2670
139
Yang
X
Letterio
JJ
Lechleider
RJ
et al
Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-β.
EMBO J.
18
1999
1280
1291
140
Takaku
K
Oshima
M
Miyoshi
H
Matsui
M
Seldin
MF
Taketo
MM
Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes.
Cell.
92
1998
645
656
141
Chang
H
Huylebroeck
D
Verschueren
K
Guo
Q
Matzuk
MM
Zwijsen
A
Smad5 knockout mice die at mid-gestation due to multiple embryonic and extraembryonic defects.
Development.
126
1999
1631
1642
142
Till
JE
McCulloch
EA
Direct measurement of the radiation sensitivity of normal mouse bone marrow cells.
Radiat Res.
14
1961
213
222
143
Berardi
AC
Wang
A
Levine
JD
Lopez
P
Scadden
DT
Functional isolation and characterization of human hematopoietic stem cells.
Science.
267
1995
104
108
144
Ohta
M
Greenberger
JS
Anklesaria
P
Bassols
A
Massagué
J
Two forms of transforming growth factor-β distinguished by multipotential haematopoietic progenitor cells.
Nature.
329
1987
539
541
145
Keller
JR
Mantel
C
Sing
GK
Ellingsworth
LR
Ruscetti
SK
Ruscetti
FW
Transforming growth factor-β1 selectively regulates early murine hematopoietic progenitors and inhibits the growth of IL-3-dependent myeloid leukemia cell lines.
J Exp Med.
168
1988
737
750
146
Keller
JR
McNiece
IK
Sill
KT
et al
Transforming growth factor β directly regulates primitive murine hematopoietic cell proliferation.
Blood.
75
1990
596
602
147
Ottmann
OG
Pelus
LM
Differential proliferative effects of transforming growth factor-β on human hematopoietic progenitor cells.
J Immunol.
140
1988
2661
2665
148
Sing
GK
Keller
JR
Ellingsworth
LR
Ruscetti
FW
Transforming growth factor β selectively inhibits normal and leukemic human bone marrow cell growth in vitro.
Blood.
72
1988
1504
1511
149
Jacobsen
SE
Keller
JR
Ruscetti
FW
Kondaiah
P
Roberts
AB
Falk
LA
Bidirectional effects of transforming growth factor-β (TGF-β) on colony-stimulating factor-induced human myelopoiesis in vitro: differential effects of distinct TGF-β isoforms.
Blood.
78
1991
2239
2247
150
Hatzfeld
J
Li
ML
Brown
EL
et al
Release of early human hematopoietic progenitors from quiescence by antisense transforming growth factor β1 or Rb oligonucleotides.
J Exp Med.
174
1991
925
929
151
Ploemacher
RE
van Soest
PL
Boudewijn
A
Autocrine transforming growth factor β1 blocks colony formation and progenitor cell generation by hemopoietic stem cells stimulated with steel factor.
Stem Cells.
11
1993
336
347
152
Li
ML
Cardoso
AA
Sansilvestri
P
et al
Additive effects of steel factor and antisense TGF-β1 oligonucleotide on CD34+ hematopoietic progenitor cells.
Leukemia.
8
1994
441
445
153
Lu
L
Xiao
M
Grigsby
S
et al
Comparative effects of suppressive cytokines on isolated single CD34+++ stem/progenitor cells from human bone marrow and umbilical cord blood plated with and without serum.
Exp Hematol.
21
1993
1442
1446
154
Fortunel
N
Hatzfeld
J
Aoustin
L
et al
Specific dose-response effects of TGF-β1 on developmentally distinct hematopoietic stem/progenitor cells from human umbilical cord blood.
Hematol J.
1
2000
126
134
155
Cardoso
AA
Li
ML
Batard
P
et al
Release from quiescence of CD34+CD38− human umbilical cord blood cells reveals their potentiality to engraft adults.
Proc Natl Acad Sci U S A.
90
1993
8707
8711
156
van Ranst
PCF
Snoeck
HW
Lardon
F
et al
TGF-β and MIP-1α exert their main inhibitory activity on very primitive CD34++CD38− cells but show opposite effects on more mature CD34+CD38+ human hematopoietic progenitors.
Exp Hematol.
24
1996
1509
1515
157
Junker
U
Knoefel
B
Nuske
K
et al
Transforming growth factor-β1 is significantly elevated in plasma of patients suffering from renal cell carcinoma.
Cytokine.
8
1996
794
798
158
Sargiacomo
M
Valtieri
M
Gabbianelli
M
et al
Pure human hematopoietic progenitors: direct inhibitory effect of transforming growth factors-β1 and -β2.
Ann N Y Acad Sci.
628
1991
84
91
159
Strife
A
Lambek
C
Perez
A
et al
The effects of transforming growth factor β3 on the growth of highly enriched hematopoietic progenitor cells derived from normal human bone marrow and peripheral blood.
Cancer Res.
51
1991
4828
4836
160
Dexter
TM
Haemopoiesis in long-term bone marrow cultures. A review.
Acta Haematol.
62
1979
299
305
161
Eaves
CJ
Cashman
JD
Sutherland
HJ
et al
Molecular analysis of primitive hematopoietic cell proliferation control mechanisms.
Ann N Y Acad Sci.
628
1991
298
306
162
Eaves
CJ
Cashman
JD
Kay
RJ
et al
Mechanisms that regulate the cell cycle status of very primitive hematopoietic cells in long-term human marrow cultures, II: Analysis of positive and negative regulators produced by stromal cells within the adherent layer.
Blood.
78
1991
110
117
163
Moore
SC
Theus
SA
Barnett
JB
Soderberg
LS
Bone marrow natural suppressor cells inhibit the growth of myeloid progenitor cells and the synthesis of colony-stimulating factors.
Exp Hematol.
20
1992
1178
1183
164
Tang
J
Nuccie
BL
Ritterman
I
Liesveld
JL
Abboud
CN
Ryan
DH
TGF-β down-regulates stromal IL-7 secretion and inhibits proliferation of human B cell precursors.
J Immunol.
159
1997
117
125
165
Gautam
SC
Noth
CJ
Janakiraman
N
Pindolia
KR
Chapman
RA
Induction of chemokine mRNA in bone marrow stromal cells: modulation by TGF-β1 and IL-4.
Exp Hematol.
23
1995
482
491
166
Sakamaki
S
Hirayama
Y
Matsunaga
T
et al
Transforming growth factor-β1 (TGF-β1) induces thrombopoietin from bone marrow stromal cells, which stimulates the expression of TGF-β receptor on megakaryocytes and, in turn, renders them susceptible to suppression by TGF-β itself with high specificity.
Blood.
94
1999
1961
1970
167
Nemunaitis
J
Tompkins
CK
Andrews
DF
Singer
JW
Transforming growth factor-β expression in human marrow stromal cells.
Eur J Haematol.
46
1991
140
145
168
Cashman
JD
Eaves
AC
Raines
EW
Ross
R
Eaves
CJ
Mechanisms that regulate the cell cycle status of very primitive hematopoietic cells in long-term human marrow cultures, I: Stimulatory role of a variety of mesenchymal cell activators and inhibitory role of TGF-β.
Blood.
75
1990
96
101
169
Dexter
TM
Allen
TD
Latjha
LG
Conditions controlling the proliferation of haematopoietic stem cells in vitro.
J Cell Physiol.
91
1977
335
344
170
Waegell
WO
Higley
HR
Kincade
PW
Dasch
JR
Growth acceleration and stem cell expansion in Dexter-type cultures by neutralization of TGF-β.
Exp Hematol.
22
1994
1051
1057
171
Cashman
JD
Clark-Lewis
I
Eaves
AC
Eaves
CJ
Differentiation stage-specific regulation of primitive human hematopoietic progenitor cycling by exogenous and endogenous inhibitors in an in vivo model.
Blood.
94
1999
3722
3729
172
Weekx
SF
Plum
J
Van Cauwelaert
P
et al
Developmentally regulated responsiveness to transforming growth factor-β is correlated with functional differences between human adult and fetal primitive hematopoietic progenitor cells.
Leukemia.
13
1999
1266
1272
173
Dubois
CM
Ruscetti
FW
Palaszynski
EW
Falk
LA
Oppenheim
JJ
Keller
JR
Transforming growth factor β is a potent inhibitor of interleukin 1 (IL-1) receptor expression: proposed mechanism of inhibition of IL-1 action.
J Exp Med.
172
1990
737
744
174
Jacobsen
SE
Ruscetti
FW
Dubois
CM
Lee
J
Boone
TC
Keller
JR
Transforming growth factor-β trans-modulates the expression of colony stimulating factor receptors on murine hematopoietic progenitor cell lines.
Blood.
77
1991
1706
1716
175
Keller
JR
Jacobsen
SE
Sill
KT
Ellingsworth
LR
Ruscetti
FW
Stimulation of granulopoiesis by transforming growth factor: synergy with granulocyte/macrophage-colony-stimulating factor.
Proc Natl Acad Sci U S A.
88
1991
7190
7194
176
Jacobsen
SE
Ruscetti
FW
Roberts
AB
Keller
JR
TGF-β is a bidirectional modulator of cytokine receptor expression on murine bone marrow cells. Differential effects of TGF-β1 and TGF-β3.
J Immunol.
151
1993
4534
4544
177
Dubois
CM
Ruscetti
FW
Stankova
J
Keller
JR
Transforming growth factor-β regulates c-kit message stability and cell-surface protein expression in hematopoietic progenitors.
Blood.
83
1994
3138
3145
178
Heinrich
MC
Dooley
DC
Keeble
WW
Transforming growth factor-β1 inhibits expression of the gene products for steel factor and its receptor (c-kit).
Blood.
85
1995
1769
1780
179
Sansilvestri
P
Cardoso
AA
Batard
P
et al
Early CD34high cells can be separated into KIThigh cells in which transforming growth factor-β (TGF-β) downmodulates c-kit and KITlow cells in which anti-TGF-β upmodulates c-kit.
Blood.
86
1995
1729
1735
180
McNiece
IK
Bertoncello
I
Keller
JR
Ruscetti
FW
Hartley
CA
Zsebo
KM
Transforming growth factor-β inhibits the action of stem cell factor on mouse and human hematopoietic progenitors.
Int J Cell Cloning.
10
1992
80
86
181
Fortunel
N
Batard
P
Hatzfeld
A
et al
High proliferative potential-quiescent cells: a working model to study primitive quiescent hematopoietic cells.
J Cell Sci.
111
1998
1867
1875
182
Jacobsen
SEW
Veiby
OP
Myklebust
J
Okkenhaug
C
Lyman
SD
Ability of flt3 ligand to stimulate the in vitro growth of primitive murine hematopoietic progenitors is potently and directly inhibited by transforming growth factor-β and tumor necrosis factor-α.
Blood.
87
1996
5016
5026
183
Ohishi
K
Katayama
N
Itoh
R
et al
Accelerated cell-cycling of hematopoietic progenitors by the flt3 ligand that is modulated by transforming growth factor-β.
Blood.
87
1996
1718
1727
184
Ramsfjell
V
Borge
OJ
Cui
L
Jacobsen
SE
Thrombopoietin directly and potently stimulates multilineage growth and progenitor cell expansion from primitive (CD34+CD38−) human bone marrow progenitor cells: distinct and key interactions with the ligands for c-kit and flt3, and inhibitory effects of TGF-β and TNF-α.
J Immunol.
158
1997
5169
5177
185
Zanjani
ED
Pallavicini
MG
Ascensao
JL
et al
Engraftment and long-term expression of human fetal hemopoietic stem cells in sheep following transplantation in utero.
J Clin Invest.
89
1992
1178
1188
186
Kawashima
I
Zanjani
ED
Almaida-Porada
G
Flake
AW
Zeng
H
Ogawa
M
CD34+ human marrow cells that express low levels of Kit protein are enriched for long-term marrow-engrafting cells.
Blood.
87
1996
4136
4142
187
Lotem
J
Sachs
L
Hematopoietic cytokines inhibit apoptosis induced by transforming growth factor β1 and cancer chemotherapy compounds in myeloid leukemic cells.
Blood.
80
1992
1750
1757
188
Saltzman
A
Munro
R
Searfoss
G
Franks
C
Jaye
M
Ivashchenko
Y
Transforming growth factor-β-mediated apoptosis in the Ramos B-lymphoma cell line is accompanied by caspase activation and Bcl-XL downregulation.
Exp Cell Res.
242
1998
244
254
189
Schrantz
N
Blanchard
DA
Auffredou
MT
Sharma
S
Leca
G
Vazquez
A
Role of caspases and possible involvement of retinoblastoma protein during TGFβ-mediated apoptosis of human B lymphocytes.
Oncogene.
18
1999
3511
3519
190
Taetle
R
Payne
C
Dos Santos
B
Russell
M
Segarini
P
Effects of transforming growth factor β1 on growth and apoptosis of human acute myelogenous leukemia cells.
Cancer Res.
53
1993
3386
3393
191
Buske
C
Becker
D
Feuring-Buske
M
et al
TGF-β inhibits growth and induces apoptosis in leukemic B cell precursors.
Leukemia.
11
1997
386
392
192
Kuo
ML
Chen
CW
Jee
SH
Chuang
SE
Cheng
AL
Transforming growth factor-β1 attenuates ceramide-induced CPP32/Yama activation and apoptosis in human leukaemic HL-60 cells.
Biochem J.
327
1997
663
667
193
Saeki
K
Yuo
A
Kato
M
Miyazono
K
Yazaki
Y
Takaku
F
Cell density-dependent apoptosis in HL-60 cells, which is mediated by an unknown soluble factor, is inhibited by transforming growth factor-β1 and overexpression of Bcl-2.
J Biol Chem.
272
1997
20003
20010
194
Alam
R
Forsythe
P
Stafford
S
Fukuda
Y
Transforming growth factor β abrogates the effects of hematopoietins on eosinophils and induces their apoptosis.
J Exp Med.
179
1994
1041
1045
195
Schiott
A
Sjogren
HO
Lindvall
M
Three isoforms of transforming growth factor-β co-stimulate rat T cells and inhibit lymphocyte apoptosis.
Scand J Immunol.
48
1998
371
378
196
Genestier
L
Kasibhatla
S
Brunner
T
Green
DR
Transforming growth factor-β1 inhibits Fas ligand expression and subsequent activation-induced cell death in T cells via downregulation of c-Myc.
J Exp Med.
189
1999
231
239
197
Riedl
E
Strobl
H
Majdic
O
Knapp
W
TGF-β1 promotes in vitro generation of dendritic cells by protecting progenitor cells from apoptosis.
J Immunol.
158
1997
1591
1597
198
Sitnicka
E
Ruscetti
FW
Priestley
GV
Wolf
NS
Bartelmez
SH
Transforming growth factor β1 directly and reversibly inhibits the initial cell division of long-term repopulating stem cells.
Blood.
88
1996
82
88
199
Garbe
A
Spyridonidis
A
Mobest
D
Schmoor
C
Mertelsmann
R
Hensler
R
Transforming growth factor-β1 delays formation of granulocyte-macrophage colony-forming cells, but spares more primitive progenitors during ex vivo expansion of CD34+ haemopoietic progenitor cells.
Br J Haematol.
99
1997
951
958
200
Dybedal
I
Guan
F
Borge
OJ
et al
Transforming growth factor-β1 abrogates Fas-induced growth suppression and apoptosis of murine bone marrow progenitor cells.
Blood.
90
1997
3395
3403
201
Jacobsen
FW
Stokke
T
Jacobsen
SE
Transforming growth factor-β potently inhibits the viability-promoting activity of stem cell factor and other cytokines and induces apoptosis of primitive murine hematopoietic progenitor cells.
Blood.
86
1995
2957
2966
202
Veiby
OP
Jacobsen
FW
Cui
L
Lyman
SD
Jacobsen
SE
The flt3 ligand promotes the survival of primitive hemopoietic progenitor cells with myeloid as well as B lymphoid potential. Suppression of apoptosis and counteraction by TNF-α and TGF-β.
J Immunol.
157
1996
2953
2960
203
Batard
P
Monier
MN
Fortunel
N
et al
TGF-β1 maintains hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation.
J Cell Sci.
113
2000
383
390
204
Pierelli
L
Marone
M
Bonanno
G
et al
Modulation of bcl-2 and p27 in human primitive proliferating hematopoietic progenitors by autocrine TGF-β1 is a cell cycle-independent effect and influences their hematopoietic potential.
Blood.
95
2000
3001
3009
205
Del Rizzo
DF
Eskinazi
D
Axelrad
AA
Interleukin 3 opposes the action of negative regulatory protein (NRP) and transforming growth factor-β (TGF-β) in their inhibition of DNA synthesis of the erythroid stem cell BFU-E.
Exp Hematol.
18
1990
138
142
206
Gandrillon
O
Schmidt
U
Beug
H
Samarut
J
TGF-β cooperates with TGF-α to induce the self-renewal of normal erythrocytic progenitors: evidence for an autocrine mechanism.
EMBO J.
18
1999
2764
2781
207
Chen
LL
Dean
A
Jenkinson
T
Mendelsohn
J
Effect of transforming growth factor-β1 on proliferation and induction of hemoglobin accumulation in K-562 cells.
Blood.
74
1989
2368
2375
208
Zermati
Y
Varet
B
Hermine
O
TGF-β1 drives and accelerates erythroid differentiation in the Epo-dependent UT-7 cell line even in the absence of erythropoietin.
Exp Hematol.
28
2000
256
266
209
Krystal
G
Lam
V
Dragowska
W
et al
Transforming growth factor-β1 is an inducer of erythroid differentiation.
J Exp Med.
180
1994
851
860
210
Dai
CH
Krantz
CB
Zsebo
KM
Human burst-forming units-erythroid need direct interaction with stem cell factor for further development.
Blood.
78
1991
2493
2497
211
Lu
L
Heinrich
MC
Wang
LS
et al
Retroviral-mediated gene transduction of c-kit into single hematopoietic progenitor cells from cord blood enhances erythroid colony formation and decreases sensitivity to inhibition by tumor necrosis factor-α and transforming growth factor-β1.
Blood.
94
1999
2319
2332
212
Ogawa
K
Tashima
M
Toi
T
et al
Inhibition of erythroid differentiation by stem cell factor in K-562 cells expressing the c-kit gene.
Exp Hematol.
22
1994
45
51
213
Muta
K
Krantz
SB
Bondurant
MC
Dai
CH
Stem cell factor retards differentiation of normal human erythroid progenitor cells while stimulating proliferation.
Blood.
86
1995
572
580
214
Dybedal
I
Jacobsen
SE
Transforming growth factor-β (TGF-β), a potent inhibitor of erythropoiesis: neutralizing TGF-β antibodies show erythropoietin as a potent stimulator of murine burst-forming unit erythroid colony formation in the absence of a burst-promoting activity.
Blood.
86
1995
949
957
215
Leveque
E
Nagel
MD
Haye
B
Effect of the interaction between transforming growth factor β and erythropoietin on the proliferation of normal erythroid progenitors and leukemic UT-7 cells: action of transforming growth factor β on the erythropoietin receptor.
Hematol Oncol.
14
1996
137
146
216
Richon
VM
Rifkind
RA
Marks
PA
Expression and phosphorylation of the retinoblastoma protein during induced differentiation of murine erythroleukemia cells.
Cell Growth Differ.
3
1992
413
420
217
Sehy
DW
Shao
LE
Yu
AL
Tsai
WM
Yu
J
Activin A-induced differentiation in K562 cells is associated with a transient hypophosphorylation of RB protein and the concomitant block of cell cycle at G1 phase.
J Cell Biochem.
50
1992
255
265
218
Zhuo
S
Fan
S
Huang
S
Kaufman
S
Study of the role of retinoblastoma protein in terminal differentiation of murine erythroleukemia cells.
Proc Natl Acad Sci U S A.
92
1995
4234
4238
219
Condorelli
GL
Testa
U
Valtieri
M
et al
Modulation of retinoblastoma gene in normal adult hematopoiesis: peak expression and functional role in advanced erythroid differentiation.
Proc Natl Acad Sci U S A.
92
1995
4808
4812
220
Jacks
T
Fazeli
A
Schmitt
EM
Bronson
RT
Goodell
MA
Weinberg
RA
Effects of an Rb mutation in the mouse.
Nature.
359
1992
295
300
221
Lee
EY
Chang
CY
Hu
N
et al
Mice deficient for Rb are nonviable and show defects in neurogenesis and haematopoiesis.
Nature.
359
1992
288
294
222
Hu
N
Gulley
ML
Kung
JT
Lee
EY
Retinoblastoma gene deficiency has mitogenic but not tumorigenic effects on erythropoiesis.
Cancer Res.
57
1997
4123
4129
223
Celada
A
Maki
RA
Transforming growth factor-β enhances the M-CSF and GM-CSF-stimulated proliferation of macrophages.
J Immunol.
148
1992
1102
1105
224
Fan
K
Ruan
Q
Sensenbrenner
L
Chen
B
Transforming growth factor-β1 bifunctionally regulates murine macrophage proliferation.
Blood.
79
1992
1679
1685
225
Turley
JM
Falk
LA
Ruscetti
FW
et al
Transforming growth factor β1 functions in monocytic differentiation of hematopoietic cells through autocrine and paracrine mechanisms.
Cell Growth Differ.
7
1996
1535
1544
226
Bruno
E
Miller
ME
Hoffman
R
Interacting cytokines regulate in vitro human megakaryocytopoiesis.
Blood.
73
1989
671
677
227
Jackson
H
Williams
N
Westcott
KR
Green
R
Differential effects of transforming growth factor-β1 on distinct developmental stages of murine megakaryocytopoiesis.
J Cell Physiol.
161
1994
312
318
228
Broudy
VC
Lin
NL
Kaushansky
K
Thrombopoietin (c-mpl ligand) acts synergistically with erythropoietin, stem cell factor, and interleukin-11 to enhance murine megakaryocyte colony growth and increases megakaryocyte ploidy in vitro.
Blood.
85
1995
1719
1726
229
Kaushansky
K
Brouty
VC
Lin
N
et al
Thrombopoietin, the Mpl ligand, is essential for full megakaryocyte development.
Proc Natl Acad Sci U S A.
92
1995
3234
3238
230
Young
JC
Bruno
E
Luens
KM
Wu
S
Backer
M
Murray
LJ
Thrombopoietin stimulates megakaryocytopoiesis, myelopoiesis, and expansion of CD34+ progenitor cells from single CD34+Thy-1+Lin− primitive progenitor cells.
Blood.
88
1996
1619
1631
231
Kuter
DJ
Gminski
DM
Rosenberg
RD
Transforming growth factor β inhibits megakaryocyte growth and endomitosis.
Blood.
79
1992
619
626
232
Berthier
R
Valiron
O
Schweitzer
A
Marguerie
G
Serum-free medium allows the optimal growth of human megakaryocyte progenitors compared with human plasma supplemented cultures: role of TGF β.
Stem Cells.
11
1993
120
129
233
Zucker-Franklin
D
Kaushansky
K
Effect of thrombopoietin on the development of megakaryocytes and platelets: an ultrastructure analysis.
Blood.
88
1996
1632
1638
234
Rojnuckarin
P
Drachman
JG
Kaushansky
K
Thrombopoietin-induced activation of the mitogen-activated protein kinase (MAPK) pathway in normal megakaryocytes: role in endomitosis.
Blood.
94
1999
1273
1282
235
Greenberg
SM
Chandrasekhar
C
Golan
DE
Handin
RI
Transforming growth factor β inhibits endomitosis in the Dami human megakaryocytic cell line.
Blood.
76
1990
533
537
236
Cowley
SA
Groopman
JE
Avraham
H
Effects of transforming growth factor β on megakaryocytic cell fusion and endomitosis.
Int J Cell Cloning.
10
1992
223
231
237
Mitjavila
MT
Vinci
G
Villeval
JL
et al
Human platelet α granules contain a nonspecific inhibitor of megakaryocyte colony formation: its relationship to type β transforming growth factor (TGF-β).
J Cell Physiol.
134
1988
93
100
238
Fava
RA
Casey
TT
Wilcox
J
Pelton
RW
Moses
HL
Nanney
LB
Synthesis of transforming growth factor-β1 by megakaryocytes and its localization to megakaryocyte and platelet α-granules.
Blood.
76
1990
1946
1955
239
Wickenhauser
C
Hillienhof
A
Jungheim
K
et al
Detection and quantification of transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) release by normal human megakaryocytes.
Leukemia.
9
1995
310
315
240
Zauli
G
Catani
L
Gugliotta
L
et al
Essential thrombocythemia: impaired regulation of megakaryocyte progenitors.
Int J Cell Cloning.
9
1991
43
56
241
Zauli
G
Visani
G
Catani
L
Vianelli
N
Gugliotta
L
Capitani
S
Reduced responsiveness of bone marrow megakaryocyte progenitors to platelet-derived transforming growth factor β1, produced in normal amounts, in patients with essential thrombocythaemia.
Br J Haematol.
83
1993
14
20
242
Broudy
VC
Lin
NL
Fox
N
Taga
T
Saito
M
Kaushansky
K
Thrombopoietin stimulates colony-forming unit-megakaryocyte proliferation and megakaryocyte maturation independently of cytokines that signal through the gp130 receptor subunit.
Blood.
88
1996
2026
2032
243
Mouthon
MA
Navarro
S
Katz
A
Breton-Gorius
J
Vainchenker
W
c-jun and c-fos are expressed by human megakaryocytes.
Exp Hematol.
20
1992
909
915
244
Panterne
B
Hatzfeld
J
Blanchard
JM
et al
c-fos mRNA constitutive expression by mature human megakaryocytes.
Oncogene.
7
1992
2341
2344
245
Strobl
H
Riedl
E
Scheinecker
C
et al
TGF-β1 promotes in vitro development of dendritic cells from CD34+ hemopoietic progenitors.
J Immunol.
157
1996
1499
1507
246
Zhang
Y
Zhang
YY
Ogata
M
et al
Transforming growth factor-β1 polarizes murine hematopoietic progenitor cells to generate Langerhans cell-like dendritic cells through monocyte/macrophage differentiation pathway.
Blood.
93
1999
1208
1220
247
Geissmann
F
Prost
C
Monnet
JP
Dy
M
Brousse
N
Hermine
O
Transforming growth factor-β1, in the presence of granulocyte/macrophage colony-stimulating factor and interleukin 4, induces differentiation of human peripheral blood monocytes into dendritic Langerhans cells.
J Exp Med.
187
1998
961
966
248
Geissmann
F
Revy
P
Regnault
A
et al
TGF-β1 prevents the noncognate maturation of human dendritic Langerhans cells.
J Immunol.
162
1999
4567
4575
249
Yang
D
Howard
OM
Chen
Q
Oppenheim
JJ
Cutting edge: immature dendritic cells generated from monocytes in the presence of TGF-β1 express functional C-C chemokine receptor 6.
J Immunol.
163
1999
1737
1741
250
Jaksits
S
Kriehuber
E
Charbonnier
AS
Rappersberger
K
Stingl
G
Maurer
D
CD34+ cell-derived CD14+ precursor cells develop into Langerhans cells in a TGF-β1-dependent manner.
J Immunol.
163
1999
4869
4877
251
Strobl
H
Bello-Fernandez
C
Riedl
E
et al
FLT3 ligand in cooperation with transforming growth factor-β1 potentiates in vitro development of Langerhans-type dendritic cells and allows single-cell dendritic cell cluster formation under serum-free conditions.
Blood.
90
1997
1425
1434
252
Miller
DG
Adam
MA
Miller
AD
Gene transfer by retrovirus vector occurs only in cells that are actively replicating at the time of infection.
Mol Cell Biol.
10
1990
4239
4242
253
Nolta
JA
Kohn
DB
Comparison of the effects of growth factors on retroviral vector-mediated gene transfer and the proliferative status of human hematopoietic progenitor cells.
Hum Gene Ther.
1
1990
257
268
254
Hatzfeld
A
Batard
P
Panterne
B
Taieb
F
Hatzfeld
J
Increased stable retroviral gene transfer in early hematopoietic progenitors released from quiescence.
Hum Gene Ther.
7
1996
207
213
255
Dao
MA
Taylor
N
Nolta
JA
Reduction in levels of the cyclin-dependent kinase inhibitor p27(kip-1) coupled with transforming growth factor β neutralization induces cell-cycle entry and increases retroviral transduction of primitive human hematopoietic cells.
Proc Natl Acad Sci U S A.
95
1998
13006
13011
256
Imbert
AM
Bagnis
C
Galindo
R
Chabannon
C
Mannoni
P
A neutralizing anti-TGF-β1 antibody promotes proliferation of CD34+Thy-1+ peripheral blood progenitors and increases the number of transduced progenitors.
Exp Hematol.
26
1998
374
381
257
Yu
J
Soma
T
Hanazono
Y
Dunbar
CE
Abrogation of TGF-β activity during retroviral transduction improves murine hematopoietic progenitor and repopulating cell gene transfer efficiency.
Gene Ther.
5
1998
1265
1271
258
Ducos
K
Panterne
B
Fortunel
N
Hatzfeld
A
Monier
MN
Hatzfeld
J
p21 mRNA is controlled by endogenous transforming growth factor-β1 in quiescent human hematopoietic stem/progenitor cells.
J Cell Physiol.
184
2000
80
85
259
Ando
K
Griffin
JD
Cdk4 integrates growth stimulatory and inhibitory signals during G1 phase of hematopoietic cells.
Oncogene.
10
1995
751
755
260
Bhatia
M
Bonnet
D
Wu
D
et al
Bone morphogenetic proteins regulate the developmental program of human hematopoietic stem cells.
J Exp Med.
189
1999
1139
1148

Author notes

Jacques A. Hatzfeld, Laboratoire de Biologie des Cellules Souches Somatiques Humaines, UPR 1983, Centre National de la Recherche Scientifique, IFC1, 7, rue Guy Môquet, 94800, Villejuif, France; e-mail: hatzfeld@infobiogen.fr.

Sign in via your Institution