TGFβ restores hematopoietic homeostasis after myelosuppressive chemotherapy

Fabienne Brenet, Pouneh Kermani, Roman Spektor, Shahin Rafii, Joseph M Scandura, Fabienne Brenet, Pouneh Kermani, Roman Spektor, Shahin Rafii, Joseph M Scandura

Abstract

Myelosuppression is a life-threatening complication of antineoplastic therapy, but treatment is restricted to a few cytokines with unilineage hematopoietic activity. Although hematopoietic stem cells (HSCs) are predominantly quiescent during homeostasis, they are rapidly recruited into cell cycle by stresses, including myelosuppressive chemotherapy. Factors that induce HSCs to proliferate during stress have been characterized, but it is not known how HSC quiescence is then reestablished. In this study, we show that TGFβ signaling is transiently activated in hematopoietic stem and progenitor cells (HSPCs) during hematopoietic regeneration. Blockade of TGFβ signaling after chemotherapy accelerates hematopoietic reconstitution and delays the return of cycling HSCs to quiescence. In contrast, TGFβ blockade during homeostasis fails to induce cycling of HSPCs. We identified the cyclin-dependent kinase inhibitor Cdkn1c (p57) as a key downstream mediator of TGFβ during regeneration because the recovery of chimeric mice, incapable of expressing p57 in HSPCs, phenocopies blockade of TGFβ signaling after chemotherapy. This study demonstrates that context-dependent activation of TGFβ signaling is central to an unrecognized counterregulatory mechanism that promotes homeostasis once hematopoiesis has sufficiently recovered from myelosuppressive chemotherapy. These results open the door to new, potentially superior, approaches to promote multilineage hematopoietic recovery by blocking the TGFβ signaling that dampens regeneration.

Figures

Figure 1.
Figure 1.
TGFβ signaling is activated during BM recovery from chemotherapy. WT mice (n = 16) were treated with a single dose of 5FU (250 mg/kg, i.p.) on day 0. (A) IHC staining for pSmad2 was performed on BM sections collected before and after chemotherapy. Sections were counterstained with the nuclear stain, methyl green, to assess cellularity (bar, 100 µm). (B) ELISA measurement of active TGFβ in the BM (gray squares), quantification of pSmad2-positive cells per high-power (HP) IHC field (green circles) and BM cellularity (black diamonds) were assessed before (D0) and at the indicated times after chemotherapy. (C; top) Representative immunoblot of pSmad2 (∼55-kD) in Lin− BM and spleen cells during homeostasis (D0) and during recovery from chemotherapy (D15). (bottom) Replicate blots were quantified using ImageJ and pSmad2 was normalized to the α-tubulin (∼50 kD) loading control (n = 3). (D) FACS-purified LKS+ cells were stained for pSmad2 (green) and DAPI (blue) on day 0 and 15 (bar, 50 µm). (E) pSmad2 fluorescence intensity (FI) was quantified using ImageJ software. The day 15/day 0 FI ratio (left) and the percentage of pSmad2+ LKS+ cells are shown at day 0 and 15 (right). All experiments were performed at least twice, usually three to five times. All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 2.
Figure 2.
Blockade of TGFβ during recovery from chemotherapy promotes hematopoietic regeneration. (A) Cohorts of mice were treated with 5FU on day 0, and then with no additional agent (black diamonds, 5FU), the TGFβ-neutralizing antibody, 1D11 (gray squares, 5FU-I), or a nontargeted isotype control antibody, 13C4 (white circles, 5FU-C; n = 6) on day 5, 7, and 9 (arrows). (B; top) Representative immunoblot of pSmad2 in Lin− BM during homeostasis (D0) and during recovery from chemotherapy (D15) with 1D11 (I) or 13C4 (C) antibody treatment. (bottom) Replicate blots were quantified using ImageJ and pSmad2 (∼55 kD) was normalized to the Smad2 band (top) of the Smad 2/3 (∼55/50 kD) loading control (n = 3). (C–F) Recovery of WBCs (C), platelets (D), and RBCs (E) is shown for treated mice (n = 6). (F) Cohorts of untreated, homeostatic mice (n = 10) were administered either the TGFβ-neutralizing antibody 1D11 (I; 10 mg/kg) or 13C4 control (C; 10 mg/kg) on days −3, −1 before analysis on day 0. Total BM cellularity and Lin-depleted cell count, WBCs, RBCs, and platelets (PLTs) were assessed after treatment. All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 3.
Figure 3.
TGFβ limits HSPC proliferation after chemotherapy. (A–D) BM cell counts before (D0) and after (D15) treatment with 5FU and the TGFβ-neutralizing antibody, 1D11 (D15-I), or the control antibody (D15-C) were assessed by flow cytometry. Results were normalized per leg (femur + tibia) as indicated. Expansion of LKS+ (Lin−cKit+Sca1+) cells (A), LKS+Flk2−CD34− (LT), LKS+Flk2−CD34+ (ST), and LKS−FCRγII/IIIdimCD34+ CMPs and LKS + SLAM cells (D, right) is shown for untreated mice (black bars, D0) and mice treated with 5FU, and then 1D11 (gray bars, 5FU-I) or the control antibody (white bars, 5FU-C) during hematopoietic regeneration (n = 5). (D, left) Representative flow cytometry of LKS + SLAM (LKS+CD48−CD150+) on day 15 after treatment with 5FU and either 1D11 (D15-I) or the control antibody (D15-C) is shown. (E) CFC assay for committed HPCs in D15-I BM and D15-C BM (n = 3). (F) Schematic of competitive repopulation analysis. EGFP or CD45.1-marked D15-C BMMC were mixed 1:1 with reciprocally marked D15-I BMMCs, and then transplanted into lethally irradiated recipient mice (n = 10). (G) Competitive repopulation by D15-C and D15-I cells of multilineage peripheral blood was analyzed by flow cytometry at the indicated times after transplantation (n = 10). All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 4.
Figure 4.
Blockade of TGFβ during recovery from myelotoxic stress prolongs HSPC cycling. (A) Flow cytometry gating strategy for cell cycle analysis of LKS+ (Lin−cKit+Sca1+) and LKS + SLAM (Lin−cKit+Sca1+CD48−CD150+) cells is shown (top images show frequency in total BM; bottom images show percentage of LKS + SLAM gated cells). (B) Mice were treated with 5FU, and then with the TGFβ-neutralizing antibody, 1D11 (5FU-I), or the isotype control antibody 13C4 (5FU-C) on day 5, 7, and 9. The percentage of quiescent LKS+ cells is shown before treatment (D0) and at various times after treatment with 5FU and 1D11 (5FU-I, gray bars) or the control antibody (5FU-C, white bars; n = 5/group). (C) Cell cycle status of BM LKS + SLAM HSCs is shown before (D0) and on day 15 after treatment with 5FU and 1D11 (5FU-I) or the control antibody (n = 5). (D) Cohorts of untreated, homeostatic mice (n = 10) were administered either the TGFβ-neutralizing antibody 1D11 (I; 10 mg/kg) or control antibody 13C4 (C; 10 mg/kg) on days −3 and −1 before analysis on day 0. Bivariate cell cycle status of BM LKS + SLAM HSC on day 0 is shown. All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 5.
Figure 5.
TGFβ is required for p57 up-regulation during recovery from myelosuppressive chemotherapy. (A) The expression of CDKIs in Lin− BM cells of C57BL/6J mice before (D0) or after (D15) administration of 5FU (250 mg/kg, i.p.) was assessed by qRT-PCR. Expression of p57, p21, p18, and p27 mRNA was normalized to Hprt1 at each time point (n = 3). (B) Wild-type mice were killed before (D0) and at the indicated times after a single dose of 5FU chemotherapy. Femurs were stained by IHC for p57 and nuclei were counterstained with Methyl Green. p57 staining was seen in both megakaryocytes (Mk, small arrows) and small mononuclear cells (Mn, large arrow heads). Expression of p57 (reddish-brown) was maximally up-regulated 15 d after 5FU treatment (bar, 100 µm). (C) Cohorts of mice were treated with 5FU on day 0, and then with the TGFβ-neutralizing antibody 1D11 or a nontargeted control antibody 13C4 on day 5, 7, and 9. Immunoblots are shown for p57 (∼51 kD), pSmad2 (∼55 kD), and p27 (∼27 kD), in Lin− BM cells. (D) Replicate blots were quantified using ImageJ and expression normalized to β-actin (∼45 kD; n = 3). (E) LKS+ cells were FACS purified from mice before (D0), and on day 15 after 5FU chemotherapy, followed by either 1D11 (D15-I) or 13C4 (D15-C), as described previously. Purified cells were stained for p57 (red), pSmad2 (green), and for DNA using DAPI (blue). Images were acquired using a Zeiss 710 laser scanning confocal microscope and (F) fluorescence intensity (FI) was quantified using ImageJ software (bar, 10 µm). All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 6.
Figure 6.
p57-KO HSPCs recover more robustly after myelosuppressive chemotherapy. (A) Table comparing hematologic parameters for p57-WT and p57-KO hematopoiesis. FLMCs were harvested from littermates and transplanted into lethally irradiated (9 Gr) C57BL/6 recipients to assess adult hematopoiesis. Once steady-state hematopoiesis was reached (≥12 wk), BM LKS+ cells were enumerated by flow cytometry and normalized to cells per leg (n = 5 for each genotype). Blood cell counts (n = 20) are shown for WT and KO-transplanted mice after they reached homeostasis (>12 wk). BM, total BM cellularity, WBC, white blood cells; RBC, red blood cells; HGB, hemoglobin; HCT, hematocrit; PLT, platelets; MCV, Mean Corpuscular Volume. (B) CFC assay for committed HPCs is shown for chimeric mice with p57-WT (black bars) and p57-KO (white bars) BM cells. Total CFCs, CFU-GM (CFU-granulocyte, macrophage), CFU-GEMM (CFU-granulocyte, erythrocyte, macrophage, megakaryocyte), and BFU-E (Burst forming unit-erythroid) are presented in the graph (n = 3 in triplicate for each genotype). (C) Littermate FLMCs were isolated from p57-WT or p57-KO embryos and transplanted into lethally irradiated C57BL/6J recipient mice (n = 10). Once homeostasis was reached (∼10 wk), recipient mice were treated with a single myelosuppressive dose of 5FU (150 mg/kg, i.p) as indicated. Peripheral blood WBCs (D), RBCs (E), platelets (F), and BM cellularity (G) are shown for chimeric mice with p57-WT (black circles) and p57-KO (white squares) hematopoiesis. All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 7.
Figure 7.
p57-KO hematopoiesis phenocopies inhibition of TGFβ signaling during BM regeneration. (A) Representative multidimensional flow cytometry of p57-WT and p57-KO BM LKS+ cells is shown before treatment (D0) and day 15 after treatment with 5FU (frequency in total BM). (B and C) Flow cytometric enumeration of LKS+ (Lin−cKit+Sca1+) cells, and LKS+Flk2−CD34− (LT), LKS+Flk2−CD34+ (ST), LKS+Flk2+CD34+ (MPP) subpopulations (n = 5) is shown before (D0) and during hematopoietic regeneration (D15). (D) Immunophenotypic CMP (C), GMP (G) and MEP (M) is shown before (D0) and after (D15) chemotherapy (n = 5). (E) Expansion of immature MPPs was functionally validated using the CFU-S assay (n = 3). (F) Bi-dimensional cell cycle analysis is shown for p57-KO LKS + SLAM cells isolated during regeneration (D15) from chimeric mice with p57-WT (black bars) or p57-KO (white bars) hematopoiesis. (G) The population of quiescent LKS+ cells before treatment (D0) and at various times after treatment with 5FU is shown. (H) Expression of p57, p21, p18, and p27 mRNA in Lin− BM cells of p57-WT and p57-KO recipients was assessed by qRT-PCR before and at the indicated times after chemotherapy administration. Expression was normalized to Hprt1 at each time point (n = 3). All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 8.
Figure 8.
Blockade of TGFβ during recovery from hematologic stress prolongs HSPC cycling. (A) Chimeric mice with p57-WT or p57-KO hematopoiesis were injected with phenylhydrazine hydrochloride (PHZ) on day 1 and 2, and then killed on day 8 for LKS + SLAM cell cycle analysis. (B) C57BL/6J mice were injected with PHZ as previously described, and then with either the TGFβ-neutralizing antibody 1D11 (PHZ-I) or control antibody 13C4 (PHZ-C) on day 3 and 4 before analysis on day 8. (C) Chimeric mice with either p57-WT or p57-KO hematopoiesis (top) and C57BL/6J mice (bottom) were treated with PHZ as described, and RBC recovery was monitored during recovery. (D and E) C57BL/6J mice were injected with LPS on day 1, and then with either 1D11 (LPS-I) or 13C4 (LPS-C) 6 h later. Cell cycle status of LKS + SLAM cells was analyzed on day 3 (D) and 4 (E) after LPS administration. (F–I) Lethally irradiated mice were transplanted with 2 × 106 WT BM cells on day 1 and administered either 1D11 (BMT-I) or 13C4 (BMT-C) on day 7, 9, 11, and 14 after transplantation. Mice were sacrificed on day 19 for LKS + SLAM cell cycle analysis (F). Recovery of blood WBCs (G), RBCs (H), and PLTs (I) was monitored over time for transplanted mice treated with either 1D11 (BMT-I, white squares) or 13C4 (BMT-C, black circles). All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).
Figure 9.
Figure 9.
Hematopoietic stress confers a competitive advantage to p57-KO HSCs. (A) Schematic representation of competitive repopulation analysis. CD45.1+ control FLMCs were mixed 1:1 with a test population of either p57-WT or p57-KO FLMCs (EGFP+/+), and then transplanted into lethally irradiated recipient mice (n = 10). Stably engrafted mice with p57-WT or p57-KO hematopoiesis were analyzed at steady state (B) or after chemotherapy (C). (B) Multilineage peripheral blood engraftment was analyzed by flow cytometry at the indicated times after reconstitution to assess the proportion of p57-WT or p57-KO cells (n = 10). (C) To study the effect of myelosuppressive stress in competitively transplanted recipient mice, recipients were allowed to reach homeostasis (10 wk), and then given a single dose of myelosuppressive chemotherapy. Multilineage engraftment was assessed as described previously to monitor the relative contribution of p57-WT or p57-KO hematopoiesis during regeneration (n = 10). The percentage of donor cells was normalized to the day 0 measure for each mouse. The orange box represents the standard deviation of the day 0 measurements. (D) Kaplan-Meier survival curve is shown for chimeric mice stably engrafted with p57-WT or p57-KO hematopoiesis (n = 10), and then cyclically treated every 4 wk with a myelosuppressive dose of 5FU. All quantified data are shown as mean ± SEM (*, P < 0.05; **, P < 0.01; ***, P < 0.001, or if undesignated, the comparison was not significant).

References

    1. Annes J.P., Munger J.S., Rifkin D.B. 2003. Making sense of latent TGFbeta activation. J. Cell Sci. 116:217–224 10.1242/jcs.00229
    1. Barcellos-Hoff M.H., Dix T.A. 1996. Redox-mediated activation of latent transforming growth factor-beta 1. Mol. Endocrinol. 10:1077–1083 10.1210/me.10.9.1077
    1. Batard P., Monier M.N., Fortunel N., Ducos K., Sansilvestri-Morel P., Phan T., Hatzfeld A., Hatzfeld J.A. 2000. TGF-(beta)1 maintains hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation. J. Cell Sci. 113:383–390
    1. Biswas S., Guix M., Rinehart C., Dugger T.C., Chytil A., Moses H.L., Freeman M.L., Arteaga C.L. 2007. Inhibition of TGF-beta with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Invest. 117:1305–1313 10.1172/JCI30740
    1. Blank U., Karlsson S. 2011. The role of Smad signaling in hematopoiesis and translational hematology. Leukemia. 25:1379–1388 10.1038/leu.2011.95
    1. Blank U., Karlsson G., Moody J.L., Utsugisawa T., Magnusson M., Singbrant S., Larsson J., Karlsson S. 2006. Smad7 promotes self-renewal of hematopoietic stem cells. Blood. 108:4246–4254 10.1182/blood-2006-02-005611
    1. Bradford G.B., Williams B., Rossi R., Bertoncello I. 1997. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp. Hematol. 25:445–453
    1. Capron C., Lacout C., Lécluse Y., Jalbert V., Chagraoui H., Charrier S., Galy A., Bennaceur-Griscelli A., Cramer-Bordé E., Vainchenker W. 2010. A major role of TGF-beta1 in the homing capacities of murine hematopoietic stem cell/progenitors. Blood. 116:1244–1253 10.1182/blood-2009-05-221093
    1. Cheng T., Rodrigues N., Dombkowski D., Stier S., Scadden D.T. 2000a. Stem cell repopulation efficiency but not pool size is governed by p27(kip1). Nat. Med. 6:1235–1240 10.1038/81335
    1. Cheng T., Rodrigues N., Shen H., Yang Y., Dombkowski D., Sykes M., Scadden D.T. 2000b. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science. 287:1804–1808 10.1126/science.287.5459.1804
    1. Cheng T., Shen H., Rodrigues N., Stier S., Scadden D.T. 2001. Transforming growth factor beta 1 mediates cell-cycle arrest of primitive hematopoietic cells independent of p21(Cip1/Waf1) or p27(Kip1). Blood. 98:3643–3649 10.1182/blood.V98.13.3643
    1. Cheshier S.H., Morrison S.J., Liao X., Weissman I.L. 1999. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc. Natl. Acad. Sci. USA. 96:3120–3125 10.1073/pnas.96.6.3120
    1. Crawford S.E., Stellmach V., Murphy-Ullrich J.E., Ribeiro S.M., Lawler J., Hynes R.O., Boivin G.P., Bouck N. 1998. Thrombospondin-1 is a major activator of TGF-beta1 in vivo. Cell. 93:1159–1170 10.1016/S0092-8674(00)81460-9
    1. Csaszar E., Kirouac D.C., Yu M., Wang W., Qiao W., Cooke M.P., Boitano A.E., Ito C., Zandstra P.W. 2012. Rapid expansion of human hematopoietic stem cells by automated control of inhibitory feedback signaling. Cell Stem Cell. 10:218–229 10.1016/j.stem.2012.01.003
    1. Dickson M.C., Martin J.S., Cousins F.M., Kulkarni A.B., Karlsson S., Akhurst R.J. 1995. Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. Development. 121:1845–1854
    1. Ehninger A., Trumpp A. 2011. The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J. Exp. Med. 208:421–428 10.1084/jem.20110132
    1. Fortunel N., Hatzfeld J., Aoustin L., Batard P., Ducos K., Monier M.N., Charpentier A., Hatzfeld A. 2000a. Specific dose-response effects of TGF-beta1 on developmentally distinct hematopoietic stem/progenitor cells from human umbilical cord blood. Hematol. J. 1:126–135 10.1038/sj.thj.6200021
    1. Fortunel N.O., Hatzfeld A., Hatzfeld J.A. 2000b. Transforming growth factor-beta: pleiotropic role in the regulation of hematopoiesis. Blood. 96:2022–2036
    1. Gorelik L., Flavell R.A. 2000. Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity. 12:171–181 10.1016/S1074-7613(00)80170-3
    1. Heissig B., Hattori K., Dias S., Friedrich M., Ferris B., Hackett N.R., Crystal R.G., Besmer P., Lyden D., Moore M.A., et al. 2002. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 109:625–637 10.1016/S0092-8674(02)00754-7
    1. Iwata J.-I., Hacia J.G., Suzuki A., Sanchez-Lara P.A., Urata M., Chai Y. 2012. Modulation of noncanonical TGF-β signaling prevents cleft palate in Tgfbr2 mutant mice. J. Clin. Invest. 122:873–885 10.1172/JCI61498
    1. Karlsson G., Blank U., Moody J.L., Ehinger M., Singbrant S., Deng C.X., Karlsson S. 2007. Smad4 is critical for self-renewal of hematopoietic stem cells. J. Exp. Med. 204:467–474 10.1084/jem.20060465
    1. Kiel M.J., Yilmaz O.H., Iwashita T., Yilmaz O.H., Terhorst C., Morrison S.J. 2005. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 121:1109–1121 10.1016/j.cell.2005.05.026
    1. Kollet O., Dar A., Shivtiel S., Kalinkovich A., Lapid K., Sztainberg Y., Tesio M., Samstein R.M., Goichberg P., Spiegel A., et al. 2006. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat. Med. 12:657–664 10.1038/nm1417
    1. Kopp H.G., Avecilla S.T., Hooper A.T., Shmelkov S.V., Ramos C.A., Zhang F., Rafii S. 2005. Tie2 activation contributes to hemangiogenic regeneration after myelosuppression. Blood. 106:505–513 10.1182/blood-2004-11-4269
    1. Lapidot T., Petit I. 2002. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp. Hematol. 30:973–981 10.1016/S0301-472X(02)00883-4
    1. Larsson J., Goumans M.J., Sjöstrand L.J., van Rooijen M.A., Ward D., Levéen P., Xu X., ten Dijke P., Mummery C.L., Karlsson S. 2001. Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptor-deficient mice. EMBO J. 20:1663–1673 10.1093/emboj/20.7.1663
    1. Larsson J., Blank U., Helgadottir H., Björnsson J.M., Ehinger M., Goumans M.J., Fan X., Levéen P., Karlsson S. 2003. TGF-beta signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro. Blood. 102:3129–3135 10.1182/blood-2003-04-1300
    1. Larsson J., Blank U., Klintman J., Magnusson M., Karlsson S. 2005. Quiescence of hematopoietic stem cells and maintenance of the stem cell pool is not dependent on TGF-beta signaling in vivo. Exp. Hematol. 33:592–596 10.1016/j.exphem.2005.02.003
    1. Letterio J.J., Geiser A.G., Kulkarni A.B., Dang H., Kong L., Nakabayashi T., Mackall C.L., Gress R.E., Roberts A.B. 1996. Autoimmunity associated with TGF-beta1-deficiency in mice is dependent on MHC class II antigen expression. J. Clin. Invest. 98:2109–2119 10.1172/JCI119017
    1. Levéen P., Larsson J., Ehinger M., Cilio C.M., Sundler M., Sjöstrand L.J., Holmdahl R., Karlsson S. 2002. Induced disruption of the transforming growth factor beta type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable. Blood. 100:560–568 10.1182/blood.V100.2.560
    1. Lévesque J.P., Winkler I.G. 2011. Hierarchy of immature hematopoietic cells related to blood flow and niche. Curr. Opin. Hematol. 18:220–225 10.1097/MOH.0b013e3283475fe7
    1. Lévesque J.P., Hendy J., Takamatsu Y., Simmons P.J., Bendall L.J. 2003. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide. J. Clin. Invest. 111:187–196
    1. Matsumoto A., Takeishi S., Kanie T., Susaki E., Onoyama I., Tateishi Y., Nakayama K., Nakayama K.I. 2011. p57 is required for quiescence and maintenance of adult hematopoietic stem cells. Cell Stem Cell. 9:262–271 10.1016/j.stem.2011.06.014
    1. McCulloch E.A. 2002. CFU-S. In Hematopoietic Stem Cell Protocols. Klug C.A., Jordan C.T., Humana Press, Inc, Totowa, NJ. 153-160
    1. Miyamoto K., Araki K.Y., Naka K., Arai F., Takubo K., Yamazaki S., Matsuoka S., Miyamoto T., Ito K., Ohmura M., et al. 2007. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell. 1:101–112 10.1016/j.stem.2007.02.001
    1. Morrison S.J., Wright D.E., Weissman I.L. 1997. Cyclophosphamide/granulocyte colony-stimulating factor induces hematopoietic stem cells to proliferate prior to mobilization. Proc. Natl. Acad. Sci. USA. 94:1908–1913 10.1073/pnas.94.5.1908
    1. Mu D., Cambier S., Fjellbirkeland L., Baron J.L., Munger J.S., Kawakatsu H., Sheppard D., Broaddus V.C., Nishimura S.L. 2002. The integrin alpha(v)beta8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-beta1. J. Cell Biol. 157:493–507 10.1083/jcb.200109100
    1. Munger J.S., Harpel J.G., Giancotti F.G., Rifkin D.B. 1998. Interactions between growth factors and integrins: latent forms of transforming growth factor-beta are ligands for the integrin alphavbeta1. Mol. Biol. Cell. 9:2627–2638
    1. Munger J.S., Huang X., Kawakatsu H., Griffiths M.J., Dalton S.L., Wu J., Pittet J.F., Kaminski N., Garat C., Matthay M.A., et al. 1999. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 96:319–328 10.1016/S0092-8674(00)80545-0
    1. Muraoka R.S., Dumont N., Ritter C.A., Dugger T.C., Brantley D.M., Chen J., Easterly E., Roebuck L.R., Ryan S., Gotwals P.J., et al. 2002. Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J. Clin. Invest. 109:1551–1559
    1. Oshima M., Oshima H., Taketo M.M. 1996. TGF-beta receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis. Dev. Biol. 179:297–302 10.1006/dbio.1996.0259
    1. Passegué E., Wagers A.J., Giuriato S., Anderson W.C., Weissman I.L. 2005. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J. Exp. Med. 202:1599–1611 10.1084/jem.20050967
    1. Petit I., Szyper-Kravitz M., Nagler A., Lahav M., Peled A., Habler L., Ponomaryov T., Taichman R.S., Arenzana-Seisdedos F., Fujii N., et al. 2002. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat. Immunol. 3:687–694 10.1038/ni813
    1. Pfeilschifter J., Diel I., Scheppach B., Bretz A., Krempien R., Erdmann J., Schmid G., Reske N., Bismar H., Seck T., et al. 1998. Concentration of transforming growth factor beta in human bone tissue: relationship to age, menopause, bone turnover, and bone volume. J. Bone Miner. Res. 13:716–730 10.1359/jbmr.1998.13.4.716
    1. Qian H., Buza-Vidas N., Hyland C.D., Jensen C.T., Antonchuk J., Månsson R., Thoren L.A., Ekblom M., Alexander W.S., Jacobsen S.E. 2007. Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells. Cell Stem Cell. 1:671–684 10.1016/j.stem.2007.10.008
    1. Ribeiro S.M., Poczatek M., Schultz-Cherry S., Villain M., Murphy-Ullrich J.E. 1999. The activation sequence of thrombospondin-1 interacts with the latency-associated peptide to regulate activation of latent transforming growth factor-beta. J. Biol. Chem. 274:13586–13593 10.1074/jbc.274.19.13586
    1. Sato Y., Tsuboi R., Lyons R., Moses H., Rifkin D.B. 1990. Characterization of the activation of latent TGF-beta by co-cultures of endothelial cells and pericytes or smooth muscle cells: a self-regulating system. J. Cell Biol. 111:757–763 10.1083/jcb.111.2.757
    1. Scandura J.M., Boccuni P., Massagué J., Nimer S.D. 2004. Transforming growth factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation. Proc. Natl. Acad. Sci. USA. 101:15231–15236 10.1073/pnas.0406771101
    1. Schaefer B.C., Schaefer M.L., Kappler J.W., Marrack P., Kedl R.M. 2001. Observation of antigen-dependent CD8+ T-cell/ dendritic cell interactions in vivo. Cell. Immunol. 214:110–122 10.1006/cimm.2001.1895
    1. Sitnicka E., Ruscetti F.W., Priestley G.V., Wolf N.S., Bartelmez S.H. 1996. Transforming growth factor beta 1 directly and reversibly inhibits the initial cell divisions of long-term repopulating hematopoietic stem cells. Blood. 88:82–88
    1. Takahashi K., Nakayama K., Nakayama K. 2000. Mice lacking a CDK inhibitor, p57Kip2, exhibit skeletal abnormalities and growth retardation. J. Biochem. 127:73–83 10.1093/oxfordjournals.jbchem.a022586
    1. Tang Y., Wu X., Lei W., Pang L., Wan C., Shi Z., Zhao L., Nagy T.R., Peng X., Hu J., et al. 2009. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat. Med. 15:757–765 10.1038/nm.1979
    1. Tesio M., Trumpp A. 2011. Breaking the cell cycle of HSCs by p57 and friends. Cell Stem Cell. 9:187–192 10.1016/j.stem.2011.08.005
    1. Wilson A., Trumpp A. 2006. Bone-marrow haematopoietic-stem-cell niches. Nat. Rev. Immunol. 6:93–106 10.1038/nri1779
    1. Wilson A., Laurenti E., Oser G., van der Wath R.C., Blanco-Bose W., Jaworski M., Offner S., Dunant C.F., Eshkind L., Bockamp E., et al. 2008. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 135:1118–1129 10.1016/j.cell.2008.10.048
    1. Wu X., Pang L., Lei W., Lu W., Li J., Li Z., Frassica F.J., Chen X., Wan M., Cao X. 2010. Inhibition of Sca-1-positive skeletal stem cell recruitment by alendronate blunts the anabolic effects of parathyroid hormone on bone remodeling. Cell Stem Cell. 7:571–580 10.1016/j.stem.2010.09.012
    1. Yamazaki S., Iwama A., Takayanagi S., Morita Y., Eto K., Ema H., Nakauchi H. 2006. Cytokine signals modulated via lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells. EMBO J. 25:3515–3523 10.1038/sj.emboj.7601236
    1. Yamazaki S., Iwama A., Takayanagi S., Eto K., Ema H., Nakauchi H. 2009. TGF-beta as a candidate bone marrow niche signal to induce hematopoietic stem cell hibernation. Blood. 113:1250–1256 10.1182/blood-2008-04-146480
    1. Yamazaki S., Ema H., Karlsson G., Yamaguchi T., Miyoshi H., Shioda S., Taketo M.M., Karlsson S., Iwama A., Nakauchi H. 2011. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell. 147:1146–1158 10.1016/j.cell.2011.09.053
    1. Yan Y., Frisén J., Lee M.H., Massagué J., Barbacid M. 1997. Ablation of the CDK inhibitor p57Kip2 results in increased apoptosis and delayed differentiation during mouse development. Genes Dev. 11:973–983
    1. Yang Y.-A., Dukhanina O., Tang B., Mamura M., Letterio J.J., MacGregor J., Patel S.C., Khozin S., Liu Z.-Y., Green J., et al. 2002. Lifetime exposure to a soluble TGF-beta antagonist protects mice against metastasis without adverse side effects. J. Clin. Invest. 109:1607–1615
    1. Yang Z., Mu Z., Dabovic B., Jurukovski V., Yu D., Sung J., Xiong X., Munger J.S. 2007. Absence of integrin-mediated TGFbeta1 activation in vivo recapitulates the phenotype of TGFbeta1-null mice. J. Cell Biol. 176:787–793 10.1083/jcb.200611044
    1. Yaswen L., Kulkarni A.B., Fredrickson T., Mittleman B., Schiffman R., Payne S., Longenecker G., Mozes E., Karlsson S. 1996. Autoimmune manifestations in the transforming growth factor-beta 1 knockout mouse. Blood. 87:1439–1445
    1. Yilmaz O.H., Kiel M.J., Morrison S.J. 2006. SLAM family markers are conserved among hematopoietic stem cells from old and reconstituted mice and markedly increase their purity. Blood. 107:924–930 10.1182/blood-2005-05-2140
    1. Yingling J.M., Blanchard K.L., Sawyer J.S. 2004. Development of TGF-beta signalling inhibitors for cancer therapy. Nat. Rev. Drug Discov. 3:1011–1022 10.1038/nrd1580
    1. Yoshihara H., Arai F., Hosokawa K., Hagiwara T., Takubo K., Nakamura Y., Gomei Y., Iwasaki H., Matsuoka S., Miyamoto K., et al. 2007. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 1:685–697 10.1016/j.stem.2007.10.020
    1. Yu Q., Stamenkovic I. 2000. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes Dev. 14:163–176
    1. Yuan Y., Shen H., Franklin D.S., Scadden D.T., Cheng T. 2004. In vivo self-renewing divisions of haematopoietic stem cells are increased in the absence of the early G1-phase inhibitor, p18INK4C. Nat. Cell Biol. 6:436–442 10.1038/ncb1126
    1. Zhang P., Liégeois N.J., Wong C., Finegold M., Hou H., Thompson J.C., Silverman A., Harper J.W., DePinho R.A., Elledge S.J. 1997. Altered cell differentiation and proliferation in mice lacking p57KIP2 indicates a role in Beckwith-Wiedemann syndrome. Nature. 387:151–158 10.1038/387151a0
    1. Zou P., Yoshihara H., Hosokawa K., Tai I., Shinmyozu K., Tsukahara F., Maru Y., Nakayama K., Nakayama K.I., Suda T. 2011. p57(Kip2) and p27(Kip1) cooperate to maintain hematopoietic stem cell quiescence through interactions with Hsc70. Cell Stem Cell. 9:247–261 10.1016/j.stem.2011.07.003

Source: PubMed

3
Tilaa