Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways

Jian Huang, Michelle Nguyen-McCarty, Elizabeth O Hexner, Gwenn Danet-Desnoyers, Peter S Klein, Jian Huang, Michelle Nguyen-McCarty, Elizabeth O Hexner, Gwenn Danet-Desnoyers, Peter S Klein

Abstract

Hematopoietic stem cell (HSC) self renewal and lineage commitment depend on complex interactions with the microenvironment. The ability to maintain or expand HSCs for clinical applications or basic research has been substantially limited because these interactions are not well defined. Recent evidence suggests that HSCs reside in a low-perfusion, reduced-nutrient niche and that nutrient-sensing pathways contribute to HSC homeostasis. Here we report that suppression of the mTOR pathway, an established nutrient sensor, combined with activation of canonical Wnt-β-catenin signaling, allows for the ex vivo maintenance of human and mouse long-term HSCs under cytokine-free conditions. We also show that the combination of two clinically approved medications that together activate Wnt-β-catenin and inhibit mTOR signaling increases the number (but not the proportion) of long-term HSCs in vivo.

Conflict of interest statement

The authors have no conflicts of interest.

Figures

Figure 1. Inhibition of GSK-3 and mTOR…
Figure 1. Inhibition of GSK-3 and mTOR preserves HSPCs
(a) Experiment design: Mouse c-Kit+ or LSK cells were cultured in cytokine-free medium with inhibitors (CR) or vehicle for 7 d, then either plated on OP9 stromal cells or transplanted into lethally irradiated mice. FCM was performed after 3 weeks (OP9) or 4 months (transplantation, Figure 2). (b) Sorted LSK cells were cultured for 7 d, then plated on OP9 cells. Markers for myeloid (OP9), T and B lineages (OP9-DL1) (left) were measured at 3 weeks by FCM. Representative FCM data on left, quantification of myeloid, T and B lineages on right.
Figure 2. Maintenance of long-term HSCs ex…
Figure 2. Maintenance of long-term HSCs ex vivo by inhibition of GSK-3 and mTOR
(a) Mouse c-Kit+ cells were cultured in cytokine-free medium for 7 d with the inhibitors as indicated, then competitive serial transplants were performed. Chimerism in bone marrow (BM) was measured by FCM after 4 months. The data are from 3 independent experiments. (b–d) LSK cells were cultured in cytokine-free medium for 7 d, then competitive serial transplants were performed in primary (b), secondary (c), and tertiary recipients (d). Bone marrow was harvested after 4 months for analysis of chimerism and for transplant to subsequent recipients. (e) Limiting dilution experiments were performed with three doses of cultured c-Kit+ test marrow (CD45.1+) from control or CR treated cells combined with 2×105 competing cells (CD45.2+) transplanted into groups of at least nine recipients per dose. Chimerism at 4 months for each dose is represented as the percentage of mice negative for engraftment in BM for vehicle control (green triangle), CR treated cells (red square), and uncultured cells (blue diamond).
Figure 3. Human long-term HSCs maintained in…
Figure 3. Human long-term HSCs maintained in cytokine-free culture
(a) Human umbilical cord blood (UCB) CD34+ cells were cultured in cytokine-free medium with or without Chiron+rapamycin (CR) for 7 d, then competitive serial transplants were performed by transplanting all the cells into sub-lethally irradiated NSG recipients. Engraftment was assessed as percent of human CD45+ cells in BM after 4 months. (b) Representative FCM data (CD45+) are shown for BM from primary recipients of human CD34+ cells that were directly transplanted (uncultured) or cultured in vehicle (DMSO), CR, or conventional cytokines. (c) Progenitors, lymphoid (T-cell and B-cell), and myeloid lineages derived from human CD34+ cells that were directly transplanted (uncultured) or cultured in CR for 7 d before transplant. Similar numbers of each lineage were detected by FCM. (d) Four months after primary transplantation, BM was transplanted into 2° NOD-SCID recipients and chimerism was assessed after 4 months as in A.
Figure 4. Maintenance of long-term HSCs in…
Figure 4. Maintenance of long-term HSCs in culture requires β-catenin
(a) BM was harvested from mice with a conditional deletion of β-catenin (using cre-ERT2) or cre- controls, and LSK cells were cultured in cytokine-free medium with or without inhibitors (CR) for 7 d. Cultured cells were then plated on OP9 cells and assessed by FCM after 3 weeks as in Fig. 1. (b) C-Kit+ cells were harvested from mice with a conditional deletion of β-catenin (using mx-cre) or cre- controls and cultured in cytokine-free medium with or without inhibitors (CR) for 7 d, then competitive transplants were performed by transplanting all the cells in each group mixed with 3X105 fresh BM cells into lethally irradiated recipients. Chimerism in BM was measured by FCM for CD45.1+LSK,CD150+CD48−Flk2− cells after 4 months. For panels (a) and (b), depletion of β-catenin was confirmed by western blot (Supplementary Fig. 5c,d). (c) To assess cell cycle status, c-Kit+ cells were cultured for 4 d and analyzed by FCM for LSK markers, DAPI staining, and Ki67. Representative FACS data are shown for control (DMSO) vs. CR cultured cells. (d) To assess apoptosis, c-Kit+ cells were cultured as in A and analyzed by FCM for LSK, 7AAD, and Annexin V.
Figure 5. Lithium and rapamycin increase the…
Figure 5. Lithium and rapamycin increase the number of long-term HSCs in vivo
(a) Mouse c-Kit+ cells were cultured for 7 d in cytokine-free medium with or without lithium and rapamycin (LR) and transplanted into lethally irradiated recipients. Survival of recipients receiving cells cultured with vehicle (blue diamond), lithium (red square), rapamycin (purple circle), or LR (green triangle) is shown as a Kaplan-Meier plot. (b) Representative FCM of donor-derived phenotypic HSCs in BM (upper panels) and myeloid and lymphoid lineages in peripheral blood (lower panels) for recipients of LR cultured cells. (c) Mice received lithium and rapamycin for 3 weeks. Cellularity of BM in vehicle control, lithium, rapamycin, and LR treated mice was measured as the number of nucleated cells/mouse recovered from one femur and tibia. (d) Absolute number of phenotypic HSCs (LSK,CD34−Flk2−CD150+CD48−) in BM in control, lithium, rapamycin, and LR treated mice. (e) Mice were treated with lithium and rapamycin for 3 weeks. BM was harvested, limiting dilution assay was performed, and competitive rescue units for control and LR are shown. (f) Western blot analysis for β-catenin, phosphorylated-S6, S6, phosphorylated-4EBP in BM cells harvested from control, lithium, rapamycin, and LR treated mice.

References

    1. Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell. 2000;100:157–68.
    1. Purton LE, Scadden DT. Limiting factors in murine hematopoietic stem cell assays. Cell Stem Cell. 2007;1:263–270.
    1. Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology. Cell. 2008;132:631–44.
    1. Chou S, Chu P, Hwang W, Lodish H. Expansion of human cord blood hematopoietic stem cells for transplantation. Cell Stem Cell. 2010;7:427–428.
    1. Lymperi S, Ferraro F, Scadden DT. The HSC niche concept has turned 31. Has our knowledge matured? Ann N Y Acad Sci. 2010;1192:12–18.
    1. Bowman TV, Zon LI. Lessons from the niche for generation and expansion of hematopoietic stem cells. Drug Discovery Today: Therapeutic Strategies. 2009;6:135–140.
    1. Parmar K, Mauch P, Vergilio JA, Sackstein R, Down JD. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A. 2007;104:5431–6.
    1. Simsek T, et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell. 2010;7:380–90.
    1. Miharada K, et al. Cripto regulates hematopoietic stem cells as a hypoxic-niche-related factor through cell surface receptor GRP78. Cell Stem Cell. 2011;9:330–44.
    1. Takubo K, et al. Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell. 2010;7:391–402.
    1. Krock B, Skuli N, Simon MC. The tumor suppressor LKB1 emerges as a critical factor in hematopoietic stem cell biology. Cell Metabolism. 2011;13:8–10.
    1. Gan B, et al. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature. 2010;468:701–4.
    1. Gurumurthy S, et al. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature. 2010;468:659–63.
    1. Nakada D, Saunders TL, Morrison SJ. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature. 2010;468:653–8.
    1. Yilmaz OH, et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature. 2006;441:475–82.
    1. Zhang J, et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature. 2006;441:518–22.
    1. Chen C, et al. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. Journal of Experimental Medicine. 2008;205:2397–408.
    1. Hofmeister CC, Zhang J, Knight KL, Le P, Stiff PJ. Ex vivo expansion of umbilical cord blood stem cells for transplantation: growing knowledge from the hematopoietic niche. Bone Marrow Transplant. 2007;39:11–23.
    1. Zhang CC, Kaba M, Iizuka S, Huynh H, Lodish HF. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood. 2008;111:3415–23.
    1. Zhang CC, et al. Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med. 2006;12:240–245.
    1. Himburg HA, et al. Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells. Nat Med. 2010;16:475–482.
    1. Delaney C, et al. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nature Medicine. 2010;16:232–236.
    1. Goessling W, et al. Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell. 2009;136:1136–47.
    1. Boitano AE, et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329:1345–1348.
    1. Perry JM, et al. Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 2011;25:1928–42.
    1. Zhang CC, Lodish HF. Murine hematopoietic stem cells change their surface phenotype during ex vivo expansion. Blood. 2005;105:4314–20.
    1. Staal FJ, Burgering BM, van de Wetering M, Clevers HC. Tcf-1-mediated transcription in T lymphocytes: differential role for glycogen synthase kinase-3 in fibroblasts and T cells. Int Immunol. 1999;11:317–323.
    1. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434:843–50.
    1. Staal FJ, Sen JM. The canonical Wnt signaling pathway plays an important role in lymphopoiesis and hematopoiesis. European Journal of Immunology. 2008;38:1788–94.
    1. Malhotra S, Kincade PW. Wnt-related molecules and signaling pathway equilibrium in hematopoiesis. Cell Stem Cell. 2009;4:27–36.
    1. Sato N, Meijer L, Skaltsounis L, Greengard P, Brivanlou AH. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med. 2004;10:55–63.
    1. Trowbridge JJ, Xenocostas A, Moon RT, Bhatia M. Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation. Nat Med. 2006;12:89–98.
    1. Luis TC, et al. Canonical wnt signaling regulates hematopoiesis in a dosage-dependent fashion. Cell Stem Cell. 2011;9:345–56.
    1. Luis TC, et al. Wnt3a deficiency irreversibly impairs hematopoietic stem cell self-renewal and leads to defects in progenitor cell differentiation. Blood. 2009;113:546–554.
    1. Zhao C, et al. Loss of beta-Catenin Impairs the Renewal of Normal and CML Stem Cells In vivo. Cancer Cell. 2007;12:528–541.
    1. Fleming HE, et al. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell. 2008;2:274–83.
    1. Willert K, et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature. 2003;423:448–52.
    1. Reya T, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003;423:409–14.
    1. Cobas M, et al. Beta-catenin is dispensable for hematopoiesis and lymphopoiesis. Journal of Experimental Medicine. 2004;199:221–9.
    1. Jeannet G, et al. Long-term, multilineage hematopoiesis occurs in the combined absence of beta-catenin and gamma-catenin. Blood. 2008;111:142–149.
    1. Koch U, et al. Simultaneous loss of beta- and gamma-catenin does not perturb hematopoiesis or lymphopoiesis. Blood. 2008;111:160–4.
    1. Kirstetter P, Anderson K, Porse BT, Jacobsen SE, Nerlov C. Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nature Immunology. 2006;7:1048–56.
    1. Inoki K, et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell. 2006;126:955–968.
    1. Holmes R, Zuniga-Pflucker JC. The OP9-DL1 system: generation of T-lymphocytes from embryonic or hematopoietic stem cells in vitro. Cold Spring Harb Protoc. 2009 doi: 10.1101/pdb.prot5156.
    1. Kodama H, Nose M, Niida S, Nishikawa S, Nishikawa S. Involvement of the c-Kit receptor in the adhesion of hematopoietic stem cells to stromal cells. Exp Hematol. 1994;22:979–84.
    1. Zediak VP, Maillard I, Bhandoola A. Multiple prethymic defects underlie age-related loss of T progenitor competence. Blood. 2007;110:1161–7.
    1. Richie Ehrlich LI, Serwold T, Weissman IL. In vitro assays misrepresent in vivo lineage potentials of murine lymphoid progenitors. Blood. 2011;117:2618–24.
    1. Maretto S, et al. Mapping Wnt/beta-catenin signaling during mouse development and in colorectal tumors. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:3299–304.
    1. Holmes T, O’Brien TA, Knight R, Lindeman R, Shen S, Song E, Symonds G, Dolnikov A. Glycogen synthase kinase-3beta inhibition preserves hematopoietic stem cell activity and inhibits leukemic cell growth. Stem Cells. 2008;26:1288–1297.
    1. Castilho RM, Squarize CH, Chodosh LA, Williams BO, Gutkind JS. mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell. 2009;5:279–289.
    1. Boggs DR, Joyce RA. The Hematopoietic Effects of Lithium. Seminars in Hematology. 1983;20:129–138.
    1. Ballin A, Lehman D, Sirota P, Litvinjuk U, Meytes D. Increased number of peripheral blood CD34+ cells in lithium-treated patients. Br J Haematol. 1998;100:219–21.
    1. Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837–51.
    1. Wang Y, et al. The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science. 2010;327:1650–3.
    1. Cohen Y, Chetrit A, Sirota P, Modan B. Cancer morbidity in psychiatric patients: influence of lithium carbonate treatment. Medical Oncology. 1998;15:32–6.
    1. Gattinoni L, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–1297.
    1. Gattinoni L, Klebanoff CA, Restifo NP. Pharmacologic induction of CD8+ T cell memory: better living through chemistry. Sci Transl Med. 2009;1:1–6.
    1. Schmitt TM, Zuniga-Pflucker JC. Induction of Tcell development from hematopoietic progenitor cells by delta-like-1 in vitro. Immunity. 2002;17:749–756.

Source: PubMed

3
Předplatit