Skip to main content
Top
Published in: Current Osteoporosis Reports 4/2011

01-12-2011 | Epidemiology and Pathophysiology (Mone Zaidi and Jeffrey I. Mechanick, Section Editors)

Stem Cell Interactions in a Bone Marrow Niche

Authors: Joan Isern, Simón Méndez-Ferrer

Published in: Current Osteoporosis Reports | Issue 4/2011

Login to get access

Abstract

Stem cells differ from other cells of the body in their potential for multilineage differentiation and their continued proliferation without substantial loss of potential (so-called self-renewal). These properties are maintained and regulated by a specific microenvironment referred to as “niche.” This term has been used to indicate the specific location of stem cells within tissues, as well as the cellular and molecular components that critically determine stem cell behavior. Whereas other, perhaps less complex, stem cell niches (e.g., Drosophila germarium) have been more clearly dissected in the 30 years that have passed since these observations, the hematopoietic stem cell (HSC) niche has proven challenging due to the difficulty to detect HSCs under normal conditions and the dynamism of HSCs and other cells of the bone marrow that influence HSC behavior. This article reviews the recent development of the HSC niche field with emphasis on prospective integrative mechanisms within bone marrow homeostasis and multisystem physiology. For that purpose, we will first highlight anatomical and histological features of the bone marrow of relevance for HSC behavior; then, we will summarize the principal findings concerning different cell types and potential mechanisms by which they critically regulate HSC function.
Literature
1.
go back to reference Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cell. 1978;4(1–2):7–25. Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cell. 1978;4(1–2):7–25.
2.
go back to reference Kiel MJ, Morrison SJ. Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol. 2008;8(4):290–301.PubMedCrossRef Kiel MJ, Morrison SJ. Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol. 2008;8(4):290–301.PubMedCrossRef
3.
go back to reference • Karsenty G, Oury F. The central regulation of bone mass, the first link between bone remodeling and energy metabolism. J Clin Endocrinol Metabol. 2010;95(11):4795–801. This is a review of the central regulation of the bone and its endocrine function. CrossRef • Karsenty G, Oury F. The central regulation of bone mass, the first link between bone remodeling and energy metabolism. J Clin Endocrinol Metabol. 2010;95(11):4795–801. This is a review of the central regulation of the bone and its endocrine function. CrossRef
4.
go back to reference Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841–6.PubMedCrossRef Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841–6.PubMedCrossRef
5.
go back to reference Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425(6960):836–41.PubMedCrossRef Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425(6960):836–41.PubMedCrossRef
6.
go back to reference Nilsson SK, Johnston HM, Coverdale JA. Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches. Blood. 2001;97(8):2293–9.PubMedCrossRef Nilsson SK, Johnston HM, Coverdale JA. Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches. Blood. 2001;97(8):2293–9.PubMedCrossRef
7.
go back to reference Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky MC, et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439(7076):599–603.PubMedCrossRef Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky MC, et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439(7076):599–603.PubMedCrossRef
8.
go back to reference Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149–61.PubMedCrossRef Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149–61.PubMedCrossRef
9.
go back to reference Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, Denhardt DT, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005;106(4):1232–9.PubMedCrossRef Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, Denhardt DT, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005;106(4):1232–9.PubMedCrossRef
10.
go back to reference Stier S, Ko Y, Forkert R, Lutz C, Neuhaus T, Grunewald E, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med. 2005;201(11):1781–91.PubMedCrossRef Stier S, Ko Y, Forkert R, Lutz C, Neuhaus T, Grunewald E, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med. 2005;201(11):1781–91.PubMedCrossRef
11.
go back to reference Wilson A, Murphy MJ, Oskarsson T, Kaloulis K, Bettess MD, Oser GM, et al. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Gene Dev. 2004;18(22):2747–63.PubMedCrossRef Wilson A, Murphy MJ, Oskarsson T, Kaloulis K, Bettess MD, Oser GM, et al. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Gene Dev. 2004;18(22):2747–63.PubMedCrossRef
12.
go back to reference Haug JS, He XC, Grindley JC, Wunderlich JP, Gaudenz K, Ross JT, et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell. 2008;2(4):367–79.PubMedCrossRef Haug JS, He XC, Grindley JC, Wunderlich JP, Gaudenz K, Ross JT, et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell. 2008;2(4):367–79.PubMedCrossRef
13.
go back to reference •• Xie Y, Yin T, Wiegraebe W, He XC, Miller D, Stark D, et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature. 2009;457(7225):97–101. This is one of the first studies that applied sophisticated in vivo imaging technologies to study the localization of purified HSC homing to the bone marrow of recipient mice. PubMedCrossRef •• Xie Y, Yin T, Wiegraebe W, He XC, Miller D, Stark D, et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature. 2009;457(7225):97–101. This is one of the first studies that applied sophisticated in vivo imaging technologies to study the localization of purified HSC homing to the bone marrow of recipient mice. PubMedCrossRef
14.
go back to reference Dzierzak E, Speck NA. Of lineage and legacy: the development of mammalian hematopoietic stem cells. Nat Immunol. 2008;9(2):129–36.PubMedCrossRef Dzierzak E, Speck NA. Of lineage and legacy: the development of mammalian hematopoietic stem cells. Nat Immunol. 2008;9(2):129–36.PubMedCrossRef
15.
go back to reference Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004;103(9):3258–64.PubMedCrossRef Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004;103(9):3258–64.PubMedCrossRef
16.
go back to reference Zhu J, Garrett R, Jung Y, Zhang Y, Kim N, Wang J, et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood. 2007;109(9):3706–12.PubMedCrossRef Zhu J, Garrett R, Jung Y, Zhang Y, Kim N, Wang J, et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood. 2007;109(9):3706–12.PubMedCrossRef
17.
go back to reference •• Kiel MJ, Acar M, Radice GL, Morrison SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell. 2009;4(2):170–9. The authors report here that N-cadherin is not necessary to maintain hematopoietic stem cells. PubMedCrossRef •• Kiel MJ, Acar M, Radice GL, Morrison SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell. 2009;4(2):170–9. The authors report here that N-cadherin is not necessary to maintain hematopoietic stem cells. PubMedCrossRef
18.
go back to reference Lymperi S, Horwood N, Marley S, Gordon MY, Cope AP, Dazzi F. Strontium can increase some osteoblasts without increasing hematopoietic stem cells. Blood. 2008;111(3):1173–81.PubMedCrossRef Lymperi S, Horwood N, Marley S, Gordon MY, Cope AP, Dazzi F. Strontium can increase some osteoblasts without increasing hematopoietic stem cells. Blood. 2008;111(3):1173–81.PubMedCrossRef
19.
go back to reference Kiel MJ, Radice GL, Morrison SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell. 2007;1:204–17.PubMedCrossRef Kiel MJ, Radice GL, Morrison SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell. 2007;1:204–17.PubMedCrossRef
20.
go back to reference Li P, Zon LI. Resolving the controversy about N-cadherin and hematopoietic stem cells. Cell Stem Cell. 2010;6(3):199–202.PubMedCrossRef Li P, Zon LI. Resolving the controversy about N-cadherin and hematopoietic stem cells. Cell Stem Cell. 2010;6(3):199–202.PubMedCrossRef
21.
go back to reference •• Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7. This article showed that disruption of the ribonuclease Dicer1 in preosteoblasts, but not in mature osteoblasts, is associated with myelodysplasia and secondary leukemia. PubMedCrossRef •• Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7. This article showed that disruption of the ribonuclease Dicer1 in preosteoblasts, but not in mature osteoblasts, is associated with myelodysplasia and secondary leukemia. PubMedCrossRef
22.
go back to reference Chan CK, Chen CC, Luppen CA, Kim JB, Deboer AT, Wei K, et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature. 2008 Dec 10 Chan CK, Chen CC, Luppen CA, Kim JB, Deboer AT, Wei K, et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature. 2008 Dec 10
23.
go back to reference •• Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, Fujisaki J, et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457(7225):92–6. This is an in vivo imaging study of hematopoietic stem and progenitor cells in the skull bone marrow describing their detailed localization relative to osteoblasts, blood vessels, and the endosteal surface. PubMedCrossRef •• Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, Fujisaki J, et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457(7225):92–6. This is an in vivo imaging study of hematopoietic stem and progenitor cells in the skull bone marrow describing their detailed localization relative to osteoblasts, blood vessels, and the endosteal surface. PubMedCrossRef
24.
go back to reference Mendes SC, Robin C, Dzierzak E. Mesenchymal progenitor cells localize within hematopoietic sites throughout ontogeny. Development. 2005;132(5):1127–36.PubMedCrossRef Mendes SC, Robin C, Dzierzak E. Mesenchymal progenitor cells localize within hematopoietic sites throughout ontogeny. Development. 2005;132(5):1127–36.PubMedCrossRef
25.
go back to reference Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121(7):1109–21.PubMedCrossRef Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121(7):1109–21.PubMedCrossRef
26.
go back to reference Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25(6):977–88.PubMedCrossRef Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25(6):977–88.PubMedCrossRef
27.
go back to reference Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185(1):111–20.PubMedCrossRef Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185(1):111–20.PubMedCrossRef
28.
go back to reference Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hematopoietic stem cells are uniquely selective in their migratory response to chemokines. J Exp Med. 2002;195(9):1145–54.PubMedCrossRef Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hematopoietic stem cells are uniquely selective in their migratory response to chemokines. J Exp Med. 2002;195(9):1145–54.PubMedCrossRef
29.
go back to reference • Tzeng YS, Li H, Kang YL, Chen WC, Cheng WC, Lai DM. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood. 2010;117(2):429–39. The authors showed that conditional depletion of the chemokine CXCL12 in adult mice increases the proliferation and reduces the number of long-term hematopoietic stem cells in the bone marrow.PubMedCrossRef • Tzeng YS, Li H, Kang YL, Chen WC, Cheng WC, Lai DM. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood. 2010;117(2):429–39. The authors showed that conditional depletion of the chemokine CXCL12 in adult mice increases the proliferation and reduces the number of long-term hematopoietic stem cells in the bone marrow.PubMedCrossRef
30.
go back to reference •• Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460(7252):259–63. Description of the negative influence of adipocytes on hematopoiesis. PubMedCrossRef •• Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460(7252):259–63. Description of the negative influence of adipocytes on hematopoiesis. PubMedCrossRef
31.
go back to reference Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3(4):393–403.PubMed Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3(4):393–403.PubMed
32.
go back to reference Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276(5309):71–4.PubMedCrossRef Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276(5309):71–4.PubMedCrossRef
33.
go back to reference • Bernardo ME, Locatelli F, Fibbe WE. Mesenchymal stromal cells. Ann N Y Acad Sci. 2009;1176:101–17. This is a review of the biological and functional properties of mesenchymal stromal cells. PubMedCrossRef • Bernardo ME, Locatelli F, Fibbe WE. Mesenchymal stromal cells. Ann N Y Acad Sci. 2009;1176:101–17. This is a review of the biological and functional properties of mesenchymal stromal cells. PubMedCrossRef
34.
go back to reference Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301–13.PubMedCrossRef Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301–13.PubMedCrossRef
35.
go back to reference Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 1991;78(1):55–62.PubMed Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 1991;78(1):55–62.PubMed
36.
go back to reference Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007;131(2):324–36.PubMedCrossRef Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007;131(2):324–36.PubMedCrossRef
37.
go back to reference •• Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34. This article identifies murine mesenchymal stem cells using nestin expression and their essential function in the HSC niche. PubMedCrossRef •• Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34. This article identifies murine mesenchymal stem cells using nestin expression and their essential function in the HSC niche. PubMedCrossRef
38.
go back to reference Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2008;2(4):313–9.PubMedCrossRef Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2008;2(4):313–9.PubMedCrossRef
39.
go back to reference • Bianco P. Back to the future: moving beyond “mesenchymal stem cells”. J Cell Biochem. 2011;112(7):1713–21. This review provides a historical perspective on skeletal stem cells. PubMedCrossRef • Bianco P. Back to the future: moving beyond “mesenchymal stem cells”. J Cell Biochem. 2011;112(7):1713–21. This review provides a historical perspective on skeletal stem cells. PubMedCrossRef
40.
go back to reference Le Douarin NM, Calloni GW, Dupin E. The stem cells of the neural crest. Cell Cycle. 2008;7(8):1013–9.PubMedCrossRef Le Douarin NM, Calloni GW, Dupin E. The stem cells of the neural crest. Cell Cycle. 2008;7(8):1013–9.PubMedCrossRef
41.
go back to reference Takashima Y, Era T, Nakao K, Kondo S, Kasuga M, Smith AG, et al. Neuroepithelial cells supply an initial transient wave of MSC differentiation. Cell. 2007;129(7):1377–88.PubMedCrossRef Takashima Y, Era T, Nakao K, Kondo S, Kasuga M, Smith AG, et al. Neuroepithelial cells supply an initial transient wave of MSC differentiation. Cell. 2007;129(7):1377–88.PubMedCrossRef
42.
43.
go back to reference Abkowitz JL, Robinson AE, Kale S, Long MW, Chen J. Mobilization of hematopoietic stem cells during homeostasis and after cytokine exposure. Blood. 2003;102(4):1249–53.PubMedCrossRef Abkowitz JL, Robinson AE, Kale S, Long MW, Chen J. Mobilization of hematopoietic stem cells during homeostasis and after cytokine exposure. Blood. 2003;102(4):1249–53.PubMedCrossRef
44.
go back to reference Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294(5548):1933–6.PubMedCrossRef Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294(5548):1933–6.PubMedCrossRef
45.
go back to reference Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452(7186):442–7.PubMedCrossRef Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452(7186):442–7.PubMedCrossRef
46.
go back to reference Lucas D, Battista M, Shi PA, Isola L, Frenette PS. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. 2008;3(4):364–6.PubMedCrossRef Lucas D, Battista M, Shi PA, Isola L, Frenette PS. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. 2008;3(4):364–6.PubMedCrossRef
47.
go back to reference Massberg S, Schaerli P, Knezevic-Maramica I, Kollnberger M, Tubo N, Moseman EA, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131(5):994–1008.PubMedCrossRef Massberg S, Schaerli P, Knezevic-Maramica I, Kollnberger M, Tubo N, Moseman EA, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131(5):994–1008.PubMedCrossRef
48.
go back to reference Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124(2):407–21.PubMedCrossRef Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124(2):407–21.PubMedCrossRef
49.
go back to reference Chai Y, Jiang X, Ito Y, Bringas Jr P, Han J, Rowitch DH, et al. Fate of the mammalian cranial neural crest during tooth and mandibular morphogenesis. Development. 2000;127(8):1671–9.PubMed Chai Y, Jiang X, Ito Y, Bringas Jr P, Han J, Rowitch DH, et al. Fate of the mammalian cranial neural crest during tooth and mandibular morphogenesis. Development. 2000;127(8):1671–9.PubMed
50.
go back to reference Jiang X, Iseki S, Maxson RE, Sucov HM, Morriss-Kay GM. Tissue origins and interactions in the mammalian skull vault. Dev Biol. 2002;241(1):106–16.PubMedCrossRef Jiang X, Iseki S, Maxson RE, Sucov HM, Morriss-Kay GM. Tissue origins and interactions in the mammalian skull vault. Dev Biol. 2002;241(1):106–16.PubMedCrossRef
51.
go back to reference Takeda S, Karsenty G. Molecular bases of the sympathetic regulation of bone mass. Bone. 2008;42(5):837–40.PubMedCrossRef Takeda S, Karsenty G. Molecular bases of the sympathetic regulation of bone mass. Bone. 2008;42(5):837–40.PubMedCrossRef
52.
go back to reference Adams GB, Martin RP, Alley IR, Chabner KT, Cohen KS, Calvi LM, et al. Therapeutic targeting of a stem cell niche. Nat Biotechnol. 2007;25(2):238–43.PubMedCrossRef Adams GB, Martin RP, Alley IR, Chabner KT, Cohen KS, Calvi LM, et al. Therapeutic targeting of a stem cell niche. Nat Biotechnol. 2007;25(2):238–43.PubMedCrossRef
53.
go back to reference • Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116(23):4815–28. The authors report that macrophages promote the retention of hematopoietic stem cells in the bone marrow. PubMedCrossRef • Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116(23):4815–28. The authors report that macrophages promote the retention of hematopoietic stem cells in the bone marrow. PubMedCrossRef
54.
go back to reference • Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208(2):251–60. The authors report that expression of the receptor for granulocyte colony-stimulating factor in monocytic cells is sufficient to trigger hematopoietic stem cell mobilization from the bone marrow to peripheral circulation. PubMedCrossRef • Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208(2):251–60. The authors report that expression of the receptor for granulocyte colony-stimulating factor in monocytic cells is sufficient to trigger hematopoietic stem cell mobilization from the bone marrow to peripheral circulation. PubMedCrossRef
55.
go back to reference • Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, Scheiermann C, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011. The authors report that bone marrow CD169 + macrophages increase the attraction of hematopoietic stem cells by mesenchymal stem cells in the bone marrow. • Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, Scheiermann C, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011. The authors report that bone marrow CD169 + macrophages increase the attraction of hematopoietic stem cells by mesenchymal stem cells in the bone marrow.
56.
go back to reference Mendez-Ferrer S, Frenette PS. Galpha(s) uncouples hematopoietic stem cell homing and mobilization. Cell Stem Cell. 2009;4(5):379–80.PubMedCrossRef Mendez-Ferrer S, Frenette PS. Galpha(s) uncouples hematopoietic stem cell homing and mobilization. Cell Stem Cell. 2009;4(5):379–80.PubMedCrossRef
57.
go back to reference •• Sanchez-Aguilera A, Lee YJ, Lo Celso C, Ferraro F, Brumme K, Mondal S, et al. Guanine nucleotide exchange factor Vav1 regulates perivascular homing and bone marrow retention of hematopoietic stem and progenitor cells. Proc Natl Acad Sci U S A. 2011;108(23):9607–12. Abnormal localization of Vav1-deficient hematopoietic stem and progenitor cells relative to bone marrow nestin + cells is associated with engraftment failure. PubMedCrossRef •• Sanchez-Aguilera A, Lee YJ, Lo Celso C, Ferraro F, Brumme K, Mondal S, et al. Guanine nucleotide exchange factor Vav1 regulates perivascular homing and bone marrow retention of hematopoietic stem and progenitor cells. Proc Natl Acad Sci U S A. 2011;108(23):9607–12. Abnormal localization of Vav1-deficient hematopoietic stem and progenitor cells relative to bone marrow nestin + cells is associated with engraftment failure. PubMedCrossRef
58.
go back to reference Yamazaki K, Allen TD. Ultrastructural morphometric study of efferent nerve terminals on murine bone marrow stromal cells, and the recognition of a novel anatomical unit: the “neuro-reticular complex”. Am J Anat. 1990;187(3):261–76.PubMedCrossRef Yamazaki K, Allen TD. Ultrastructural morphometric study of efferent nerve terminals on murine bone marrow stromal cells, and the recognition of a novel anatomical unit: the “neuro-reticular complex”. Am J Anat. 1990;187(3):261–76.PubMedCrossRef
59.
go back to reference •• Schajnovitz A, Itkin T, D'Uva G, Kalinkovich A, Golan K, Ludin A, et al. CXCL12 secretion by bone marrow stromal cells is dependent on cell contact and mediated by connexin-43 and connexin-45 gap junctions. Nat Immunol. 2011;12(5):391–8. Connexin-43- and −45–mediated electromechanical coupling among bone marrow stromal cells is reported here to be necessary for secretion of the chemokine CXCL12. PubMedCrossRef •• Schajnovitz A, Itkin T, D'Uva G, Kalinkovich A, Golan K, Ludin A, et al. CXCL12 secretion by bone marrow stromal cells is dependent on cell contact and mediated by connexin-43 and connexin-45 gap junctions. Nat Immunol. 2011;12(5):391–8. Connexin-43- and −45–mediated electromechanical coupling among bone marrow stromal cells is reported here to be necessary for secretion of the chemokine CXCL12. PubMedCrossRef
60.
go back to reference • Omatsu Y, Sugiyama T, Kohara H, Kondoh G, Fujii N, Kohno K, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33(3):387–99. The authors report that previously characterized bone marrow CXCL12-abundant reticular cells contain mesenchymal precursors that regulate hematopoietic progenitor cells. PubMedCrossRef • Omatsu Y, Sugiyama T, Kohara H, Kondoh G, Fujii N, Kohno K, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33(3):387–99. The authors report that previously characterized bone marrow CXCL12-abundant reticular cells contain mesenchymal precursors that regulate hematopoietic progenitor cells. PubMedCrossRef
Metadata
Title
Stem Cell Interactions in a Bone Marrow Niche
Authors
Joan Isern
Simón Méndez-Ferrer
Publication date
01-12-2011
Publisher
Current Science Inc.
Published in
Current Osteoporosis Reports / Issue 4/2011
Print ISSN: 1544-1873
Electronic ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-011-0075-y

Other articles of this Issue 4/2011

Current Osteoporosis Reports 4/2011 Go to the issue

Pediatrics and Skeletal Development (Craig Langman and Maria Luisa Bianchi, Section Editors)

Bone Development in the Fetus and Neonate: Role of the Calciotropic Hormones

Epidemiology and Pathophysiology (Mone Zaidi and Jeffrey I. Mechanick, Section Editors)

Nutrition, Bone, and Aging: An Integrative Physiology Approach

Pediatrics and Skeletal Development (Craig Langman and Maria Luisa Bianchi, Section Editors)

Bone Development: Overview of Bone Cells and Signaling

Epidemiology and Pathophysiology (Mone Zaidi and Jeffrey I. Mechanick, Section Editors)

Anti-Cancer Actions of Denosumab

Epidemiology and Pathophysiology (Mone Zaidi and Jeffrey I. Mechanick, Section Editors)

Mechanical Loading: Bone Remodeling and Cartilage Maintenance

Pediatrics and Skeletal Development (Craig Langman and Maria Luisa Bianchi, Section Editors)

Quantitative Computed Tomography and Computed Tomography in Children