Skip to main content
Erschienen in: International Journal of Hematology 6/2014

01.06.2014 | Progress in Hematology

Regulation of hematopoiesis in endosteal microenvironments

verfasst von: Noboru Asada, Yoshio Katayama

Erschienen in: International Journal of Hematology | Ausgabe 6/2014

Einloggen, um Zugang zu erhalten

Abstract

After birth, the hematopoietic system develops along with bone formation in mammals. Osteolineage cells are derived from mesenchymal progenitor cells, and differentiate into several types of bone-forming cells. Of the various types of cell constituents in bone marrow, osteolineage cells have been shown to play important roles in hematopoiesis. Early studies have identified osteoblasts as a hematopoietic stem cell niche component. Since that time, the role of endosteal microenvironment as a critical regulator of hematopoietic stem/progenitor cell (HSC/HPC) behavior has been appreciated particularly under stress conditions, such as cytokine-induced HSC/HPC mobilization, homing/engraftment after bone marrow transplantation, and disease models of leukemia/myelodysplasia. Recent studies revealed that the most differentiated osteolineage cells, i.e., osteocytes, play important roles in the regulation of hematopoiesis. In this review, we provide an overview of recent advances in knowledge of regulatory hematopoietic mechanisms in the endosteal area.
Literatur
1.
Zurück zum Zitat Omatsu Y, Sugiyama T, Kohara H, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33(3):387–99.PubMedCrossRef Omatsu Y, Sugiyama T, Kohara H, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33(3):387–99.PubMedCrossRef
2.
Zurück zum Zitat Mendez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34.PubMedCentralPubMedCrossRef Mendez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34.PubMedCentralPubMedCrossRef
3.
Zurück zum Zitat Greenbaum A, Hsu YM, Day RB, et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature. 2013;495(7440):227–30.PubMedCentralPubMedCrossRef Greenbaum A, Hsu YM, Day RB, et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature. 2013;495(7440):227–30.PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495(7440):231–5.PubMedCentralPubMedCrossRef Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495(7440):231–5.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Kunisaki Y, Bruns I, Scheiermann C, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502(7473):637–43.PubMedCrossRef Kunisaki Y, Bruns I, Scheiermann C, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502(7473):637–43.PubMedCrossRef
6.
Zurück zum Zitat Komori T, Yagi H, Nomura S, et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997;89(5):755–64.PubMedCrossRef Komori T, Yagi H, Nomura S, et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997;89(5):755–64.PubMedCrossRef
7.
Zurück zum Zitat Nakashima K, Zhou X, Kunkel G, et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 2002;108(1):17–29.PubMedCrossRef Nakashima K, Zhou X, Kunkel G, et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 2002;108(1):17–29.PubMedCrossRef
8.
Zurück zum Zitat Nakashima K, de Crombrugghe B. Transcriptional mechanisms in osteoblast differentiation and bone formation. Trends Genet. 2003;19(8):458–66.PubMedCrossRef Nakashima K, de Crombrugghe B. Transcriptional mechanisms in osteoblast differentiation and bone formation. Trends Genet. 2003;19(8):458–66.PubMedCrossRef
9.
Zurück zum Zitat Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21(2):115–37.PubMed Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21(2):115–37.PubMed
10.
Zurück zum Zitat Jilka RL, Weinstein RS, Bellido T, et al. Osteoblast programmed cell death (apoptosis): modulation by growth factors and cytokines. J Bone Miner Res. 1998;13(5):793–802.PubMedCrossRef Jilka RL, Weinstein RS, Bellido T, et al. Osteoblast programmed cell death (apoptosis): modulation by growth factors and cytokines. J Bone Miner Res. 1998;13(5):793–802.PubMedCrossRef
12.
Zurück zum Zitat Fulzele K, Krause DS, Panaroni C, et al. Myelopoiesis is regulated by osteocytes through Gsalpha-dependent signaling. Blood. 2013;121(6):930–9.PubMedCentralPubMedCrossRef Fulzele K, Krause DS, Panaroni C, et al. Myelopoiesis is regulated by osteocytes through Gsalpha-dependent signaling. Blood. 2013;121(6):930–9.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Asada N, Katayama Y, Sato M, et al. Matrix-embedded osteocytes regulate mobilization of hematopoietic stem/progenitor cells. Cell Stem Cell. 2013;12(6):737–47.PubMedCrossRef Asada N, Katayama Y, Sato M, et al. Matrix-embedded osteocytes regulate mobilization of hematopoietic stem/progenitor cells. Cell Stem Cell. 2013;12(6):737–47.PubMedCrossRef
14.
Zurück zum Zitat Sato M, Asada N, Kawano Y, et al. Osteocytes regulate primary lymphoid organs and fat metabolism. Cell Metab. 2013;18(5):749–58.PubMedCrossRef Sato M, Asada N, Kawano Y, et al. Osteocytes regulate primary lymphoid organs and fat metabolism. Cell Metab. 2013;18(5):749–58.PubMedCrossRef
15.
Zurück zum Zitat Taichman RS, Emerson SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med. 1994;179(5):1677–82.PubMedCrossRef Taichman RS, Emerson SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med. 1994;179(5):1677–82.PubMedCrossRef
16.
Zurück zum Zitat Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841–6.PubMedCrossRef Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841–6.PubMedCrossRef
17.
Zurück zum Zitat Zhang J, Niu C, Ye L, 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, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425(6960):836–41.PubMedCrossRef
18.
Zurück zum Zitat Lymperi S, Horwood N, Marley S, et al. Strontium can increase some osteoblasts without increasing hematopoietic stem cells. Blood. 2008;111(3):1173–81.PubMedCrossRef Lymperi S, Horwood N, Marley S, et al. Strontium can increase some osteoblasts without increasing hematopoietic stem cells. Blood. 2008;111(3):1173–81.PubMedCrossRef
19.
Zurück zum Zitat Nakamura Y, Arai F, Iwasaki H, et al. Isolation and characterization of endosteal niche cell populations that regulate hematopoietic stem cells. Blood. 2010;116(9):1422–32.PubMedCrossRef Nakamura Y, Arai F, Iwasaki H, et al. Isolation and characterization of endosteal niche cell populations that regulate hematopoietic stem cells. Blood. 2010;116(9):1422–32.PubMedCrossRef
20.
Zurück zum Zitat Arai F, Hirao A, Ohmura M, 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, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149–61.PubMedCrossRef
21.
Zurück zum Zitat Yoshihara H, Arai F, Hosokawa K, et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007;1(6):685–97.PubMedCrossRef Yoshihara H, Arai F, Hosokawa K, et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007;1(6):685–97.PubMedCrossRef
22.
Zurück zum Zitat Qian H, Buza-Vidas N, Hyland CD, et al. Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells. Cell Stem Cell. 2007;1(6):671–84.PubMedCrossRef Qian H, Buza-Vidas N, Hyland CD, et al. Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells. Cell Stem Cell. 2007;1(6):671–84.PubMedCrossRef
23.
Zurück zum Zitat Sugimura R, He XC, Venkatraman A, et al. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell. 2012;150(2):351–65.PubMedCrossRef Sugimura R, He XC, Venkatraman A, et al. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell. 2012;150(2):351–65.PubMedCrossRef
24.
Zurück zum Zitat Adams GB, Chabner KT, Alley IR, 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, et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439(7076):599–603.PubMedCrossRef
25.
Zurück zum Zitat Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12(6):657–64.PubMedCrossRef Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12(6):657–64.PubMedCrossRef
26.
Zurück zum Zitat Mansour A, Abou-Ezzi G, Sitnicka E, et al. Osteoclasts promote the formation of hematopoietic stem cell niches in the bone marrow. J Exp Med. 2012;209(3):537–49.PubMedCentralPubMedCrossRef Mansour A, Abou-Ezzi G, Sitnicka E, et al. Osteoclasts promote the formation of hematopoietic stem cell niches in the bone marrow. J Exp Med. 2012;209(3):537–49.PubMedCentralPubMedCrossRef
27.
Zurück zum Zitat Miyamoto K, Yoshida S, Kawasumi M, et al. Osteoclasts are dispensable for hematopoietic stem cell maintenance and mobilization. J Exp Med. 2011;208(11):2175–81.PubMedCentralPubMedCrossRef Miyamoto K, Yoshida S, Kawasumi M, et al. Osteoclasts are dispensable for hematopoietic stem cell maintenance and mobilization. J Exp Med. 2011;208(11):2175–81.PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Tatsumi S, Ishii K, Amizuka N, et al. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 2007;5(6):464–75.PubMedCrossRef Tatsumi S, Ishii K, Amizuka N, et al. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 2007;5(6):464–75.PubMedCrossRef
29.
Zurück zum Zitat Marotti G, Ferretti M, Muglia MA, et al. A quantitative evaluation of osteoblast-osteocyte relationships on growing endosteal surface of rabbit tibiae. Bone. 1992;13(5):363–8.PubMedCrossRef Marotti G, Ferretti M, Muglia MA, et al. A quantitative evaluation of osteoblast-osteocyte relationships on growing endosteal surface of rabbit tibiae. Bone. 1992;13(5):363–8.PubMedCrossRef
30.
Zurück zum Zitat Yellowley CE, Li Z, Zhou Z, et al. Functional gap junctions between osteocytic and osteoblastic cells. J Bone Miner Res. 2000;15(2):209–17.PubMedCrossRef Yellowley CE, Li Z, Zhou Z, et al. Functional gap junctions between osteocytic and osteoblastic cells. J Bone Miner Res. 2000;15(2):209–17.PubMedCrossRef
31.
Zurück zum Zitat Crucian B, Sams C. Immune system dysregulation during spaceflight: clinical risk for exploration-class missions. J Leukoc Biol. 2009;86(5):1017–8.PubMedCrossRef Crucian B, Sams C. Immune system dysregulation during spaceflight: clinical risk for exploration-class missions. J Leukoc Biol. 2009;86(5):1017–8.PubMedCrossRef
32.
Zurück zum Zitat Gueguinou N, Huin-Schohn C, Bascove M, et al. Could spaceflight-associated immune system weakening preclude the expansion of human presence beyond Earth’s orbit? J Leukoc Biol. 2009;86(5):1027–38.PubMedCrossRef Gueguinou N, Huin-Schohn C, Bascove M, et al. Could spaceflight-associated immune system weakening preclude the expansion of human presence beyond Earth’s orbit? J Leukoc Biol. 2009;86(5):1027–38.PubMedCrossRef
33.
Zurück zum Zitat Christopher MJ, Link DC. Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation. J Bone Miner Res. 2008;23(11):1765–74.PubMedCentralPubMedCrossRef Christopher MJ, Link DC. Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation. J Bone Miner Res. 2008;23(11):1765–74.PubMedCentralPubMedCrossRef
34.
Zurück zum Zitat Katayama Y, Battista M, Kao WM, 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, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124(2):407–21.PubMedCrossRef
35.
Zurück zum Zitat Kawamori Y, Katayama Y, Asada N, et al. Role for vitamin D receptor in the neuronal control of the hematopoietic stem cell niche. Blood. 2010;116(25):5528–35.PubMedCrossRef Kawamori Y, Katayama Y, Asada N, et al. Role for vitamin D receptor in the neuronal control of the hematopoietic stem cell niche. Blood. 2010;116(25):5528–35.PubMedCrossRef
36.
Zurück zum Zitat 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
37.
Zurück zum Zitat Chow A, Lucas D, Hidalgo A, 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;208(2):261–71.PubMedCentralPubMedCrossRef Chow A, Lucas D, Hidalgo A, 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;208(2):261–71.PubMedCentralPubMedCrossRef
38.
Zurück zum Zitat Christopher MJ, Rao M, Liu F, et al. 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.PubMedCentralPubMedCrossRef Christopher MJ, Rao M, Liu F, et al. 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.PubMedCentralPubMedCrossRef
39.
Zurück zum Zitat Winkler IG, Sims NA, Pettit AR, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116(23):4815–28.PubMedCrossRef Winkler IG, Sims NA, Pettit AR, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116(23):4815–28.PubMedCrossRef
40.
Zurück zum Zitat Taylor AF, Saunders MM, Shingle DL, et al. Mechanically stimulated osteocytes regulate osteoblastic activity via gap junctions. Am J Physiol Cell Physiol. 2007;292(1):C545–52.PubMedCrossRef Taylor AF, Saunders MM, Shingle DL, et al. Mechanically stimulated osteocytes regulate osteoblastic activity via gap junctions. Am J Physiol Cell Physiol. 2007;292(1):C545–52.PubMedCrossRef
41.
Zurück zum Zitat Chang MK, Raggatt LJ, Alexander KA, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol. 2008;181(2):1232–44.PubMedCrossRef Chang MK, Raggatt LJ, Alexander KA, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol. 2008;181(2):1232–44.PubMedCrossRef
42.
Zurück zum Zitat Kamioka H, Honjo T, Takano-Yamamoto T. A three-dimensional distribution of osteocyte processes revealed by the combination of confocal laser scanning microscopy and differential interference contrast microscopy. Bone. 2001;28(2):145–9.PubMedCrossRef Kamioka H, Honjo T, Takano-Yamamoto T. A three-dimensional distribution of osteocyte processes revealed by the combination of confocal laser scanning microscopy and differential interference contrast microscopy. Bone. 2001;28(2):145–9.PubMedCrossRef
43.
Zurück zum Zitat Gonzalez-Nieto D, Li L, Kohler A, et al. Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors. Blood. 2012;119(22):5144–54.PubMedCentralPubMedCrossRef Gonzalez-Nieto D, Li L, Kohler A, et al. Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors. Blood. 2012;119(22):5144–54.PubMedCentralPubMedCrossRef
44.
Zurück zum Zitat Ishikawa F, Yoshida S, Saito Y, et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007;25(11):1315–21.PubMedCrossRef Ishikawa F, Yoshida S, Saito Y, et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007;25(11):1315–21.PubMedCrossRef
45.
Zurück zum Zitat Ishikawa F. Modeling normal and malignant human hematopoiesis in vivo through newborn NSG xenotransplantation. Int J Hematol. 2013;98(6):634–40.PubMedCrossRef Ishikawa F. Modeling normal and malignant human hematopoiesis in vivo through newborn NSG xenotransplantation. Int J Hematol. 2013;98(6):634–40.PubMedCrossRef
46.
Zurück zum Zitat Schepers K, Pietras EM, Reynaud D, et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell. 2013;13(3):285–99.PubMedCrossRef Schepers K, Pietras EM, Reynaud D, et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell. 2013;13(3):285–99.PubMedCrossRef
47.
Zurück zum Zitat Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7.PubMedCentralPubMedCrossRef Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7.PubMedCentralPubMedCrossRef
48.
Zurück zum Zitat Raaijmakers MH. Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol. 2012;95(1):17–25.PubMedCrossRef Raaijmakers MH. Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol. 2012;95(1):17–25.PubMedCrossRef
49.
Zurück zum Zitat Kode A, Manavalan JS, Mosialou I, et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature. 2014;506(7487):240–4.PubMedCrossRef Kode A, Manavalan JS, Mosialou I, et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature. 2014;506(7487):240–4.PubMedCrossRef
Metadaten
Titel
Regulation of hematopoiesis in endosteal microenvironments
verfasst von
Noboru Asada
Yoshio Katayama
Publikationsdatum
01.06.2014
Verlag
Springer Japan
Erschienen in
International Journal of Hematology / Ausgabe 6/2014
Print ISSN: 0925-5710
Elektronische ISSN: 1865-3774
DOI
https://doi.org/10.1007/s12185-014-1583-1

Weitere Artikel der Ausgabe 6/2014

International Journal of Hematology 6/2014 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.