清晨好,您是今天最早来到科研通的研友!由于当前在线用户较少,发布求助请尽量完整的填写文献信息,科研通机器人24小时在线,伴您科研之路漫漫前行!

“Caught in the net”: the extracellular matrix of the bone marrow in normal hematopoiesis and leukemia

造血 骨髓 白血病 细胞外基质 癌症研究 生物 免疫学 细胞生物学 干细胞
作者
Costanza Zanetti,Daniela Krause
出处
期刊:Experimental Hematology [Elsevier]
卷期号:89: 13-25 被引量:21
标识
DOI:10.1016/j.exphem.2020.07.010
摘要

•The ECM supports the HSC-supportive function of the bone marrow microenvironment.microenvironment•The ECM provides a scaffold and serves as a source of growth factors and proteases.•ECM proteins influence HSC function and the course of a leukemia.•Interactions between ECM proteins and HSCs or LSCs can be targeted therapeutically.•Novel technologies aid the understanding of the ECM in the BMM. The influence of the bone marrow microenvironment on normal hematopoiesis, but also leukemia, has largely been accepted. However, the focus has been predominantly on the role of various cell types or cytokines maintaining hematopoietic stem cells or protecting leukemia stem cells from different therapies. A frequently overlooked component of the bone marrow microenvironment is the extracellular matrix, which not only provides a mechanical scaffold, but also serves as a source of growth factors. We discuss here how extracellular matrix proteins directly or indirectly modulate hematopoietic stem cell physiology and influence leukemia progression. It is hoped that existing and future studies on this topic may propel forward the possibility of augmenting normal hematopoiesis and improving therapies for leukemia, for instance, by targeting of the extracellular matrix in the bone marrow. The influence of the bone marrow microenvironment on normal hematopoiesis, but also leukemia, has largely been accepted. However, the focus has been predominantly on the role of various cell types or cytokines maintaining hematopoietic stem cells or protecting leukemia stem cells from different therapies. A frequently overlooked component of the bone marrow microenvironment is the extracellular matrix, which not only provides a mechanical scaffold, but also serves as a source of growth factors. We discuss here how extracellular matrix proteins directly or indirectly modulate hematopoietic stem cell physiology and influence leukemia progression. It is hoped that existing and future studies on this topic may propel forward the possibility of augmenting normal hematopoiesis and improving therapies for leukemia, for instance, by targeting of the extracellular matrix in the bone marrow. After many decades of research, we can now postulate that hematopoiesis is a process in which hematopoietic stem cells (HSCs) are responsible for the production of mature differentiated blood cells via intermediate progenitor cells, but at the same time HSCs self-renew to maintain their own pool. Both processes occur through the regulation of key genes and defined genetic programs [1Riddell J Gazit R Garrison BS et al.Reprogramming committed murine blood cells to induced hematopoietic stem cells with defined factors.Cell. 2014; 157: 549-564Abstract Full Text Full Text PDF PubMed Scopus (199) Google Scholar]. To maintain a steady state, most HSCs remain in quiescence, which has been linked to long-term reconstitution capacity. However, HSCs rapidly switch to active proliferation under stress conditions [2Wilson A Laurenti E Oser G et al.Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair.Cell. 2008; 135: 1118-1129Abstract Full Text Full Text PDF PubMed Scopus (1130) Google Scholar] and during differentiation [3Passegue E Wagers AJ Giuriato S Anderson WC Weissman IL Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates.J Exp Med. 2005; 202: 1599-1611Crossref PubMed Scopus (433) Google Scholar]. Overall, studies on hematopoiesis and HSCs have also provided the basis for the “cancer stem cell” theory, that is, cancer stem cells represent a small, functionally distinct subset of tumor cells, which have the ability to reconstitute the tumor because of their ability to self-renew and differentiate into the various cancerous cells. The bulk tumor cells, however, do not have this ability. Leukemias are blood cancers characterized by abnormalities of leukocytes. The concept of leukemia stem cells (LSCs) was introduced when it was found that CD34+CD38− cells from acute myeloid leukemia (AML) patients have leukemia-initiating capacity upon transplantation into immunosuppressed mice [4Bonnet D Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.Nat Med. 1997; 3: 730-737Crossref PubMed Scopus (4838) Google Scholar]. Therefore, a leukemia was proven to be composed of heterogeneous cells, with only a subset of them having self-renewal capacity similar to that of normal HSCs. Complexity has been added to this question by the discovery of the contributions of the bone marrow microenvironment (BMM) or niche to the development, progression, and therapy resistance of leukemia. The term stem cell niche, referring to a specific location and entity extrinsically orchestrating and controlling the self-renewal and differentiation capacity of HSCs, was coined in 1978 [5Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell.Blood Cells. 1978; 4: 7-25PubMed Google Scholar]. The BMM has largely been recognized as a unique structure providing a complex cellular, chemical, and mechanical microenvironment [6Morrison SJ Scadden DT. The bone marrow niche for haematopoietic stem cells.Nature. 2014; 505: 327-334Crossref PubMed Scopus (1100) Google Scholar, 7Birbrair A Frenette PS. Niche heterogeneity in the bone marrow.Ann NY Acad Sci. 2016; 1370: 82-96Crossref PubMed Scopus (124) Google Scholar, 8Crane GM Jeffery E Morrison SJ Adult haematopoietic stem cell niches.Nat Rev Immunol. 2017; 17: 573-590Crossref PubMed Scopus (197) Google Scholar]. At first, in the early years of the 21st century, the BMM was somewhat formally divided into endosteal [9Calvi LM Adams GB Weibrecht KW et al.Osteoblastic cells regulate the haematopoietic stem cell niche.Nature. 2003; 425: 841-846Crossref PubMed Scopus (2581) Google Scholar, 10Zhang J Niu C Ye L et al.Identification of the haematopoietic stem cell niche and control of the niche size.Nature. 2003; 425: 836-841Crossref PubMed Scopus (2211) Google Scholar, 11Taichman R Reilly M Verma R Ehrenman K Emerson S Hepatocyte growth factor is secreted by osteoblasts and cooperatively permits the survival of haematopoietic progenitors.Br J Haematol. 2001; 112: 438-448Crossref PubMed Scopus (0) Google Scholar, 12Nilsson SK Johnston HM Coverdale JA Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches.Blood. 2001; 97: 2293-2299Crossref PubMed Scopus (437) Google Scholar] and vascular [13Kiel MJ Yilmaz OH Iwashita T Yilmaz OH Terhorst C Morrison SJ SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells.Cell. 2005; 121: 1109-1121Abstract Full Text Full Text PDF PubMed Scopus (2148) Google Scholar, 14Kusumbe AP Ramasamy SK Adams RH Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone.Nature. 2014; 507: 323-328Crossref PubMed Scopus (565) Google Scholar, 15Kunisaki Y Bruns I Scheiermann C et al.Arteriolar niches maintain haematopoietic stem cell quiescence.Nature. 2013; 502: 637-643Crossref PubMed Scopus (591) Google Scholar] niches. Since then, via the generation of a myriad of transgenic mouse models [16Ding L Morrison SJ Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches.Nature. 2013; 495: 231-235Crossref PubMed Scopus (622) Google Scholar, 17Mendez-Ferrer S Michurina TV Ferraro F et al.Mesenchymal and haematopoietic stem cells form a unique bone marrow niche.Nature. 2010; 466: 829-834Crossref PubMed Scopus (1997) Google Scholar, 18Mizoguchi T Pinho S Ahmed J et al.Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development.Dev Cell. 2014; 29: 340-349Abstract Full Text Full Text PDF PubMed Scopus (196) Google Scholar, 19Ding L Saunders TL Enikolopov G Morrison SJ Endothelial and perivascular cells maintain haematopoietic stem cells.Nature. 2012; 481: 457-462Crossref PubMed Scopus (980) Google Scholar], sophisticated imaging technologies [17Mendez-Ferrer S Michurina TV Ferraro F et al.Mesenchymal and haematopoietic stem cells form a unique bone marrow niche.Nature. 2010; 466: 829-834Crossref PubMed Scopus (1997) Google Scholar,20Itkin T Gur-Cohen S Spencer JA et al.Distinct bone marrow blood vessels differentially regulate haematopoiesis.Nature. 2016; 532: 323-328Crossref PubMed Google Scholar, 21Hawkins ED Duarte D Akinduro O et al.T-cell acute leukaemia exhibits dynamic interactions with bone marrow microenvironments.Nature. 2016; 538: 518-522Crossref PubMed Scopus (89) Google Scholar, 22Duarte D Hawkins ED Akinduro O et al.Inhibition of endosteal vascular niche remodeling rescues hematopoietic stem cell loss in AML.Cell Stem Cell. 2018; 22: 64-77.e66Abstract Full Text Full Text PDF PubMed Scopus (71) Google Scholar, 23Lo Celso C Fleming HE Wu JW et al.Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche.Nature. 2009; 457: 92-96Crossref PubMed Scopus (591) Google Scholar], and single-cell studies [24Tikhonova AN Dolgalev I Hu H et al.The bone marrow microenvironment at single-cell resolution.Nature. 2019; 569: 222-228Crossref PubMed Scopus (106) Google Scholar, 25Baccin C Al-Sabah J Velten L et al.Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization.Nat Cell Biol. 2020; 22: 38-48Crossref PubMed Scopus (4) Google Scholar, 26Wolock SL Krishnan I Tenen DE et al.Mapping distinct bone marrow niche populations and their differentiation paths.Cell Rep. 2019; 28 (302–311.e305)Abstract Full Text Full Text PDF PubMed Scopus (17) Google Scholar], the field has come to the understanding that the BMM is a continuum in which specific locations may be associated with specific features of HSCs [9Calvi LM Adams GB Weibrecht KW et al.Osteoblastic cells regulate the haematopoietic stem cell niche.Nature. 2003; 425: 841-846Crossref PubMed Scopus (2581) Google Scholar,15Kunisaki Y Bruns I Scheiermann C et al.Arteriolar niches maintain haematopoietic stem cell quiescence.Nature. 2013; 502: 637-643Crossref PubMed Scopus (591) Google Scholar,20Itkin T Gur-Cohen S Spencer JA et al.Distinct bone marrow blood vessels differentially regulate haematopoiesis.Nature. 2016; 532: 323-328Crossref PubMed Google Scholar,27Lo Celso C Wu JW Lin CP In vivo imaging of hematopoietic stem cells and their microenvironment.J Biophotonics. 2009; 2: 619-631Crossref PubMed Scopus (0) Google Scholar, 28Sugiyama 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: 977-988Abstract Full Text Full Text PDF PubMed Scopus (1380) Google Scholar, 29Cordeiro Gomes A Hara T Lim VY et al.Hematopoietic stem cell niches produce lineage-instructive signals to control multipotent progenitor differentiation.Immunity. 2016; 45: 1219-1231Abstract Full Text Full Text PDF PubMed Scopus (74) Google Scholar, 30Tokoyoda K Egawa T Sugiyama T Choi BI Nagasawa T Cellular niches controlling B lymphocyte behavior within bone marrow during development.Immunity. 2004; 20: 707-718Abstract Full Text Full Text PDF PubMed Scopus (536) Google Scholar, 31Zhao M Perry JM Marshall H et al.Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells.Nat Med. 2014; 20: 1321-1326Crossref PubMed Scopus (242) Google Scholar] and LSCs [32Bowers M Zhang B Ho Y Agarwal P Chen CC Bhatia R Osteoblast ablation reduces normal long-term hematopoietic stem cell self-renewal but accelerates leukemia development.Blood. 2015; 125: 2678-2688Crossref PubMed Scopus (55) Google Scholar,33Ishikawa 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: 1315-1321Crossref PubMed Scopus (612) Google Scholar]. In general, it is understood that the BMM is a complex entity composed of several cellular and acellular elements such as mesenchymal stromal cells (MSCs), endothelial cells (ECs), osteoblasts, osteoclasts, osteocytes, adipocytes, C–X–C motif chemokine ligand (CXCL)12-abundant reticular (CAR) cells, fibroblasts, sympathetic neurons, megakaryocytes, and macrophages, as well as chemical and mechanical factors and the extracellular matrix (ECM) [34Mendez-Ferrer S Bonnet D Steensma DP et al.Bone marrow niches in haematological malignancies.Nat Rev Cancer. 2020; 20: 285-298Crossref PubMed Scopus (2) Google Scholar]. In addition, a specific pH, an oxygen gradient, cytokines, and mechanical factors, most of which are reviewed elsewhere [35Schepers K Campbell TB Passegue E Normal and leukemic stem cell niches: insights and therapeutic opportunities.Cell Stem Cell. 2015; 16: 254-267Abstract Full Text Full Text PDF PubMed Scopus (196) Google Scholar], contribute to the BMM's unique features (Figure 1). This review, however, is focused on the structure and biochemical properties of the proteins in the ECM of the BMM, as well as their functions in normal and malignant hematopoiesis. In general, the ECM is a three-dimensional, complex and highly dynamic noncellular network composed of (1) proteoglycans; (2) fibrous proteins such as collagens, fibronectins, elastins, tenascins, vitronectin, and laminins; and (3) glycosaminoglycans, such as hyaluronic acid, chondroitin sulfate, heparan sulfate, keratan sulfate, dermatan sulfate, and heparin; and (4) matricellular proteins such as osteocalcin and periostin, which assist in the linkage between other ECM proteins and cellular receptors [36Coutu DL Kokkaliaris KD Kunz L Schroeder T Three-dimensional map of nonhematopoietic bone and bone-marrow cells and molecules.Nat Biotechnol. 2017; 35: 1202-1210Crossref PubMed Scopus (28) Google Scholar,37Klamer S Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment.Cell Adh Migr. 2014; 8: 563-577Crossref PubMed Scopus (38) Google Scholar]. The most abundant proteins in the ECM of the BMM are fibronectin, collagens I, II, III, IV, and X, laminin, tenascin, thrombospondin, and elastin. Soluble or membrane-bound glycoproteins of the sialomucin family, such as platelet–selectin glycoprotein ligand (PSGL1/CD162) and intercellular adhesion molecule 1 (ICAM1; CD54), are also associated with the ECM [37Klamer S Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment.Cell Adh Migr. 2014; 8: 563-577Crossref PubMed Scopus (38) Google Scholar]. The ECM regulates structural scaffolding and, here in particular, the stiffness and deformability of tissues and tissue homeostasis. It also serves as a major source of proteases and growth factors [38Hynes RO. The extracellular matrix: not just pretty fibrils.Science. 2009; 326: 1216-1219Crossref PubMed Scopus (1749) Google Scholar]. Possibly consistent with the early discovery that localization within the BM cavity may lead to differences in the proliferation rate of colony-forming units [39Lord BI Testa NG Hendry JH The relative spatial distributions of CFUs and CFUc in the normal mouse femur.Blood. 1975; 46: 65-72Crossref PubMed Google Scholar], the distribution of ECM components in the BMM has been reported to be spatially organized [40Nilsson SK Debatis ME Dooner MS Madri JA Quesenberry PJ Becker PS Immunofluorescence characterization of key extracellular matrix proteins in murine bone marrow in situ.J Histochem Cytochem. 1998; 46: 371-377Crossref PubMed Google Scholar]. In particular, collagens I and III, osteocalcin, vitronectin, osteopontin, and periostin were found predominantly in the bone matrix. Collagen IV and laminin were localized largely in the vascular basement membrane and the bone marrow parenchyma, while fibronectin was found only in the latter. The heparan sulfate proteoglycan perlecan was found in the bone matrix and arteriolar basement membrane [36Coutu DL Kokkaliaris KD Kunz L Schroeder T Three-dimensional map of nonhematopoietic bone and bone-marrow cells and molecules.Nat Biotechnol. 2017; 35: 1202-1210Crossref PubMed Scopus (28) Google Scholar]. Hyaluronan (HA) was highly expressed in the endothelium of blood vessels in the metaphysis, where it may be involved in the homing of hematopoietic stem and progenitor cells (HSPCs) [41Ellis SL Grassinger J Jones A et al.The relationship between bone, hemopoietic stem cells, and vasculature.Blood. 2011; 118: 1516-1524Crossref PubMed Scopus (103) Google Scholar]. ECM composition and remodeling vary from tissue to tissue, leading to biochemical and biophysical properties that specifically reflect each environment [42Mouw JK Ou G Weaver VM Extracellular matrix assembly: a multiscale deconstruction.Nat Rev Mol Cell Biol. 2014; 15: 771-785Crossref PubMed Scopus (468) Google Scholar]. Deregulation of components of the ECM can contribute to several pathological conditions such as cardiomyopathy [43Pauschinger M Knopf D Petschauer S et al.Dilated cardiomyopathy is associated with significant changes in collagen type I/III ratio.Circulation. 1999; 99: 2750-2756Crossref PubMed Google Scholar], fibrosis [44Walraven M Hinz B Therapeutic approaches to control tissue repair and fibrosis: extracellular matrix as a game changer.Matrix Biol. 2018; 71-72: 205-224Crossref PubMed Scopus (59) Google Scholar,45Bhattacharyya S Tamaki Z Wang W et al.FibronectinEDA promotes chronic cutaneous fibrosis through Toll-like receptor signaling.Sci Transl Med. 2014; 6 (232ra250)Crossref PubMed Scopus (81) Google Scholar], and cancers [46Butcher DT Alliston T Weaver VM A tense situation: forcing tumour progression.Nat Rev Cancer. 2009; 9: 108-122Crossref PubMed Scopus (1051) Google Scholar,47Kessenbrock K Plaks V Werb Z Matrix metalloproteinases: regulators of the tumor microenvironment.Cell. 2010; 141: 52-67Abstract Full Text Full Text PDF PubMed Scopus (2715) Google Scholar]. In particular, mutations in the COL1A1 and COL1A2 genes, encoding the two α chains of type I collagen, cause autosomal dominant osteogenesis imperfecta. This results in abnormal synthesis and secretion of collagen type I, leading to bone fragility and deformities in patients [48Pollitt R McMahon R Nunn J et al.Mutation analysis of COL1A1 and COL1A2 in patients diagnosed with osteogenesis imperfecta type I–IV.Hum Mutat. 2006; 27: 716Crossref PubMed Google Scholar]. In addition, mutations in the COL7A1 and COL17A1 genes are responsible for a dystrophic form of epidermolysis bullosa [49Pasmooij AM Garcia M Escamez MJ et al.Revertant mosaicism due to a second-site mutation in COL7A1 in a patient with recessive dystrophic epidermolysis bullosa.J Invest Dermatol. 2010; 130: 2407-2411Abstract Full Text Full Text PDF PubMed Scopus (42) Google Scholar], and mutations in the COL5A1 or COL5A2 genes give rise to classic Ehlers–Danlos syndrome, a rare autosomal dominant connective tissue disorder [50Ritelli M Dordoni C Venturini M et al.Clinical and molecular characterization of 40 patients with classic Ehlers–Danlos syndrome: identification of 18 COL5A1 and 2 COL5A2 novel mutations.Orphanet J Rare Dis. 2013; 8: 58Crossref PubMed Scopus (43) Google Scholar]. ECM proteins are important constituents of the BM milieu, where they are involved in adhesion, binding of cytokines, and the general compartmentalization of the BM [51Klein G. The extracellular matrix of the hematopoietic microenvironment.Experientia. 1995; 51: 914-926Crossref PubMed Scopus (0) Google Scholar] (Figure 2). The fact that ECM proteins can be extracted and purified from different animal sources and used widely for the coating of cell culture wells has formed the basis for studies of the interactions of HSCs with the ECM. In addition, novel technologies, as mentioned below, have provided insight into the biochemical-functional properties of the ECM. Changes in the cellular microenvironment, in general, are sensed by cell–ECM interactions and are mediated mainly by integrins [52Kechagia JZ Ivaska J Roca-Cusachs P Integrins as biomechanical sensors of the microenvironment.Nat Rev Mol Cell Biol. 2019; 20: 457-473Crossref PubMed Scopus (71) Google Scholar] as well as discoidin domain receptors (DDRs) [53Leitinger B Hohenester E. Mammalian collagen receptors.Matrix Biol. 2007; 26: 146-155Crossref PubMed Scopus (253) Google Scholar], leukocyte-associated Ig-like receptor (LAIR) 1 [54Meyaard L. The inhibitory collagen receptor LAIR-1 (CD305).J Leukoc Biol. 2008; 83: 799-803Crossref PubMed Scopus (156) Google Scholar], and syndecans [55Morgan MR Humphries MJ Bass MD Synergistic control of cell adhesion by integrins and syndecans.Nat Rev Mol Cell Biol. 2007; 8: 957-969Crossref PubMed Scopus (385) Google Scholar], regulating cell migration, adhesion, survival, shape, and differentiation [56Bissell MJ Hall HG Parry G How does the extracellular matrix direct gene expression?.J Theor Biol. 1982; 99: 31-68Crossref PubMed Scopus (980) Google Scholar]. Integrins are heterodimers, consisting of an α subunit and a β subunit, of which there are 18 and 8, respectively. Integrins span the cell membrane with a usually short cytoplasmic domain, while the N-terminal domain represents the binding region for ligands from the ECM. Murine HSPCs express integrin α4β1 (VLA4; ITGA4 and ITGB1), α5β1 (VLA5; ITGA5 and ITGB1), α6β1, αLβ2 (LFA1; ITGAL and ITGB2), and αMβ2 (MAC-1; ITGAM and ITGB2), while some HSPCs express integrin 3 (ITGB3) [37Klamer S Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment.Cell Adh Migr. 2014; 8: 563-577Crossref PubMed Scopus (38) Google Scholar]. Binding of integrins to proteins of the ECM, also termed outside-in signaling "outside-in signaling", activates intracellular signaling pathways leading to modulation of the position of other membrane receptors, the actin cytoskeleton, and possibly the cell cycle. This regulates cell spreading and retraction, migration, survival, shape, and differentiation. “Inside-out signaling,” that is, cell-intrinsic processes, which may lead to clustering or conformational changes of integrins, in contrast, stimulates ligand binding by integrins [57Calderwood DA Integrin activation.J Cell Sci. 2004; 117: 657-666Crossref PubMed Scopus (348) Google Scholar] and regulates integrin adhesiveness. This may be stimulated in normal and leukemic CD34+ HSCs by growth factors [58Lévesque JP Leavesley DI Niutta S Vadas M Simmons PJ Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins.J Exp Med. 1995; 181: 1805-1815Crossref PubMed Google Scholar,59Moore S Haylock DN Lévesque JP et al.Stem cell factor as a single agent induces selective proliferation of the Philadelphia chromosome positive fraction of chronic myeloid leukemia CD34(+) cells.Blood. 1998; 92: 2461-2470Crossref PubMed Google Scholar]. β1 integrins are essential for the homing and colonization of HSPCs in the fetal liver and the bone marrow. α4 integrins are involved in the differentiation of erythroid, myeloid, and B-cell progenitors, likely because of their role in transmigration of HSPCs through fibronectin. Humans and mice with mutations in the β2 integrin gene have leukocyte adhesion deficiency type I, leading to significant leukocytosis [60Bouvard D Brakebusch C Gustafsson E et al.Functional consequences of integrin gene mutations in mice.Circ Res. 2001; 89: 211-223Crossref PubMed Google Scholar]. Expression of DDR1 is found in epithelial cells, while DDR2 is expressed in MSCs and immature dendritic cells [61Gonzalez ME Martin EE Anwar T et al.Mesenchymal stem cell-induced DDR2 mediates stromal–breast cancer interactions and metastasis growth.Cell Rep. 2017; 18: 1215-1228Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar,62Borza CM Pozzi A. Discoidin domain receptors in disease.Matrix Biol. 2014; 34: 185-192Crossref PubMed Google Scholar]. LAIR-1, a receptor playing an inhibitory role in immune cell activation, is expressed on peripheral blood leukocytes and CD34+ HSCs [63Verbrugge A de Ruiter T Geest C Coffer PJ Meyaard L Differential expression of leukocyte-associated Ig-like receptor-1 during neutrophil differentiation and activation.J Leukoc Biol. 2006; 79: 828-836Crossref PubMed Scopus (38) Google Scholar]. LAIR-1 knockout mice are characterized by increased numbers of splenic B, regulatory T, and dendritic cells [64Tang X Tian L Esteso G et al.Leukocyte-associated Ig-like receptor-1-deficient mice have an altered immune cell phenotype.J Immunol. 2012; 188: 548-558Crossref PubMed Scopus (0) Google Scholar]. Several pieces of evidence for the involvement of the ECM in hematopoiesis exist. For example, deletion of Ext1, the gene encoding a glycosyltransferase essential for heparan sulfate production in BM stromal cells, led to mobilization of HSPCs from the BM [65Saez B Ferraro F Yusuf RZ et al.Inhibiting stromal cell heparan sulfate synthesis improves stem cell mobilization and enables engraftment without cytotoxic conditioning.Blood. 2014; 124: 2937-2947Crossref PubMed Scopus (17) Google Scholar]. Further, three-dimensional quantitative microscopy has revealed that sinusoidal endothelial cells and mesenchymal CXCL-12–abundant reticular cells closely associate with the ECM [66Gomariz A Helbling PM Isringhausen S et al.Quantitative spatial analysis of haematopoiesis-regulating stromal cells in the bone marrow microenvironment by 3D microscopy.Nat Commun. 2018; 9: 2532Crossref PubMed Scopus (28) Google Scholar]. With the use of microarray equipment, ECM ligands have been deposited on a substrate and used to study neural stem cell self-renewal and differentiation in a high-throughput assay, a strategy that can easily be adopted by the hematopoiesis field [67Soen Y Mori A Palmer TD Brown PO Exploring the regulation of human neural precursor cell differentiation using arrays of signaling microenvironments.Mol Syst Biol. 2006; 2: 37Crossref PubMed Scopus (184) Google Scholar]. Among the growth factors stored in the ECM are fibroblast growth factors (FGFs), interleukin (IL)-1, transforming growth factor (TGF) β1, and others, which may be secreted by BM stromal cells or hematopoietic precursors and influence hematopoietic lineage specification and proliferation [68Bodo M Baroni T Tabilio A Haematopoietic and stromal stem cell regulation by extracellular matrix components and growth factors.J Stem Cells. 2009; 4: 57-69PubMed Google Scholar]. Calcium ions trapped in the ECM close to the endosteum, whose concentration at this location is thought to be 20-fold higher than in serum [69Silver IA Murrills RJ Etherington DJ Microelectrode studies on the acid microenvironment beneath adherent macrophages and osteoclasts.Exp Cell Res. 1988; 175: 266-276Crossref PubMed Scopus (674) Google Scholar], also influence the localization and function of HSCs [70Adams GB Chabner KT Alley IR et al.Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor.Nature. 2006; 439: 599-603Crossref PubMed Scopus (546) Google Scholar]. Further, divalent cations, for example, Ca2+, Mg2+, and Mn2+, in the ECM modify the affinity of β1 integrins on HSPCs to their ligands, such as vascular cell adhesion molecule (VCAM) 1 [71Chigaev A Zwartz G Graves SW et al.Alpha4beta1 integrin affinity changes govern cell adhesion.J Biol Chem. 2003; 278: 38174-38182Crossref PubMed Scopus (0) Google Scholar], regulating HSPC homing in combination with mitogenic cytokines like granulocyte–macrophage colony-stimulating factor and KIT ligand [72Takamatsu Y Simmons PJ Lévesque JP Dual control by divalent cations and mitogenic cytokines of alpha 4 beta 1 and alpha 5 beta 1 integrin avidity expressed by human hemopoietic cells.Cell Adhes Commun. 1998; 5: 349-366Crossref PubMed Google Scholar]. Other ions in the BMM are zinc and copper [73Domingues MJ Cao H Heazlewood SY Cao B Nilsson SK Niche extracellular matrix components and their influence on HSC.J Cell Biochem. 2017; 118: 1984-1993Crossref PubMed Scopus (14) Google Scholar]. Lastly, prostaglandin E2, produced by osteoblasts and derived from the eicosanoid family, sphingosine 1-phosphate, and other lipids are also considered regulators of HSC homeostasis. The most abundant ECM protein in the bone marrow is collagen type I, although collagen types II, III, V, and XI, as well as proteoglycans, contribute to bone structure and micro-architecture determining bone strength [74Tzaphlidou M. The role of collagen in bone structure: an image processing approach.Micron. 2005; 36: 593-601Crossref PubMed Scopus (57) Google Scholar]. The structural hallmarks of all collagens are three individual α chains that form a triple-helical structure, which can be homotrimeric or heterotrimeric, depending on the 28 forms of collagen present [75Ricard-Blum S. The collagen family.Cold Spring Harb Perspect Biol. 2011; 3a004978Crossref PubMed Scopus (548) Google Scholar]. Collagen biosynthesis is a highly complex, multistep process, which occurs via chain association and folding of soluble precursors called pro-collagens and subsequent posttranslational modifications [76Kadler KE Holmes DF Trotter JA Chapman JA Collagen fibril formation.Biochem J. 1996; 316: 1-11Crossref PubMed Google Scholar]. The pro-α chains constituting the collagen triple helix consist of the repetitive motif glycine (Gly)–X–Y [77Ramshaw JA Shah NK Brodsky B Gly-X-Y tripeptide frequencies in collagen: a context for host–guest triple-helical peptides.J Struct Biol. 1998; 122: 86-91Crossref PubMed Scopus (0) Google Scholar]. The α chains undergo hydroxylation and glycosylation steps in the en
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
C5b6789n完成签到,获得积分10
4秒前
dio完成签到 ,获得积分10
6秒前
lemon完成签到 ,获得积分10
19秒前
zhdjj完成签到 ,获得积分10
42秒前
燕山堂完成签到 ,获得积分10
47秒前
小平完成签到 ,获得积分10
47秒前
Wgg完成签到 ,获得积分10
52秒前
1分钟前
zly完成签到 ,获得积分10
1分钟前
干就完了发布了新的文献求助10
1分钟前
chengzi完成签到 ,获得积分10
1分钟前
负责冰海完成签到 ,获得积分10
1分钟前
1分钟前
qyn1234566发布了新的文献求助10
2分钟前
qyn1234566完成签到,获得积分10
2分钟前
luochen完成签到,获得积分10
2分钟前
shiminyuan完成签到,获得积分10
2分钟前
ChaoZhang发布了新的文献求助10
2分钟前
科研通AI2S应助ZPH采纳,获得10
2分钟前
刘玲完成签到 ,获得积分10
3分钟前
keyanzhou完成签到 ,获得积分10
3分钟前
Glory完成签到 ,获得积分10
3分钟前
科研通AI2S应助Solitude采纳,获得20
3分钟前
3分钟前
Solitude发布了新的文献求助20
3分钟前
yinhe完成签到 ,获得积分10
3分钟前
Solitude完成签到,获得积分10
4分钟前
Sunny完成签到 ,获得积分10
4分钟前
美满的雁桃完成签到 ,获得积分10
4分钟前
WSY完成签到 ,获得积分10
4分钟前
独步出营完成签到 ,获得积分10
4分钟前
橙子味的邱憨憨完成签到 ,获得积分10
4分钟前
苏州九龙小7完成签到 ,获得积分10
4分钟前
自由飞翔完成签到 ,获得积分10
4分钟前
WenJun完成签到,获得积分10
5分钟前
刻苦的新烟完成签到 ,获得积分10
5分钟前
简单的惋庭完成签到 ,获得积分10
5分钟前
wyh295352318完成签到 ,获得积分10
5分钟前
退伍的三毛完成签到 ,获得积分10
5分钟前
yangzai完成签到 ,获得积分10
5分钟前
高分求助中
Sustainable Land Management: Strategies to Cope with the Marginalisation of Agriculture 1000
Corrosion and Oxygen Control 600
Yaws' Handbook of Antoine coefficients for vapor pressure 500
Python Programming for Linguistics and Digital Humanities: Applications for Text-Focused Fields 500
Love and Friendship in the Western Tradition: From Plato to Postmodernity 500
行動データの計算論モデリング 強化学習モデルを例として 500
Johann Gottlieb Fichte: Die späten wissenschaftlichen Vorlesungen / IV,1: ›Transzendentale Logik I (1812)‹ 400
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2557085
求助须知:如何正确求助?哪些是违规求助? 2180350
关于积分的说明 5623790
捐赠科研通 1901729
什么是DOI,文献DOI怎么找? 950059
版权声明 565633
科研通“疑难数据库(出版商)”最低求助积分说明 504851