24 results on '"Stem Cell Niche physiology"'
Search Results
2. Bone marrow niches for hematopoietic stem cells: life span dynamics and adaptation to acute stress.
- Author
-
Hofmann J and Kokkaliaris KD
- Subjects
- Animals, Humans, Adaptation, Physiological, Bone Marrow pathology, Bone Marrow metabolism, Bone Marrow physiology, Aging physiology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells physiology, Stem Cell Niche physiology, Stress, Physiological
- Abstract
Abstract: Hematopoietic stem cells (HSCs) are instrumental for organismal survival because they are responsible for lifelong production of mature blood lineages in homeostasis and response to external stress. To fulfill their function, HSCs rely on reciprocal interactions with specialized tissue microenvironments, termed HSC niches. From embryonic development to advanced aging, HSCs transition through several hematopoietic organs in which they are supported by distinct extrinsic cues. Here, we describe recent discoveries on how HSC niches collectively adapt to ensure robust hematopoietic function during biological aging and after exposure to acute stress. We also discuss the latest strategies leveraging niche-derived signals to revert aging-associated phenotypes and enhance hematopoietic recovery after myeloablation., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
3. The RIG-I-NRF2 axis regulates the mesenchymal stromal niche for bone marrow transplantation.
- Author
-
Lou Q, Jiang K, Xu Q, Yuan L, Xie S, Pan Y, Chen J, Wu J, Zhu J, Jiang L, and Zhao M
- Subjects
- Animals, Hematopoietic Stem Cells metabolism, Kelch-Like ECH-Associated Protein 1 metabolism, Mice, Reactive Oxygen Species metabolism, Stem Cell Niche physiology, Bone Marrow Transplantation, DEAD Box Protein 58 metabolism, NF-E2-Related Factor 2
- Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) support bone formation and constitute the stromal niche in regulating hematopoietic stem cells (HSCs). Stromal niche dysfunction affects HSC engraftment during transplantation; however, the underlying mechanisms remain elusive. In the present study, we found that all-trans retinoic acid (ATRA) and inflammation stress upregulated retinoic acid-inducible gene I (RIG-I) in BMSCs. Excess RIG-I expression damaged the clonogenicity, bone-forming ability of BMSCs and particularly their stromal niche function that supports HSC expansion in vitro and engraftment in vivo. Mechanistically, RIG-I elevation promoted the degradation of NRF2, a checkpoint for antioxidant cellular response, by altering the RIG-I-Trim25-Keap1-NRF2 complex, leading to reactive oxygen species (ROS) accumulation and BMSC damage. Genetic inhibition of RIG-I sustained NRF2 protein levels and reduced ROS levels in ATRA-treated BMSCs, thus preserving their clonogenicity, bone-forming ability, and stromal niche function in supporting HSC engraftment in mice. More importantly, RIG-I inhibition recovered the ATRA-treated stromal niche function to enhance HSC engraftment and emergency myelopoiesis for innate immunity against the bacterium Listeria monocytogenes during transplantation. Overall, we identified a noncanonical role of RIG-I in the regulation of the stromal niche for HSC transplantation., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
4. tfec controls the hematopoietic stem cell vascular niche during zebrafish embryogenesis.
- Author
-
Mahony CB, Fish RJ, Pasche C, and Bertrand JY
- Subjects
- Animals, Animals, Genetically Modified genetics, Animals, Genetically Modified metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Cytokines genetics, Cytokines metabolism, Embryo, Nonmammalian metabolism, Gene Expression Regulation, Developmental, Hematopoiesis physiology, Hematopoietic Stem Cells metabolism, Zebrafish genetics, Zebrafish metabolism, Zebrafish Proteins genetics, Animals, Genetically Modified embryology, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Embryo, Nonmammalian cytology, Embryonic Development physiology, Hematopoietic Stem Cells cytology, Stem Cell Niche physiology, Zebrafish embryology, Zebrafish Proteins metabolism
- Abstract
In mammals, embryonic hematopoiesis occurs in successive waves, culminating with the emergence of hematopoietic stem cells (HSCs) in the aorta. HSCs first migrate to the fetal liver (FL), where they expand, before they seed the bone marrow niche, where they will sustain hematopoiesis throughout adulthood. In zebrafish, HSCs emerge from the dorsal aorta and colonize the caudal hematopoietic tissue (CHT). Recent studies showed that they interact with endothelial cells (ECs), where they expand, before they reach their ultimate niche, the kidney marrow. We identified tfec, a transcription factor from the mitf family, which is highly enriched in caudal endothelial cells (cECs) at the time of HSC colonization in the CHT. Gain-of-function assays indicate that tfec is capable of expanding HSC-derived hematopoiesis in a non-cell-autonomous fashion. Furthermore, tfec mutants (generated by CRISPR/Cas9) showed reduced hematopoiesis in the CHT, leading to anemia. Tfec mediates these changes by increasing the expression of several cytokines in cECs from the CHT niche. Among these, we found kitlgb, which could rescue the loss of HSCs observed in tfec mutants. We conclude that tfec plays an important role in the niche to expand hematopoietic progenitors through the modulation of several cytokines. The full comprehension of the mechanisms induced by tfec will represent an important milestone toward the expansion of HSCs for regenerative purposes., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
5. ATF4 plays a pivotal role in the development of functional hematopoietic stem cells in mouse fetal liver.
- Author
-
Zhao Y, Zhou J, Liu D, Dong F, Cheng H, Wang W, Pang Y, Wang Y, Mu X, Ni Y, Li Z, Xu H, Hao S, Wang X, Ma S, Wang QF, Xiao G, Yuan W, Liu B, and Cheng T
- Subjects
- Activating Transcription Factor 4 genetics, Angiopoietin-Like Protein 3, Angiopoietin-like Proteins, Angiopoietins genetics, Angiopoietins metabolism, Animals, Fetus cytology, Hematopoietic Stem Cells cytology, Liver cytology, Mice, Mice, Knockout, Stromal Cells cytology, Stromal Cells metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor A metabolism, Activating Transcription Factor 4 metabolism, Cell Movement physiology, Fetus embryology, Hematopoietic Stem Cells metabolism, Liver embryology, Stem Cell Niche physiology
- Abstract
The fetal liver (FL) serves as a predominant site for expansion of functional hematopoietic stem cells (HSCs) during mouse embryogenesis. However, the mechanisms for HSC development in FL remain poorly understood. In this study, we demonstrate that deletion of activating transcription factor 4 (ATF4) significantly impaired hematopoietic development and reduced HSC self-renewal in FL. In contrast, generation of the first HSC population in the aorta-gonad-mesonephros region was not affected. The migration activity of ATF4(-/-) HSCs was moderately reduced. Interestingly, the HSC-supporting ability of both endothelial and stromal cells in FL was significantly compromised in the absence of ATF4. Gene profiling using RNA-seq revealed downregulated expression of a panel of cytokines in ATF4(-/-) stromal cells, including angiopoietin-like protein 3 (Angptl3) and vascular endothelial growth factor A (VEGFA). Addition of Angptl3, but not VEGFA, partially rescued the repopulating defect of ATF4(-/-) HSCs in the culture. Furthermore, chromatin immunoprecipitation assay in conjunction with silencing RNA-mediated silencing and complementary DNA overexpression showed transcriptional control of Angptl3 by ATF4. To summarize, ATF4 plays a pivotal role in functional expansion and repopulating efficiency of HSCs in developing FL, and it acts through upregulating transcription of cytokines such as Angptl3 in the microenvironment., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
6. ATF4, a new player in fetal HSC expansion.
- Author
-
Rieger MA
- Subjects
- Animals, Activating Transcription Factor 4 metabolism, Cell Movement physiology, Fetus embryology, Hematopoietic Stem Cells metabolism, Liver embryology, Stem Cell Niche physiology
- Published
- 2015
- Full Text
- View/download PDF
7. Investigating osteogenic differentiation in multiple myeloma using a novel 3D bone marrow niche model.
- Author
-
Reagan MR, Mishima Y, Glavey SV, Zhang Y, Manier S, Lu ZN, Memarzadeh M, Zhang Y, Sacco A, Aljawai Y, Shi J, Tai YT, Ready JE, Kaplan DL, Roccaro AM, and Ghobrial IM
- Subjects
- Cell Differentiation, Cells, Cultured, Coculture Techniques, Human Umbilical Vein Endothelial Cells cytology, Humans, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells physiology, Models, Biological, Osteoblasts cytology, Osteoblasts physiology, Tissue Scaffolds, Bone Marrow Cells pathology, Multiple Myeloma pathology, Osteogenesis physiology, Primary Cell Culture methods, Stem Cell Niche physiology
- Abstract
Clonal proliferation of plasma cells within the bone marrow (BM) affects local cells, such as mesenchymal stromal cells (MSCs), leading to osteolysis and fatality in multiple myeloma (MM). Consequently, there is an urgent need to find better mechanisms of inhibiting myeloma growth and osteolytic lesion development. To meet this need and accelerate clinical translation, better models of myeloma within the BM are required. Herein we have developed a clinically relevant, three-dimensional (3D) myeloma BM coculture model that mimics bone cell/cancer cell interactions within the bone microenvironment. The coculture model and clinical samples were used to investigate myeloma growth, osteogenesis inhibition, and myeloma-induced abnormalities in MM-MSCs. This platform demonstrated myeloma support of capillary-like assembly of endothelial cells and cell adhesion-mediated drug resistance (CAM-DR). Also, distinct normal donor (ND)- and MM-MSC miRNA (miR) signatures were identified and used to uncover osteogenic miRs of interest for osteoblast differentiation. More broadly, our 3D platform provides a simple, clinically relevant tool to model cancer growth within the bone-useful for investigating skeletal cancer biology, screening compounds, and exploring osteogenesis. Our identification and efficacy validation of novel bone anabolic miRs in MM opens more opportunities for novel approaches to cancer therapy via stromal miR modulation., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
8. CD166 regulates human and murine hematopoietic stem cells and the hematopoietic niche.
- Author
-
Chitteti BR, Kobayashi M, Cheng Y, Zhang H, Poteat BA, Broxmeyer HE, Pelus LM, Hanenberg H, Zollman A, Kamocka MM, Carlesso N, Cardoso AA, Kacena MA, and Srour EF
- Subjects
- Animals, Antigens, CD metabolism, Chromatin Immunoprecipitation, Flow Cytometry, Hematopoietic Stem Cells physiology, Humans, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, SCID, Receptors, Cell Surface metabolism, Signaling Lymphocytic Activation Molecule Family Member 1, Activated-Leukocyte Cell Adhesion Molecule physiology, Biomarkers metabolism, Bone Marrow Cells metabolism, Hematopoietic Stem Cells cytology, Stem Cell Niche physiology
- Abstract
We previously showed that immature CD166(+) osteoblasts (OB) promote hematopoietic stem cell (HSC) function. Here, we demonstrate that CD166 is a functional HSC marker that identifies both murine and human long-term repopulating cells. Both murine LSKCD48(-)CD166(+)CD150(+) and LSKCD48(-)CD166(+)CD150(+)CD9(+) cells, as well as human Lin(-)CD34(+)CD38(-)CD49f(+)CD166(+) cells sustained significantly higher levels of chimerism in primary and secondary recipients than CD166(-) cells. CD166(-/-) knockout (KO) LSK cells engrafted poorly in wild-type (WT) recipients and KO bone marrow cells failed to radioprotect lethally irradiated WT recipients. CD166(-/-) hosts supported short-term, but not long-term WT HSC engraftment, confirming that loss of CD166 is detrimental to the competence of the hematopoietic niche. CD166(-/-) mice were significantly more sensitive to hematopoietic stress. Marrow-homed transplanted WT hematopoietic cells lodged closer to the recipient endosteum than CD166(-/-) cells, suggesting that HSC-OB homophilic CD166 interactions are critical for HSC engraftment. STAT3 has 3 binding sites on the CD166 promoter and STAT3 inhibition reduced CD166 expression, suggesting that both CD166 and STAT3 may be functionally coupled and involved in HSC competence. These studies illustrate the significance of CD166 in the identification and engraftment of HSC and in HSC-niche interactions, and suggest that CD166 expression can be modulated to enhance HSC function., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
9. HSCs and niche relations marked by CD166.
- Author
-
Shiozawa Y and Taichman RS
- Subjects
- Animals, Humans, Activated-Leukocyte Cell Adhesion Molecule physiology, Biomarkers metabolism, Bone Marrow Cells metabolism, Hematopoietic Stem Cells cytology, Stem Cell Niche physiology
- Published
- 2014
- Full Text
- View/download PDF
10. In vivo time-lapse imaging shows diverse niche engagement by quiescent and naturally activated hematopoietic stem cells.
- Author
-
Rashidi NM, Scott MK, Scherf N, Krinner A, Kalchschmidt JS, Gounaris K, Selkirk ME, Roeder I, and Lo Celso C
- Subjects
- Animals, Bone Marrow immunology, Bone Marrow metabolism, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells metabolism, Hyaluronan Receptors immunology, Hyaluronan Receptors metabolism, Mice, Microscopy, Confocal, Receptors, CXCR4 immunology, Receptors, CXCR4 metabolism, Time-Lapse Imaging, Trichinella spiralis, Trichinellosis immunology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Stem Cell Niche physiology, Trichinellosis metabolism
- Abstract
Hematopoietic stem cells (HSCs) maintain the turnover of mature blood cells during steady state and in response to systemic perturbations such as infections. Their function critically depends on complex signal exchanges with the bone marrow (BM) microenvironment in which they reside, but the cellular mechanisms involved in HSC-niche interactions and regulating HSC function in vivo remain elusive. We used a natural mouse parasite, Trichinella spiralis, and multipoint intravital time-lapse confocal microscopy of mouse calvarium BM to test whether HSC-niche interactions may change when hematopoiesis is perturbed. We find that steady-state HSCs stably engage confined niches in the BM whereas HSCs harvested during acute infection are motile and therefore interact with larger niches. These changes are accompanied by increased long-term repopulation ability and expression of CD44 and CXCR4. Administration of a CXCR4 antagonist affects the duration of HSC-niche interactions. These findings suggest that HSC-niche interactions may be modulated during infection., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
11. Megakaryocytes promote murine osteoblastic HSC niche expansion and stem cell engraftment after radioablative conditioning.
- Author
-
Olson TS, Caselli A, Otsuru S, Hofmann TJ, Williams R, Paolucci P, Dominici M, and Horwitz EM
- Subjects
- Animals, Becaplermin, Cell Movement physiology, Cell Movement radiation effects, Cell Proliferation, Endothelium, Vascular, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Graft Survival, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells radiation effects, Megakaryocytes metabolism, Megakaryocytes radiation effects, Mice, Mice, Inbred C57BL, Osteoblasts metabolism, Osteoblasts radiation effects, Proto-Oncogene Proteins c-sis metabolism, Signal Transduction, Thrombopoietin metabolism, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Megakaryocytes cytology, Osteoblasts cytology, Receptors, Thrombopoietin physiology, Stem Cell Niche physiology, Whole-Body Irradiation
- Abstract
Successful hematopoietic stem cell (HSC) transplantation requires donor HSC engraftment within specialized bone marrow microenvironments known as HSC niches. We have previously reported a profound remodeling of the endosteal osteoblastic HSC niche after total body irradiation (TBI), defined as relocalization of surviving megakaryocytes to the niche site and marked expansion of endosteal osteoblasts. We now demonstrate that host megakaryocytes function critically in expansion of the endosteal niche after preparative radioablation and in the engraftment of donor HSC. We show that TBI-induced migration of megakaryocytes to the endosteal niche depends on thrombopoietin signaling through the c-MPL receptor on megakaryocytes, as well as CD41 integrin-mediated adhesion. Moreover, niche osteoblast proliferation post-TBI required megakaryocyte-secreted platelet-derived growth factor-BB. Furthermore, blockade of c-MPL-dependent megakaryocyte migration and function after TBI resulted in a significant decrease in donor HSC engraftment in primary and competitive secondary transplantation assays. Finally, we administered thrombopoietin to mice beginning 5 days before marrow radioablation and ending 24 hours before transplant to enhance megakaryocyte function post-TBI, and found that this strategy significantly enhanced donor HSC engraftment, providing a rationale for improving hematopoietic recovery and perhaps overall outcome after clinical HSC transplantation.
- Published
- 2013
- Full Text
- View/download PDF
12. Development of a vascular niche platform for expansion of repopulating human cord blood stem and progenitor cells.
- Author
-
Butler JM, Gars EJ, James DJ, Nolan DJ, Scandura JM, and Rafii S
- Subjects
- Animals, Cell Culture Techniques methods, Cells, Cultured, Cord Blood Stem Cell Transplantation methods, Fetal Blood physiology, Hematopoietic Stem Cells cytology, Humans, Mice, Mice, Inbred NOD, Mice, Transgenic, Tissue Engineering methods, Blood Vessels cytology, Cell Proliferation, Fetal Blood cytology, Hematopoietic Stem Cells physiology, Stem Cell Niche physiology, Tissue Scaffolds
- Abstract
Transplantation of ex vivo expanded human umbilical cord blood cells (hCB) only partially enhances the hematopoietic recovery after myelosuppressive therapy. Incubation of hCB with optimal combinations of cytokines and niche cells, such as endothelial cells (ECs), could augment the efficiency of hCB expansion. We have devised an approach to cultivate primary human ECs (hECs) in serum-free culture conditions. We demonstrate that coculture of CD34(+) hCB in direct cellular contact with hECs and minimal concentrations of thrombopoietin/Kit-ligand/Flt3-ligand resulted in a 400-fold expansion of total hematopoietic cells, 150-fold expansion of CD45(+)CD34(+) progenitor cells, and 23-fold expansion of CD45(+) Lin(-)CD34(hi+)CD45RA(-)CD49f(+) stem and progenitor cells over a 12-day period. Compared with cytokines alone, coculture of hCB with hECs permitted greater expansion of cells capable of multilineage engraftment and serial transplantation, hallmarks of long-term repopulating hematopoietic stem cells. Therefore, hECs establish a cellular platform for expansion of hematopoietic stem and progenitor cells and treatment of hematologic disorders.
- Published
- 2012
- Full Text
- View/download PDF
13. Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors.
- Author
-
Gonzalez-Nieto D, Li L, Kohler A, Ghiaur G, Ishikawa E, Sengupta A, Madhu M, Arnett JL, Santho RA, Dunn SK, Fishman GI, Gutstein DE, Civitelli R, Barrio LC, Gunzer M, and Cancelas JA
- Subjects
- Animals, Chemokine CXCL12 genetics, Chemokine CXCL12 metabolism, Connexin 43 genetics, Hematopoietic Stem Cells cytology, Mice, Mice, Mutant Strains, Osteoblasts cytology, Cell Movement physiology, Connexin 43 metabolism, Hematopoietic Stem Cells metabolism, Osteoblasts metabolism, Stem Cell Niche physiology
- Abstract
Connexin-43 (Cx43), a gap junction protein involved in control of cell proliferation, differentiation and migration, has been suggested to have a role in hematopoiesis. Cx43 is highly expressed in osteoblasts and osteogenic progenitors (OB/P). To elucidate the biologic function of Cx43 in the hematopoietic microenvironment (HM) and its influence in hematopoietic stem cell (HSC) activity, we studied the hematopoietic function in an in vivo model of constitutive deficiency of Cx43 in OB/P. The deficiency of Cx43 in OB/P cells does not impair the steady state hematopoiesis, but disrupts the directional trafficking of HSC/progenitors (Ps) between the bone marrow (BM) and peripheral blood (PB). OB/P Cx43 is a crucial positive regulator of transstromal migration and homing of both HSCs and progenitors in an irradiated microenvironment. However, OB/P Cx43 deficiency in nonmyeloablated animals does not result in a homing defect but induces increased endosteal lodging and decreased mobilization of HSC/Ps associated with proliferation and expansion of Cxcl12-secreting mesenchymal/osteolineage cells in the BM HM in vivo. Cx43 controls the cellular content of the BM osteogenic microenvironment and is required for homing of HSC/Ps in myeloablated animals.
- Published
- 2012
- Full Text
- View/download PDF
14. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice.
- Author
-
Gil-Bernabé AM, Ferjancic S, Tlalka M, Zhao L, Allen PD, Im JH, Watson K, Hill SA, Amirkhosravi A, Francis JL, Pollard JW, Ruf W, and Muschel RJ
- Subjects
- Animals, Blood Coagulation physiology, Cell Movement physiology, Cell Survival drug effects, Cell Survival physiology, Cells, Cultured, Humans, Macrophages metabolism, Macrophages physiology, Melanoma, Experimental metabolism, Melanoma, Experimental pathology, Mice, Mice, Inbred C57BL, Mice, SCID, Mice, Transgenic, Monocytes metabolism, Monocytes physiology, Neoplasm Metastasis, Neoplasms metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Neoplastic Stem Cells physiology, Stem Cell Niche drug effects, Thromboplastin metabolism, Blood Coagulation drug effects, Cell Movement drug effects, Macrophages drug effects, Monocytes drug effects, Neoplasms pathology, Stem Cell Niche physiology, Thromboplastin pharmacology
- Abstract
Tissue factor (TF) expression by tumor cells correlates with metastasis clinically and supports metastasis in experimental settings. However, the precise pathways coupling TF to malignancy remain incompletely defined. Here, we show that clot formation by TF indirectly enhances tumor cell survival after arrest in the lung, during experimental lung metastasis, by recruiting macrophages characterized by CD11b, CD68, F4/80, and CX(3)CR1 (but not CD11c) expression. Genetic or pharmacologic inhibition of coagulation, by either induction of TF pathway inhibitor ex-pression or by treatment with hirudin, respectively, abrogated macrophage recruitment and tumor cell survival. Furthermore, impairment of macrophage function, in either Mac1-deficient mice or in CD11b-diphtheria toxin receptor mice in which CD11b-positive cells were ablated, decreased tumor cell survival without altering clot formation, demonstrating that the recruitment of functional macrophages was essential for tumor cell survival. This effect was independent of NK cells. Moreover, a similar population of macrophages was also recruited to the lung during the formation of a premetastatic niche. Anticoagulation inhibited their accumulation and prevented the enhanced metastasis associated with the formation of the niche. Our study, for the first time, links TF induced coagulation to macrophage recruitment in the metastatic process.
- Published
- 2012
- Full Text
- View/download PDF
15. Wnt cross-talk in the niche.
- Author
-
Staal FJ and Fibbe WE
- Subjects
- Animals, Humans, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Stem Cell Niche physiology, Wnt Signaling Pathway physiology
- Published
- 2012
- Full Text
- View/download PDF
16. An intrinsic BM hematopoietic niche occupancy defect of HSC in scid mice facilitates exogenous HSC engraftment.
- Author
-
Qing Y, Lin Y, and Gerson SL
- Subjects
- Animals, Bone Marrow Cells metabolism, Bone Marrow Cells physiology, Cell Movement physiology, Cell Proliferation, Cells, Cultured, DNA End-Joining Repair genetics, DNA End-Joining Repair immunology, DNA End-Joining Repair physiology, Hematopoietic Stem Cells metabolism, Homeodomain Proteins physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Stem Cell Niche physiology, Cell Movement genetics, Graft Survival genetics, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells physiology, Homeodomain Proteins genetics, Stem Cell Niche genetics
- Abstract
Although scid mice have been widely used for human HSC engraftment studies, the function of HSCs of scid mice has not been characterized. We hypothesized that the DNA repair defect of scid mice results in a stem cell defect that facilitates HSC engraftment. scid BM cells showed severely impaired repopulation potentials in the competitive repopulation assay. To assess the BM hematopoietic niche occupancy ability of scid HSC, WT BM cells were transplanted into scid mice without any conditioning and observed to achieve long-term engraftment. Furthermore, the defects of scid HSCs are independent of their inability to perform lymphopoiesis because a similar defect in hematopoietic niche occupancy was not observed with Rag1(-/-) recipients. These results demonstrate that scid HSCs are impaired in maintenance within the niche, which may explain the nature of the conducive marrow niche environment of scid mice for xenotransplantation.
- Published
- 2012
- Full Text
- View/download PDF
17. The canonical Wnt pathway shapes niches supportive of hematopoietic stem/progenitor cells.
- Author
-
Ichii M, Frank MB, Iozzo RV, and Kincade PW
- Subjects
- Adult, Animals, Cell Differentiation genetics, Cell Proliferation, Cells, Cultured, Gene Expression Profiling, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Humans, Infant, Newborn, Lymphopoiesis genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Microarray Analysis, Stem Cell Niche genetics, Wnt Signaling Pathway genetics, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Stem Cell Niche physiology, Wnt Signaling Pathway physiology
- Abstract
Considerable information has accumulated about components of BM that regulate the survival, self-renewal, and differentiation of hematopoietic cells. In the present study, we investigated Wnt signaling and assessed its influence on human and murine hematopoiesis. Hematopoietic stem/progenitor cells (HSPCs) were placed on Wnt3a-transduced OP9 stromal cells. The proliferation and production of B cells, natural killer cells, and plasmacytoid dendritic cells were blocked. In addition, some HSPC characteristics were maintained or re-acquired along with different lineage generation potentials. These responses did not result from direct effects of Wnt3a on HSPCs, but also required alterations in the OP9 cells. Microarray, PCR, and flow cytometric experiments revealed that OP9 cells acquired osteoblastic characteristics while down-regulating some features associated with mesenchymal stem cells, including the expression of angiopoietin 1, the c-Kit ligand, and VCAM-1. In contrast, the production of decorin, tenascins, and fibromodulin markedly increased. We found that at least 1 of these extracellular matrix components, decorin, is a regulator of hematopoiesis: upon addition of this proteoglycan to OP9 cocultures, decorin caused changes similar to those caused by Wnt3a. Furthermore, hematopoietic stem cell numbers in the BM and spleen were elevated in decorin-knockout mice. These findings define one mechanism through which canonical Wnt signaling could shape niches supportive of hematopoiesis.
- Published
- 2012
- Full Text
- View/download PDF
18. Galectin-1-expressing stromal cells constitute a specific niche for pre-BII cell development in mouse bone marrow.
- Author
-
Mourcin F, Breton C, Tellier J, Narang P, Chasson L, Jorquera A, Coles M, Schiff C, and Mancini SJ
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Bone Marrow Cells physiology, Cell Differentiation immunology, Cells, Cultured, Green Fluorescent Proteins genetics, Interleukin-7 metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Pre-B Cell Receptors metabolism, Precursor Cells, B-Lymphoid cytology, Precursor Cells, B-Lymphoid metabolism, Stem Cell Niche cytology, Stem Cell Niche metabolism, Stromal Cells cytology, Stromal Cells metabolism, Bone Marrow metabolism, Bone Marrow physiology, Galectin 1 metabolism, Precursor Cells, B-Lymphoid physiology, Stem Cell Niche physiology, Stromal Cells physiology
- Abstract
In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7-producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1(+) reticular cells. We also determined that IL-7(+) and GAL1(+) cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7(+) to GAL1(+) supportive BM niches during their development.
- Published
- 2011
- Full Text
- View/download PDF
19. Inhibition of osteoclast function reduces hematopoietic stem cell numbers in vivo.
- Author
-
Lymperi S, Ersek A, Ferraro F, Dazzi F, and Horwood NJ
- Subjects
- Animals, Blotting, Western, Bone Density Conservation Agents pharmacology, Bone Marrow metabolism, Bone Marrow pathology, Bone Marrow Transplantation, Bone Resorption metabolism, Cell Division physiology, Cells, Cultured, Female, Flow Cytometry, Hematopoietic Stem Cells metabolism, Leukocyte Common Antigens physiology, Mice, Mice, Inbred C57BL, Osteoclasts cytology, Osteoclasts metabolism, Parathyroid Hormone pharmacology, S Phase physiology, Stem Cell Niche drug effects, Thy-1 Antigens physiology, Tomography, X-Ray Computed, Bone Resorption etiology, Bone Resorption pathology, Diphosphonates pharmacology, Hematopoietic Stem Cells pathology, Hematopoietic System physiology, Osteoclasts drug effects, Stem Cell Niche physiology
- Abstract
Osteoblasts play a crucial role in the hematopoietic stem cell (HSC) niche; however, an overall increase in their number does not necessarily promote hematopoiesis. Because the activity of osteoblasts and osteoclasts is coordinately regulated, we hypothesized that active bone-resorbing osteoclasts would participate in HSC niche maintenance. Mice treated with bisphosphonates exhibited a decrease in proportion and absolute number of Lin(-)cKit(+)Sca1(+) Flk2(-) (LKS Flk2(-)) and long-term culture-initiating cells in bone marrow (BM). In competitive transplantation assays, the engraftment of treated BM cells was inferior to that of controls, confirming a decrease in HSC numbers. Accordingly, bisphosphonates abolished the HSC increment produced by parathyroid hormone. In contrast, the number of colony-forming-unit cells in BM was increased. Because a larger fraction of LKS in the BM of treated mice was found in the S/M phase of the cell cycle, osteoclast impairment makes a proportion of HSCs enter the cell cycle and differentiate. To prove that HSC impairment was a consequence of niche manipulation, a group of mice was treated with bisphosphonates and then subjected to BM transplantation from untreated donors. Treated recipient mice experienced a delayed hematopoietic recovery compared with untreated controls. Our findings demonstrate that osteoclast function is fundamental in the HSC niche.
- Published
- 2011
- Full Text
- View/download PDF
20. Role for vitamin D receptor in the neuronal control of the hematopoietic stem cell niche.
- Author
-
Kawamori Y, Katayama Y, Asada N, Minagawa K, Sato M, Okamura A, Shimoyama M, Nakagawa K, Okano T, Tanimoto M, Kato S, and Matsui T
- Subjects
- Adrenergic Agonists pharmacology, Animals, Blotting, Western, Calcium administration & dosage, Cell Movement, Chemokine CXCL12 metabolism, Enzyme-Linked Immunosorbent Assay, Female, Flow Cytometry, Fluorescent Antibody Technique, Granulocyte Colony-Stimulating Factor administration & dosage, Hematopoietic Stem Cell Mobilization, Leukocyte Common Antigens physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Osteoblasts cytology, RANK Ligand genetics, RANK Ligand metabolism, RNA, Messenger genetics, Receptors, Adrenergic, beta-1 chemistry, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Vitamin D analogs & derivatives, Vitamin D pharmacology, Hematopoietic Stem Cells physiology, Osteoblasts metabolism, Receptors, Calcitriol physiology, Stem Cell Niche physiology, Sympathetic Nervous System metabolism
- Abstract
Hematopoietic stem/progenitor cells (HSPCs) are released from the bone marrow to the circulation by the cytokine, granulocyte colony-stimulating factor, via sympathetic nervous system (SNS)-mediated osteoblast suppression. Because the orientation of HSPCs in their osteoblastic niche is reported to be guided by [Ca(2+)], we speculated on a cooperation between the calcium-regulating hormones and SNS in the regulation of HSPC trafficking. Here, we present the severe impairment of granulocyte colony-stimulating factor-induced osteoblast suppression and subsequent HSPC mobilization in vitamin D receptor (VDR)-deficient mice. In osteoblasts, functional VDR possessing, at least in part, a transcriptional activity, was specifically induced by β2-adrenergic receptor (AR) agonists. While β2-AR agonists transiently increased mRNA expression of Vdr and its downstream gene, Rankl, 1α,25-dihydroxyvitamin-D(3) sustained the β2-AR-induced Rankl expression at high level by stabilizing VDR protein. These data suggest that VDR is essential for durable β2-AR signaling in the stem cell niche. Our study demonstrates not only a novel function of VDR as a critical modulator of HSPC trafficking, but also the presence of a SNS-mediated, bone-remodeling mechanism through VDR. VDR contributes to brain-bone-blood integration in an unanticipated way distinct from other classical calcium-regulating hormones.
- Published
- 2010
- Full Text
- View/download PDF
21. Endothelial protein C receptor-expressing hematopoietic stem cells reside in the perisinusoidal niche in fetal liver.
- Author
-
Iwasaki H, Arai F, Kubota Y, Dahl M, and Suda T
- Subjects
- Animals, Antigens, Ly metabolism, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Bone Marrow Cells physiology, Cell Movement physiology, Cells, Cultured, Embryo, Mammalian, Endothelial Protein C Receptor, Female, Fetus cytology, Fetus metabolism, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells metabolism, Liver cytology, Liver metabolism, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Pregnancy, Proto-Oncogene Proteins c-kit metabolism, Receptors, Cell Surface, Stem Cell Niche metabolism, Stem Cell Niche physiology, Glycoproteins metabolism, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Liver embryology, Stem Cell Niche cytology
- Abstract
Hematopoietic stem cells (HSCs) are maintained in specialized niches in adult bone marrow. However, niche and HSC maintenance mechanism in fetal liver (FL) still remains unclear. Here, we investigated the niche and the molecular mechanism of HSC maintenance in mouse FL using HSCs expressing endothelial protein C receptor (EPCR). The antiapoptotic effect of activated protein C (APC) on EPCR(+) HSCs and the expression of protease-activated receptor 1 (Par-1) mRNA in these cells suggested the involvement of the cytoprotective APC/EPCR/Par-1 pathway in HSC maintenance. Immunohistochemistry revealed that EPCR(+) cells were localized adjacent to, or integrated in, the Lyve-1(+) sinusoidal network, where APC and extracellular matrix (ECM) are abundant, suggesting that HSCs in FL were maintained in the APC- and ECM-rich perisinusoidal niche. EPCR(+) HSCs were in a relatively slow cycling state, consistent with their high expression levels of p57 and p18. Furthermore, the long-term reconstitution activity of EPCR(+) HSCs decreased significantly after short culture but not when cocultured with feeder layer of FL-derived Lyve-1(+) cells, which suggests that the maintenance of the self-renewal activity of FL HSCs largely depended on the interaction with the perisinusoidal niche. In conclusion, EPCR(+) HSCs resided in the perisinusoidal niche in mouse FL.
- Published
- 2010
- Full Text
- View/download PDF
22. Knockdown of N-cadherin suppresses the long-term engraftment of hematopoietic stem cells.
- Author
-
Hosokawa K, Arai F, Yoshihara H, Iwasaki H, Nakamura Y, Gomei Y, and Suda T
- Subjects
- Animals, Cadherins physiology, Cell Adhesion genetics, Cell Adhesion physiology, Cell Movement genetics, Cell Movement physiology, Cell Survival genetics, Cells, Cultured, Gene Knockdown Techniques, Hematopoietic Stem Cells physiology, Mice, Mice, Congenic, Mice, Inbred C57BL, NIH 3T3 Cells, Stem Cell Niche physiology, Time Factors, Cadherins genetics, Graft Survival genetics, Hematopoietic Stem Cell Transplantation
- Abstract
During postnatal life, the bone marrow (BM) supports both self-renewal and differentiation of hematopoietic stem cells (HSCs) in specialized microenvironments termed stem cell niches. Cell-cell and cell-extracellular matrix interactions between HSCs and their niches are critical for the maintenance of HSC properties. Here, we analyzed the function of N-cadherin in the regulation of the proliferation and long-term repopulation activity of hematopoietic stem/progenitor cells (HSPCs) by the transduction of N-cadherin shRNA. Inhibition of N-cadherin expression accelerated cell division in vitro and reduced the lodgment of donor HSPCs to the endosteal surface, resulting in a significant reduction in long-term engraftment. Cotransduction of N-cadherin shRNA and a mutant N-cadherin that introduced the silent mutations to shRNA target sequences rescued the accelerated cell division and reconstitution phenotypes. In addition, the requirement of N-cadherin for HSPC engraftment appears to be niche specific, as shN-cad-transduced lineage(-)Sca-1(+)c-Kit(+) cells successfully engrafted in spleen, which lacks an osteoblastic niche. These findings suggest that N-cad-mediated cell adhesion is functionally required for the establishment of hematopoiesis in the BM niche after BM transplantation.
- Published
- 2010
- Full Text
- View/download PDF
23. Impact of interactions of cellular components of the bone marrow microenvironment on hematopoietic stem and progenitor cell function.
- Author
-
Chitteti BR, Cheng YH, Poteat B, Rodriguez-Rodriguez S, Goebel WS, Carlesso N, Kacena MA, and Srour EF
- Subjects
- Animals, Cell Communication, Cell Differentiation physiology, Cell Proliferation, Cell Separation, Cells, Cultured, Coculture Techniques, Flow Cytometry, Hematopoietic Stem Cells metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Osteoblasts metabolism, Receptors, Notch metabolism, Reverse Transcriptase Polymerase Chain Reaction, Stromal Cells cytology, Stromal Cells metabolism, Bone Marrow metabolism, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Osteoblasts cytology, Signal Transduction physiology, Stem Cell Niche physiology
- Abstract
Hematopoietic stem (HSC) and progenitor (HPC) cell fate is governed by intrinsic and extrinsic parameters. We examined the impact of hematopoietic niche elements on HSC and HPC function by analyzing the combined effect of osteoblasts (OBs) and stromal cells (SCs) on Lineage(-)Sca-1(+)CD117(+) (LSK) cells. CFU expansion and marrow repopulating potential of cultured Lineage(-)Sca-1(+)CD117(+) cells were significantly higher in OB compared with SC cultures, thus corroborating the importance of OBs in the competence of the hematopoietic niche. OB-mediated enhancement of HSC and HPC function was reduced in cocultures of OBs and SCs, suggesting that SCs suppressed the OB-mediated hematopoiesis-enhancing activity. Although the suppressive effect of SC was mediated by adipocytes, probably through up-regulation of neuropilin-1, the OB-mediated enhanced hematopoiesis function was elaborated through Notch signaling. Expression of Notch 2, Jagged 1 and 2, Delta 1 and 4, Hes 1 and 5, and Deltex was increased in OB cultures and suppressed in SC and OB/SC cultures. Phenotypic fractionation of OBs did not segregate the hematopoiesis-enhancing activity but demonstrated that this function is common to OBs from different anatomic sites. These data illustrate that OBs promote in vitro maintenance of hematopoietic functions, including repopulating potential by up-regulating Notch-mediated signaling between HSCs and OBs.
- Published
- 2010
- Full Text
- View/download PDF
24. SHIP is required for a functional hematopoietic stem cell niche.
- Author
-
Hazen AL, Smith MJ, Desponts C, Winter O, Moser K, and Kerr WG
- Subjects
- Animals, Bone Marrow metabolism, Bone Marrow physiology, Cell Proliferation drug effects, Cells, Cultured, Culture Media, Conditioned pharmacology, Cytokines blood, Cytokines pharmacology, Hematopoietic Stem Cell Mobilization, Hematopoietic Stem Cells drug effects, Inositol Polyphosphate 5-Phosphatases, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphoric Monoester Hydrolases genetics, Phosphoric Monoester Hydrolases metabolism, Receptors, CXCR4 metabolism, Signal Transduction genetics, Stem Cell Niche physiology, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells physiology, Phosphoric Monoester Hydrolases physiology, Stem Cell Niche metabolism
- Abstract
SH2-domain-containing inositol 5'-phosphatase-1 (SHIP) deficiency significantly increases the number of hematopoietic stem cells (HSCs) present in the bone marrow (BM). However, the reconstitution capacity of these HSCs is severely impaired, suggesting that SHIP expression might be an intrinsic requirement for HSC function. To further examine this question, we developed a model in which SHIP expression is ablated in HSCs while they are resident in a SHIP-competent milieu. In this setting, we find that long-term repopulation by SHIP-deficient HSCs is not compromised. Moreover, SHIP-deficient HSCs from this model repopulate at levels comparable with wild-type HSCs upon serial transfer. However, when HSCs from mice with systemic ablation of SHIP are transplanted, they are functionally compromised for repopulation. These findings demonstrate that SHIP is not an intrinsic requirement for HSC function, but rather that SHIP is required for the BM milieu to support functionally competent HSCs. Consistent with these findings, cells that comprise the BM niche express SHIP and SHIP deficiency profoundly alters their function.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.