Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 177(7): 1915-1932.e16, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-31130381

RESUMO

Stroma is a poorly defined non-parenchymal component of virtually every organ with key roles in organ development, homeostasis, and repair. Studies of the bone marrow stroma have defined individual populations in the stem cell niche regulating hematopoietic regeneration and capable of initiating leukemia. Here, we use single-cell RNA sequencing (scRNA-seq) to define a cellular taxonomy of the mouse bone marrow stroma and its perturbation by malignancy. We identified seventeen stromal subsets expressing distinct hematopoietic regulatory genes spanning new fibroblastic and osteoblastic subpopulations including distinct osteoblast differentiation trajectories. Emerging acute myeloid leukemia impaired mesenchymal osteogenic differentiation and reduced regulatory molecules necessary for normal hematopoiesis. These data suggest that tissue stroma responds to malignant cells by disadvantaging normal parenchymal cells. Our taxonomy of the stromal compartment provides a comprehensive bone marrow cell census and experimental support for cancer cell crosstalk with specific stromal elements to impair normal tissue function and thereby enable emergent cancer.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular , Homeostase , Leucemia Mieloide Aguda/metabolismo , Osteoblastos/metabolismo , Osteogênese , Microambiente Tumoral , Animais , Células da Medula Óssea/patologia , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Osteoblastos/patologia , Células Estromais/metabolismo , Células Estromais/patologia
2.
Cell ; 175(1): 43-56.e21, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241615

RESUMO

Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.


Assuntos
Desenvolvimento Ósseo/fisiologia , Osso e Ossos/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Osso e Ossos/metabolismo , Cartilagem/citologia , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Análise de Célula Única/métodos , Células-Tronco/citologia , Células Estromais/citologia , Transcriptoma/genética
3.
Immunity ; 55(5): 862-878.e8, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35508166

RESUMO

Macrophage colony stimulating factor-1 (CSF-1) plays a critical role in maintaining myeloid lineage cells. However, congenital global deficiency of CSF-1 (Csf1op/op) causes severe musculoskeletal defects that may indirectly affect hematopoiesis. Indeed, we show here that osteolineage-derived Csf1 prevented developmental abnormalities but had no effect on monopoiesis in adulthood. However, ubiquitous deletion of Csf1 conditionally in adulthood decreased monocyte survival, differentiation, and migration, independent of its effects on bone development. Bone histology revealed that monocytes reside near sinusoidal endothelial cells (ECs) and leptin receptor (Lepr)-expressing perivascular mesenchymal stromal cells (MSCs). Targeted deletion of Csf1 from sinusoidal ECs selectively reduced Ly6C- monocytes, whereas combined depletion of Csf1 from ECs and MSCs further decreased Ly6Chi cells. Moreover, EC-derived CSF-1 facilitated recovery of Ly6C- monocytes and protected mice from weight loss following induction of polymicrobial sepsis. Thus, monocytes are supported by distinct cellular sources of CSF-1 within a perivascular BM niche.


Assuntos
Fator Estimulador de Colônias de Macrófagos , Células-Tronco Mesenquimais , Animais , Medula Óssea , Células da Medula Óssea , Células Endoteliais , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Monócitos
4.
Annu Rev Cell Dev Biol ; 30: 677-704, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25150008

RESUMO

Two opposing descriptions of so-called mesenchymal stem cells (MSCs) exist at this time. One sees MSCs as the postnatal, self-renewing, and multipotent stem cells for the skeleton. This cell coincides with a specific type of bone marrow perivascular cell. In skeletal physiology, this skeletal stem cell is pivotal to the growth and lifelong turnover of bone and to its native regeneration capacity. In hematopoietic physiology, its role as a key player in maintaining hematopoietic stem cells in their niche and in regulating the hematopoietic microenvironment is emerging. In the alternative description, MSCs are ubiquitous in connective tissues and are defined by in vitro characteristics and by their use in therapy, which rests on their ability to modulate the function of host tissues rather than on stem cell properties. Here, I discuss how the two views developed, conceptually and experimentally, and attempt to clarify the confusion arising from their collision.


Assuntos
Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/classificação , Células da Medula Óssea/citologia , Osso e Ossos/citologia , Antígeno CD146/análise , Separação Celular/métodos , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Células Clonais/citologia , Tecido Conjuntivo/imunologia , Humanos , Imunomodulação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/classificação , Camundongos , Modelos Biológicos , Pericitos/citologia , Células-Tronco Pluripotentes/citologia , Quimera por Radiação , Nicho de Células-Tronco , Células Estromais/classificação , Células Estromais/citologia , Transplante Heterotópico
5.
Immunity ; 48(4): 632-648, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29669248

RESUMO

Steady-state hematopoietic stem cells' (HSCs) self-renewal and differentiation toward their mature progeny in the adult bone marrow is tightly regulated by cues from the microenvironment. Recent insights into the cellular and molecular constituents have uncovered a high level of complexity. Here, we review emerging evidence showing how HSCs and their progeny are regulated by an interdependent network of mesenchymal stromal cells, nerve fibers, the vasculature, and also other hematopoietic cells. Understanding the interaction mechanisms in these intricate niches will provide great opportunities for HSC-related therapies and immune modulation.


Assuntos
Células da Medula Óssea/fisiologia , Autorrenovação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Nicho de Células-Tronco/fisiologia , Animais , Diferenciação Celular , Células-Tronco Hematopoéticas/fisiologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Fibras Nervosas/fisiologia
6.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38430523

RESUMO

Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche.


Assuntos
Linfócitos B , Medula Óssea , Quimiocina CXCL12 , Malária , Camundongos Endogâmicos C57BL , Animais , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/imunologia , Camundongos , Malária/imunologia , Malária/parasitologia , Linfócitos B/imunologia , Medula Óssea/imunologia , Medula Óssea/parasitologia , Nicho de Células-Tronco/imunologia , Interferon gama/metabolismo , Interferon gama/imunologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-38912739

RESUMO

Heparan sulfate proteoglycans are a family of glycoproteins that modulate cell signaling by binding growth factors and changing their bioavailability. Syndecans are a specific family of transmembrane heparan sulfate proteoglycans that regulate cell adhesion, migration, and signaling. In this review, we will summarize emerging evidence for the functions of Syndecans in the normal and malignant blood systems and their microenvironments. More specifically, we detail the known functions of Syndecans within normal hematopoietic stem cells. Further, we discuss the functions of Syndeans in hematological malignancies, including myeloid malignancies, lymphomas, and bleeding disorders. As normal and malignant hematopoietic cells require cues from their microenvironments to function, we also summarize the roles of Syndecans in cells of the stroma, endothelia, and osteolineage compartments. Syndecan biology is a rapidly evolving field; a comprehensive understanding of these molecules and their place in the hematopoietic system promises to improve our grasp on disease processes and better predict the efficacies of growth factor-targeting therapies.

8.
Br J Haematol ; 205(1): 175-188, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38736325

RESUMO

B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) blasts strictly depend on the transport of extra-cellular asparagine (Asn), yielding a rationale for L-asparaginase (ASNase) therapy. However, the carriers used by ALL blasts for Asn transport have not been identified yet. Exploiting RS4;11 cells as BCP-ALL model, we have found that cell Asn is lowered by either silencing or inhibition of the transporters ASCT2 or SNAT5. The inhibitors V-9302 (for ASCT2) and GluγHA (for SNAT5) markedly lower cell proliferation and, when used together, suppress mTOR activity, induce autophagy and cause a severe nutritional stress, leading to a proliferative arrest and a massive cell death in both the ASNase-sensitive RS4;11 cells and the relatively ASNase-insensitive NALM-6 cells. The cytotoxic effect is not prevented by coculturing leukaemic cells with primary mesenchymal stromal cells. Leukaemic blasts of paediatric ALL patients express ASCT2 and SNAT5 at diagnosis and undergo marked cytotoxicity when exposed to the inhibitors. ASCT2 expression is positively correlated with the minimal residual disease at the end of the induction therapy. In conclusion, ASCT2 and SNAT5 are the carriers exploited by ALL cells to transport Asn, and ASCT2 expression is associated with a lower therapeutic response. ASCT2 may thus represent a novel therapeutic target in BCP-ALL.


Assuntos
Sistema ASC de Transporte de Aminoácidos , Asparagina , Sobrevivência Celular , Antígenos de Histocompatibilidade Menor , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sistema ASC de Transporte de Aminoácidos/genética , Asparagina/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Antígenos de Histocompatibilidade Menor/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Sobrevivência Celular/efeitos dos fármacos , Sistema A de Transporte de Aminoácidos/metabolismo , Sistema A de Transporte de Aminoácidos/genética , Linhagem Celular Tumoral , Asparaginase/farmacologia , Asparaginase/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Criança
9.
Stem Cells ; 41(10): 944-957, 2023 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-37465968

RESUMO

Signal transducer and activator of transcription 5 (STAT5a and STAT5b) are intrinsically critical for normal hematopoiesis but are also expressed in stromal cells. Here, STAT5ab knockout (KO) was generated with a variety of bone marrow hematopoietic and stromal Cre transgenic mouse strains. Vav1-Cre/+STAT5abfl/fl, the positive control for loss of multipotent hematopoietic function, surprisingly dysregulated niche factor mRNA expression, and deleted STAT5ab in CD45neg cells. Single-cell transcriptome analysis of bone marrow from Vav1-Cre/+ wild-type or Vav1-Cre/+STAT5abfl/fl mice showed hematopoietic stem cell (HSC) myeloid commitment priming. Nes+ cells were detected in both CD45neg and CD45+ clusters and deletion of STAT5ab with Nes-Cre caused hematopoietic repopulating defects. To follow up on these promiscuous Cre promoter deletions in CD45neg and CD45+ bone marrow cell populations, more stroma-specific Cre strains were generated and demonstrated a reduction in multipotent hematopoietic progenitors. Functional support for niche-supporting activity was assessed using STAT5-deficient mesenchymal stem cells (MSCs). With Lepr-Cre/+STAT5abfl/fl, niche factor mRNAs were downregulated with validation of reduced IGF-1 and CXCL12 proteins. Furthermore, advanced computational analyses revealed a key role for STAT5ab/Cish balance with Cish strongly co-expressed in MSCs and HSCs primed for differentiation. Therefore, STAT5ab-associated gene regulation supports the bone marrow microenvironment.


Assuntos
Hematopoese , Fator de Transcrição STAT5 , Camundongos , Animais , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Camundongos Knockout , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Medula Óssea/metabolismo , Camundongos Transgênicos , Nicho de Células-Tronco/fisiologia
10.
Ann Hematol ; 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38684510

RESUMO

Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion.

11.
Immunol Rev ; 296(1): 87-103, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32592168

RESUMO

Plasma cells (PC) are key to protective immunity because they secrete antibodies. Surviving for periods ranging from days to decades in mammals, PC possess varying survival times that cannot be entirely stochastic or extrinsically set, as presumed half-lives vary with antigenic specificity. Here, we review the signals that impart survival potential to PC. These include signals provided during formation, and signals experienced once generated and embedded in the so-called long-lived niche. These signals all feed into survival by maintaining PC expression of MCL1, potentially synergistically with influences of other BCL2 family members. Herein, we propose that each formed PC has a capacity to respond to extrinsic cues that sets an upper maximum to its lifespan, but survival is also affected by variable availability of signals provided in BM survival niches. PC survival thus becomes a function of immunogen characteristics and niche anatomy, determined by the weighted survival benefit ascribed to each involved factor. Most factors, such as supporting cell types and secreted proteins, are predicted to influence survival times varying temporally by orders of magnitude, rather than absolute PC abundances measured at a single time, which may account for the variation in PC lifespan evident in the literature.


Assuntos
Imunidade Humoral , Memória Imunológica , Imunomodulação , Plasmócitos/imunologia , Plasmócitos/metabolismo , Animais , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Microambiente Celular/imunologia , Humanos , Imunomodulação/genética
12.
Br J Haematol ; 202(2): 294-307, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36890790

RESUMO

Mechanisms of prolonged cytopenia (PC) after chimeric antigen receptor (CAR) T-cell therapy, an emerging therapy for relapsed or refractory diffuse large B-cell lymphoma, remain elusive. Haematopoiesis is tightly regulated by the bone marrow (BM) microenvironment, called the 'niche'. To investigate whether alterations in the BM niche cells are associated with PC, we analysed CD271+ stromal cells in BM biopsy specimens and the cytokine profiles of the BM and serum obtained before and on day 28 after CAR T-cell infusion. Imaging analyses of the BM biopsy specimens revealed that CD271+ niche cells were severely impaired after CAR T-cell infusion in patients with PC. Cytokine analyses after CAR T-cell infusion showed that CXC chemokine ligand 12 and stem cell factor, niche factors essential for haematopoietic recovery, were significantly decreased in the BM of patients with PC, suggesting reduced niche cell function. The levels of inflammation-related cytokines on day 28 after CAR T-cell infusion were consistently high in the BM of patients with PC. Thus, we demonstrate for the first time that BM niche disruption and sustained elevation of inflammation-related cytokines in the BM following CAR T-cell infusion are associated with subsequent PC.


Assuntos
Leucopenia , Linfoma Difuso de Grandes Células B , Linfoma não Hodgkin , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Medula Óssea , Linfoma Difuso de Grandes Células B/terapia , Citocinas , Antígenos CD19 , Microambiente Tumoral
13.
Cell Commun Signal ; 21(1): 277, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37817179

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is an aggressive hematological malignancy, associated with unfavorable patient outcome, primarily due to disease relapse. Mesenchymal stem cells (MSCs) residing in the bone marrow (BM) niche are the source of mesenchyma-derived subpopulations, including adipocytes, and osteocytes, that are critical for normal hematopoiesis. This study aimed to characterize BM-derived adipocyte/osteocyte fractions and their crosstalk with AML cells as a potential mechanism underlying leukemogenesis. METHODS: BM cell subpopulations derived from primary AML patients were evaluated using humanized ex-vivo and in-vivo models, established for this study. The models comprised AML blasts, normal hematopoietic stem and progenitor cells and mesenchymal stromal subpopulations. ELISA, FACS analysis, colony forming unit assay, whole exome sequencing and real-time qPCR were employed to assess the differentiation capacity, genetic status, gene expression and function of these cell fractions. To explore communication pathways between AML cells and BM subpopulations, levels of signaling mediators, including cytokines and chemokines, were measured using the ProcartaPlex multiplex immunoassay. RESULTS: The study revealed deficiencies in adipogenic/osteogenic differentiation of BM-MSCs derived from AML patients, with adipocytes directly promoting survival and clonogenicity of AML cells in-vitro. In whole exome sequencing of BM-MSC/stromal cells, the AHNAK2 gene, associated with the stimulation of adipocyte differentiation, was found to be mutated and significantly under-expressed, implying its abnormal function in AML. The evaluation of communication pathways between AML cells and BM subpopulations demonstrated pronounced alterations in the crosstalk between these cell fractions. This was reflected by significantly elevated levels of signaling mediators cytokines/chemokines, in AML-induced adipocytes/osteocytes compared to non-induced MSCs, indicating abnormal hematopoiesis. Furthermore, in-vivo experiments using a fully humanized 3D scaffold model, showed that AML-induced adipocytes were the dominant component of the tumor microenvironment, providing preferential support to leukemia cell survival and proliferation. CONCLUSIONS: This study has disclosed direct contribution of impaired functional, genetic and molecular properties of AML patient-derived adipocytes to effective protection of AML blasts from apoptosis and to stimulation of their growth in vitro and in vivo, which overall leads to disease propagation and relapse. The detected AHNAK2 gene mutations in AML-MSCs point to their involvement in the mechanism underlying abnormal adipogenesis. Video Abstract.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Mesenquimais , Humanos , Adipogenia , Osteogênese , Leucemia Mieloide Aguda/metabolismo , Células da Medula Óssea/patologia , Citocinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Quimiocinas/metabolismo , Recidiva , Proliferação de Células , Microambiente Tumoral
14.
Drug Resist Updat ; 64: 100853, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35870226

RESUMO

Protein biogenesis, maturation and degradation are tightly regulated processes that are governed by a complex network of signaling pathways. The endoplasmic reticulum (ER) is responsible for biosynthesis and maturation of secretory proteins. Circumstances that alter cellular protein homeostasis, determine accumulation of misfolded and unfolded proteins in the ER, a condition defined as ER stress. In case of stress, the ER activates an adaptive response called unfolded protein response (UPR), a series of pathways of major relevance for cancer biology. The UPR plays a preeminent role in adaptation of tumor cells to the harsh conditions that they experience, due to high rates of proliferation, metabolic abnormalities and hostile environment scarce in oxygen and nutrients. Furthermore, the UPR is among the main adaptive cell stress responses contributing to the development of resistance to drugs and chemotherapy. Clinical management of Acute Myeloid Leukemia (AML) has improved significantly in the last decade, thanks to development of molecular targeted therapies. However, the emergence of treatment-resistant clones renders the rate of AML cure dismal. Moreover, different cell populations that constitute the bone marrow niche recently emerged as a main determinant leading to drug resistance. Herein we summarize the most relevant literature regarding the role played by the UPR in expansion of AML and ability to develop drug resistance and we discuss different possible modalities to overturn this adaptive response against leukemia. To this aim, we also describe the interconnection of the UPR with other cellular stress responses regulating protein homeostasis. Finally, we review the newest findings about the crosstalk between AML cells and cells of the bone marrow niche, under physiological conditions and in response to therapies, discussing in particular the importance of the niche in supporting survival of AML cells by favoring protein homeostasis.


Assuntos
Leucemia Mieloide Aguda , Resposta a Proteínas não Dobradas , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Homeostase , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Oxigênio/metabolismo , Resultado do Tratamento
15.
Rinsho Ketsueki ; 64(9): 861-868, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37793859

RESUMO

Hematopoietic stem and progenitor cells in mammals primarily reside in the bone marrow after birth. There, the cellular dynamics and subsequent fate of those cells are regulated by the adjacent microenvironment, known as the niche, to sustain lifelong blood cell production. To analyze and study physiological hematopoiesis and various hematopoietic disorders, it is essential to deeply understand how the niche regulates hematopoiesis and how niche dysregulation occurs. However, the dynamics of hematopoietic stem and progenitor cells and their interactions with the niche are dynamic and complex, and our knowledge of the spatial organization of bone marrow cells and niche factors is still limited. In this review, I provide an overview of classical techniques for spatiotemporal understanding of the cellular communities in bone marrow, as well as recent advances in bone marrow imaging techniques and valuable animal models, and discuss future prospects in this field.


Assuntos
Medula Óssea , Células-Tronco Hematopoéticas , Animais , Medula Óssea/diagnóstico por imagem , Nicho de Células-Tronco/fisiologia , Células da Medula Óssea , Hematopoese/fisiologia , Mamíferos
16.
Br J Haematol ; 199(5): 647-664, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35837798

RESUMO

Our view on the role and composition of the bone marrow (BM) has dramatically changed over time from a simple nutrient for the bone to a highly complex multicellular tissue that sustains haematopoiesis. Among these cells, multipotent haematopoietic stem cells (HSCs), which are predominantly quiescent, possess unique self-renewal capacity and multilineage differentiation potential and replenish all blood lineages to maintain lifelong haematopoiesis. Adult HSCs reside in specialised BM niches, which support their functions. Much effort has been put into deciphering HSC niches due to their potential clinical relevance. Multiple cell types have been implicated as HSC-niche components including sinusoidal endothelium, perivascular stromal cells, macrophages, megakaryocytes, osteoblasts and sympathetic nerves. In this review we provide a historical perspective on how technical advances, from genetic mouse models to imaging and high-throughput sequencing techniques, are unveiling the plethora of molecular cues and cellular components that shape the niche and regulate HSC functions.


Assuntos
Medula Óssea , Nicho de Células-Tronco , Camundongos , Animais , Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hematopoese , Diferenciação Celular
17.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35163048

RESUMO

Haematopoietic stem cells (HSCs) reside in the bone marrow and are supported by the specialised microenvironment, a niche to maintain HSC quiescence. To deal with haematopoietic equilibrium disrupted during inflammation, HSCs are activated from quiescence directly and indirectly to generate more mature immune cells, especially the myeloid lineage cells. In the process of proliferation and differentiation, HSCs gradually lose their self-renewal potential. The extensive inflammation might cause HSC exhaustion/senescence and malignant transformation. Here, we summarise the current understanding of how HSC functions are maintained, damaged, or exhausted during acute, prolonged, and pathological inflammatory conditions. We also highlight the inflammation-altered HSC niche and its impact on escalating the insults on HSCs.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Inflamação/patologia , Nicho de Células-Tronco , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco Hematopoéticas/patologia , Humanos , Inflamação/metabolismo , Transdução de Sinais
18.
Int J Mol Sci ; 24(1)2022 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-36614005

RESUMO

Acute myeloid leukaemia (AML) is a heterogeneous disease with one of the worst survival rates of all cancers. The bone marrow microenvironment is increasingly being recognised as an important mediator of AML chemoresistance and relapse, supporting leukaemia stem cell survival through interactions among stromal, haematopoietic progenitor and leukaemic cells. Traditional therapies targeting leukaemic cells have failed to improve long term survival rates, and as such, the bone marrow niche has become a promising new source of potential therapeutic targets, particularly for relapsed and refractory AML. This review briefly discusses the role of the bone marrow microenvironment in AML development and progression, and as a source of novel therapeutic targets for AML. The main focus of this review is on drugs that modulate/target this bone marrow microenvironment and have been examined in in vivo models or clinically.


Assuntos
Sistema Hematopoético , Leucemia Mieloide Aguda , Humanos , Medula Óssea , Recidiva Local de Neoplasia , Leucemia Mieloide Aguda/tratamento farmacológico , Células da Medula Óssea , Microambiente Tumoral
19.
Int J Mol Sci ; 23(4)2022 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-35216355

RESUMO

It is now well understood that the bone marrow (BM) compartment can sense systemic inflammatory signals and adapt through increased proliferation and lineage skewing. These coordinated and dynamic alterations in responding hematopoietic stem and progenitor cells (HSPCs), as well as in cells of the bone marrow niche, are increasingly viewed as key contributors to the inflammatory response. Growth factors, cytokines, metabolites, microbial products, and other signals can cause dysregulation across the entire hematopoietic hierarchy, leading to lineage-skewing and even long-term functional adaptations in bone marrow progenitor cells. These alterations may play a central role in the chronicity of disease as well as the links between many common chronic disorders. The possible existence of a form of "memory" in bone marrow progenitor cells is thought to contribute to innate immune responses via the generation of trained immunity (also called innate immune memory). These findings highlight how hematopoietic progenitors dynamically adapt to meet the demand for innate immune cells and how this adaptive response may be beneficial or detrimental depending on the context. In this review, we will discuss the role of bone marrow progenitor cells and their microenvironment in shaping the scope and scale of the immune response in health and disease.


Assuntos
Medula Óssea/patologia , Células-Tronco Hematopoéticas/patologia , Inflamação/patologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/patologia , Animais , Doença Crônica , Humanos , Imunidade Inata/fisiologia
20.
Biochem Biophys Res Commun ; 534: 843-848, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33183761

RESUMO

Multiple myeloma (MM) is an intractable hematological malignancy characterized by abnormal plasma cells in the bone marrow (BM) and increased osteolytic lesions. Within the BM niche, mesenchymal stem cells (MSCs) have been proposed to contribute to functionally important MM-MSC interactions. However, despite various studies on MM pathology, the impact of MM on MSCs during the early stages of malignancy has not been adequately addressed. We previously identified tissue inhibitor of matrix metalloproteinase 1 (TIMP-1) as a cytokine that is modulated in vivo within the MM BM niche, and highlighted its potential relevance in MM. Given the role of TIMP-1 in preventing migration of breast cancer cells, this study aimed to investigate the relationship between MSC-secreted TIMP-1 and MM progression. Here, we examined the effect of MSC-derived TIMP-1 on MM cell migration, and found that TIMP-1 secreted by human MSCs play a role in preventing migration of MM cells by reducing the levels of MM cell-derived MMP-9. We also investigated how MM cells regulate expression of TIMP-1 in MSCs. Using a knockdown approach in MSCs, we implicated TGF-B activated kinase 1 binding protein 1 (TAB1) as an upstream effector of TIMP-1 that was downregulated in the presence of MM cells, which resulted in reduced TIMP-1 secretion. Overall, our findings uncover how MSCs in the MM BM niche are modulated to promote MM progression, and unravel a previously unreported role of the TAB1-TIMP-1 axis in the context of the MM BM niche.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células-Tronco Mesenquimais/patologia , Mieloma Múltiplo/patologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Inibidor Tecidual de Metaloproteinase-1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA