Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cell ; 177(7): 1679-1681, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31199915

RESUMEN

Baryawno et al. provide a comprehensive atlas of the mouse bone marrow stroma based on single-cell RNA-sequencing data. Their analysis reveals a taxonomy of 17 distinct cell types with diverse functions that highlights the complexity of the bone marrow stroma and paves the way for future in vivo assessment.


Asunto(s)
Médula Ósea , Leucemia , Animales , Células de la Médula Ósea , Homeostasis , Ratones , Análisis de Secuencia de ARN
2.
Proc Natl Acad Sci U S A ; 120(32): e2206860120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523546

RESUMEN

Mbtd1 (mbt domain containing 1) encodes a nuclear protein containing a zinc finger domain and four malignant brain tumor (MBT) repeats. We previously generated Mbtd1-deficient mice and found that MBTD1 is highly expressed in fetal hematopoietic stem cells (HSCs) and sustains the number and function of fetal HSCs. However, since Mbtd1-deficient mice die soon after birth possibly due to skeletal abnormalities, its role in adult hematopoiesis remains unclear. To address this issue, we generated Mbtd1 conditional knockout mice and analyzed adult hematopoietic tissues deficient in Mbtd1. We observed that the numbers of HSCs and progenitors increased and Mbtd1-deficient HSCs exhibited hyperactive cell cycle, resulting in a defective response to exogenous stresses. Mechanistically, we found that MBTD1 directly binds to the promoter region of FoxO3a, encoding a forkhead protein essential for HSC quiescence, and interacts with components of TIP60 chromatin remodeling complex and other proteins involved in HSC and other stem cell functions. Restoration of FOXO3a activity in Mbtd1-deficient HSCs in vivo rescued cell cycle and pool size abnormalities. These findings indicate that MBTD1 is a critical regulator for HSC pool size and function, mainly through the maintenance of cell cycle quiescence by FOXO3a.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Ratones , Ciclo Celular/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/metabolismo
3.
Rinsho Ketsueki ; 65(9): 872-877, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-39358285

RESUMEN

Sustaining lifelong hematopoiesis requires maintenance, proliferation, and differentiation of hematopoietic stem cells. Thrombopoietin is a cytokine essential for regulation of hematopoietic stem cells as well as differentiation and maturation of megakaryocytes required for platelet production. Due to these properties, thrombopoietin agonists have been used to treat bone marrow failure syndromes such as aplastic anemia. Through analysis of thrombopoietin gene-deficient mice, my colleagues and I have demonstrated the mechanism of action of thrombopoietin receptor agonists in hematopoietic stem cell maintenance and differentiation. This review focuses on governance of homeostasis in the hematopoietic system by thrombopoietin signaling.


Asunto(s)
Células Madre Hematopoyéticas , Receptores de Trombopoyetina , Transducción de Señal , Trombopoyetina , Trombopoyetina/metabolismo , Trombopoyetina/farmacología , Animales , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Receptores de Trombopoyetina/metabolismo , Receptores de Trombopoyetina/agonistas , Diferenciación Celular
4.
Blood ; 137(19): 2609-2620, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33657206

RESUMEN

Hematopoietic stem cells (HSC) rarely divide, rest in quiescence, and proliferate only upon stress hematopoiesis. The cytokine thrombopoietin (Thpo) has been perplexingly described to induce quiescence and promote self-renewal divisions in HSCs. To clarify the contradictory effect of Thpo, we conducted a detailed analysis on conventional (Thpo-/-) and liver-specific (Thpofl/fl;AlbCre+/-) Thpo-deletion models. Thpo-/- HSCs exhibited profound loss of quiescence, impaired cell cycle progression, and increased apoptosis. Thpo-/- HSCs also exhibited diminished mitochondrial mass and impaired mitochondrial bioenergetics. Abnormal HSC phenotypes in Thpo-/- mice were reversible after HSC transplantation into wild-type recipients. Moreover, Thpo-/- HSCs acquired quiescence with extended administration of a Thpo receptor agonist, romiplostim, and were prone to subsequent stem cell exhaustion during competitive bone marrow transplantation. Thpofl/fl;AlbCre+/- HSCs exhibited similar stem cell phenotypes but to a lesser degree compared with Thpo-/- HSCs. HSCs that survive Thpo deficiency acquire quiescence in a dose-dependent manner through the modification of their metabolic state.


Asunto(s)
Células Madre Hematopoyéticas/citología , Trombopoyetina/deficiencia , Animales , Apoptosis , Ciclo Celular , Autorrenovación de las Células , Metabolismo Energético/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores Fc , Receptores de Trombopoyetina/agonistas , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal , Trombopoyetina/genética , Trombopoyetina/farmacología , Transcriptoma
5.
Blood ; 136(17): 1919-1932, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32573733

RESUMEN

RUNX1 is among the most frequently mutated genes in human leukemia, and the loss or dominant-negative suppression of RUNX1 function is found in myelodysplastic syndrome and acute myeloid leukemia (AML). How posttranslational modifications (PTMs) of RUNX1 affect its in vivo function, however, and whether PTM dysregulation of RUNX1 can cause leukemia are largely unknown. We performed targeted deep sequencing on a family with 3 occurrences of AML and identified a novel RUNX1 mutation, R237K. The mutated R237 residue is a methylation site by protein arginine methyltransferase 1, and loss of methylation reportedly impairs the transcriptional activity of RUNX1 in vitro. To explore the biologic significance of RUNX1 methylation in vivo, we used RUNX1 R233K/R237K double-mutant mice, in which 2 arginine-to-lysine mutations precluded RUNX1 methylation. Genetic ablation of RUNX1 methylation led to loss of quiescence and expansion of hematopoietic stem cells (HSCs), and it changed the genomic and epigenomic signatures of phenotypic HSCs to a poised progenitor state. Furthermore, loss of RUNX1 R233/R237 methylation suppressed endoplasmic reticulum stress-induced unfolded protein response genes, including Atf4, Ddit3, and Gadd34; the radiation-induced p53 downstream genes Bbc3, Pmaip1, and Cdkn1a; and subsequent apoptosis in HSCs. Mechanistically, activating transcription factor 4 was identified as a direct transcriptional target of RUNX1. Collectively, defects in RUNX1 methylation in HSCs confer resistance to apoptosis and survival advantage under stress conditions, a hallmark of a preleukemic clone that may predispose affected individuals to leukemia. Our study will lead to a better understanding of how dysregulation of PTMs can contribute to leukemogenesis.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/fisiología , Leucemia/genética , Metiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional/genética , Animales , Apoptosis/genética , Supervivencia Celular/genética , Familia , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patología , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Mutación Missense , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Linaje
6.
EMBO J ; 36(16): 2390-2403, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28673932

RESUMEN

Hematopoietic homeostasis depends on the maintenance of hematopoietic stem cells (HSCs), which are regulated within a specialized bone marrow (BM) niche. When HSC sense external stimuli, their adhesion status may be critical for determining HSC cell fate. The cell surface molecule, integrin αvß3, is activated through HSC adhesion to extracellular matrix and niche cells. Integrin ß3 signaling maintains HSCs within the niche. Here, we showed the synergistic negative regulation of the pro-inflammatory cytokine interferon-γ (IFNγ) and ß3 integrin signaling in murine HSC function by a novel definitive phenotyping of HSCs. Integrin αvß3 suppressed HSC function in the presence of IFNγ and impaired integrin ß3 signaling mitigated IFNγ-dependent negative action on HSCs. During IFNγ stimulation, integrin ß3 signaling enhanced STAT1-mediated gene expression via serine phosphorylation. These findings show that integrin ß3 signaling intensifies the suppressive effect of IFNγ on HSCs, which indicates that cell adhesion via integrin αvß3 within the BM niche acts as a context-dependent signal modulator to regulate the HSC function under both steady-state and inflammatory conditions.


Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/fisiología , Integrina alfaVbeta3/metabolismo , Interferón gamma/metabolismo , Animales , Regulación de la Expresión Génica , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
7.
Br J Haematol ; 193(6): 1260-1274, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34036571

RESUMEN

Adult erythropoiesis entails a series of well-coordinated events that produce mature red blood cells. One of such events is the mitochondria clearance that occurs cell-autonomously via autophagy-dependent mechanisms. Interestingly, recent studies have shown mitochondria transfer activities between various cell types. In the context of erythropoiesis, macrophages are known to interact closely with the early stages of erythroblasts to provide a specialized niche, termed erythroblastic islands (EBI). However, whether mitochondria transfer can occur in the EBI niche has not been explored. Here, we report that mitochondria transfer in the EBI niche occurs in vivo. We observed mitochondria transfer activities from the early stages of erythroblasts to macrophages in the reconstituted in vitro murine EBI via different modes, including tunnelling nanotubes (TNT). Moreover, we demonstrated that Wiskott-Aldrich syndrome protein (WASp) in macrophages mediates TNT formation and mitochondria transfer via the modulation of F-actin filamentation, thus promoting mitochondria clearance from erythroid cells, to potentially enhance their differentiation. Taken together, our findings provide novel insight into the mitochondria clearance machineries that mediate erythroid maturation.


Asunto(s)
Diferenciación Celular , Eritroblastos/metabolismo , Macrófagos/metabolismo , Mitocondrias/trasplante , Nanotubos/química , Nicho de Células Madre , Animales , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo
8.
Haematologica ; 106(7): 1883-1891, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32527954

RESUMEN

Thrombopoietin (THPO) has long been known to influence megakaryopoiesis and hematopoietic stem and progenitor cells (HSPCs), though the exact mechanisms through which it acts are unknown. Here we show that MPL expression correlates with megakaryopoietic potential of HSPCs and identify a population of quiescent progenitor cells that show limited dependence on THPO signalling. We show that THPO is primarily responsible for maintenance of hematopoietic cells with megakaryocytic (Mk) differentiation potential and their subsequent Mk differentiation and maturation. The loss of Mks in THPO knockout (KO) mouse models results in a reduction of the Mk derived chemokine platelet factor 4 (CXCL4/PF4) in the bone marrow and administration of recombinant CXCL4/PF4 rescues the loss of progenitor cell quiescence observed in these mice. CXCL4/PF4 treatment does not rescue reduced HSPC numbers suggesting that thrombopoietin directly maintains HSPC numbers.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trombopoyetina , Animales , Recuento de Células , Hematopoyesis , Células Madre Hematopoyéticas , Megacariocitos , Ratones , Trombopoyetina/farmacología
9.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33924874

RESUMEN

Hematopoietic stem cells (HSCs) reside in a hypoxic microenvironment that enables glycolysis-fueled metabolism and reduces oxidative stress. Nonetheless, metabolic regulation in organelles such as the mitochondria and lysosomes as well as autophagic processes have been implicated as essential for the determination of HSC cell fate. This review encompasses the current understanding of anaerobic metabolism in HSCs as well as the emerging roles of mitochondrial metabolism and lysosomal regulation for hematopoietic homeostasis.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Lisosomas/metabolismo , Recambio Mitocondrial , Anaerobiosis , Animales , Diferenciación Celular , Estrona/metabolismo , Glucólisis , Humanos , Tamaño Mitocondrial , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo
10.
Rinsho Ketsueki ; 62(5): 521-527, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34248130

RESUMEN

Cell cycle quiescence is a fundamental property of hematopoietic stem cells (HSCs). Quiescent HSCs form a healthy pool of cells that serve as a reserve for massive HSC expansion under various conditions of stress. We previously reported that thrombopoietin (THPO) maintains quiescent HSCs and stimulates mitochondrial metabolism, megakaryocyte-lineage differentiation, and proliferation of HSCs. The underlying mechanism by which THPO balances its contradictory effect of promoting proliferation or quiescence on HSCs remains unknown. This review explores the role of THPO signaling in HSC differentiation and quiescence regulation. We present our data, which suggests that a THPO-independent HSC subpopulation sustaining a low mitochondrial metabolic profile reverts to quiescence and regains stem cell potential with external stimuli. There is a possibility that THPO-independent HSCs form a non-quiescent reserve HSC pool from which quiescent HSCs originate in the adult bone marrow.


Asunto(s)
Células Madre Hematopoyéticas , Trombopoyetina , Ciclo Celular , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Mitocondrias , Receptores de Trombopoyetina
11.
Rinsho Ketsueki ; 61(10): 1449-1458, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-33162439

RESUMEN

Thrombopoietin (Thpo) is a hematopoietic cytokine that regulates the production of megakaryocyte/platelet lineage cells and maintains hematopoietic stem and progenitor cells (HSPCs). While Thpo directly stimulates the proliferation of HSPCs, it also maintains HSCs in quiescence to form a reserve pool of HSCs in the bone marrow. Moreover, Thpo activates mitochondria and induces HSC differentiation to megakaryocyte/platelet lineage cells. Being void of instigating anti-Thpo antibody formation in vivo, the use of Thpo receptor agonists (Mpl agonists) transcends the use of recombinant Thpo in the treatment of immune thrombocytopenia. Since its invention, the therapeutic indication of Mpl agonists has extended to the treatment of bone marrow failure in aplastic anemia. As the clinical application of Mpl agonists expands, a detailed investigation of the function and effect of Mpl agonists on physiological HSCs and bone marrow failure is necessary.


Asunto(s)
Trombopoyetina/uso terapéutico , Médula Ósea , Diferenciación Celular , Células Madre Hematopoyéticas , Humanos , Megacariocitos , Receptores de Trombopoyetina
12.
Br J Haematol ; 185(5): 888-902, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30854628

RESUMEN

Inherited thrombocytopenia is a genetically heterogeneous disease characterized by varying degrees of thrombocytopenia and risk of haematological malignancy, and the genetic cause of many cases remains unknown. We performed whole-exome sequencing of a family with thrombocytopenia and myeloid malignancy and identified a novel TUBB1 variant, T149P. Screening of other thrombocytopenia pedigrees identified another TUBB1 variant, R251H. TUBB1 encodes the tubulin ß-1 chain, a major component of microtubules abundant in megakaryocytes. Variant TUBB1 disrupted the normal assembly of microtubules and impaired proplatelet formation in vitro. In addition, DNA damage response was severely attenuated by loss of TUBB1. We found that the nuclear accumulation of p53 (also termed TP53) and the expression of pro-apoptotic genes triggered by genotoxic stress were blocked in TUBB1-deficient cells and, accordingly, apoptosis after DNA damage was diminished by knockdown of TUBB1. Thus, we have demonstrated that microtubule dysfunction confers resistance to apoptosis, even in DNA damage-accumulated cells, which explains genome instability in the affected individuals. These studies will lead us to a better understanding of how microtubule dysfunction can contribute to the accumulation of DNA damage, genetic instability and leukaemogenesis.


Asunto(s)
Trombocitopenia/genética , Tubulina (Proteína)/genética , Anciano , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Inestabilidad Genómica , Mutación de Línea Germinal , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Linaje , Alineación de Secuencia , Trombocitopenia/patología , Proteína p53 Supresora de Tumor/genética , Secuenciación del Exoma
14.
Rinsho Ketsueki ; 59(1): 3-12, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29415934

RESUMEN

The frequency of clonal hematopoiesis in humans vastly increases with aging. Older adults may develop one or several clones and this condition is called clonal hematopoiesis of indeterminate potential (CHIP). Recent genetic analyses have identified the genes inducing CHIP. These mutant genes are detected frequently in elderly people and this condition is a precursor of hematopoietic neoplasms. The prevalence of hematopoietic neoplasms in patients with CHIP is tenfold that in those without CHIP. Consequently, the mechanism of aging and leukemogenesis of hematopoietic stem cells is being understood. Furthermore, the efficacy of senolysis, selectively removing scenescent cells from tissues, has been demonstrated in mice. The clinical application of senolysis is anticipated shortly.


Asunto(s)
Senescencia Celular , Células Madre Hematopoyéticas , Envejecimiento , Animales , Evolución Clonal , Hematopoyesis , Humanos
15.
Development ; 141(24): 4656-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25468935

RESUMEN

Tissue homeostasis requires the presence of multipotent adult stem cells that are capable of efficient self-renewal and differentiation; some of these have been shown to exist in a dormant, or quiescent, cell cycle state. Such quiescence has been proposed as a fundamental property of hematopoietic stem cells (HSCs) in the adult bone marrow, acting to protect HSCs from functional exhaustion and cellular insults to enable lifelong hematopoietic cell production. Recent studies have demonstrated that HSC quiescence is regulated by a complex network of cell-intrinsic and -extrinsic factors. In addition, detailed single-cell analyses and novel imaging techniques have identified functional heterogeneity within quiescent HSC populations and have begun to delineate the topological organization of quiescent HSCs. Here, we review the current methods available to measure quiescence in HSCs and discuss the roles of HSC quiescence and the various mechanisms by which HSC quiescence is maintained.


Asunto(s)
Ciclo Celular/fisiología , Fenómenos Fisiológicos Celulares/fisiología , Células Madre Hematopoyéticas/fisiología , Homeostasis/fisiología , Modelos Biológicos , Células Madre Multipotentes/fisiología , Animales , Humanos
16.
Stem Cells ; 34(4): 1068-82, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27095138

RESUMEN

Folliculin (FLCN) is an autosomal dominant tumor suppressor gene that modulates diverse signaling pathways required for growth, proliferation, metabolism, survival, motility, and adhesion. FLCN is an essential protein required for murine embryonic development, embryonic stem cell (ESC) commitment, and Drosophila germline stem cell maintenance, suggesting that Flcn may be required for adult stem cell homeostasis. Conditional inactivation of Flcn in adult hematopoietic stem/progenitor cells (HSPCs) drives hematopoietic stem cells (HSC) into proliferative exhaustion resulting in the rapid depletion of HSPC, loss of all hematopoietic cell lineages, acute bone marrow (BM) failure, and mortality after 40 days. HSC that lack Flcn fail to reconstitute the hematopoietic compartment in recipient mice, demonstrating a cell-autonomous requirement for Flcn in HSC maintenance. BM cells showed increased phosphorylation of Akt and mTorc1, and extramedullary hematopoiesis was significantly reduced by treating mice with rapamycin in vivo, suggesting that the mTorc1 pathway was activated by loss of Flcn expression in hematopoietic cells in vivo. Tfe3 was activated and preferentially localized to the nucleus of Flcn knockout (KO) HSPCs. Tfe3 overexpression in HSPCs impaired long-term hematopoietic reconstitution in vivo, recapitulating the Flcn KO phenotype, and supporting the notion that abnormal activation of Tfe3 contributes to the Flcn KO phenotype. Flcn KO mice develop an acute histiocytic hyperplasia in multiple organs, suggesting a novel function for Flcn in macrophage development. Thus, Flcn is intrinsically required to maintain adult HSC quiescence and homeostasis, and Flcn loss leads to BM failure and mortality in mice.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Diferenciación Celular/genética , Estrona/genética , Células Madre Hematopoyéticas/patología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células de la Médula Ósea/patología , Linaje de la Célula/genética , Proliferación Celular/genética , Desarrollo Embrionario/genética , Células Madre Hematopoyéticas/metabolismo , Homeostasis/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados
17.
Blood ; 124(22): 3179-80, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25414434

RESUMEN

In this issue of Blood, Wang et al elegantly show that telomere shortening results in DNA damage that induces apoptosis and senescence in quiescent hematopoietic stem cells (HSCs).


Asunto(s)
Apoptosis/fisiología , Senescencia Celular/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Acortamiento del Telómero/fisiología , Animales
18.
Blood ; 123(16): 2540-9, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24574458

RESUMEN

Just as normal stem cells require niche cells for survival, leukemia-initiating cells (LICs) may also require niche cells for their maintenance. Chronic myeloid leukemia (CML) is caused by the activity of BCR-ABL, a constitutively active tyrosine kinase. CML therapy with tyrosine kinase inhibitors is highly effective; however, due to the persistence of residual LICs, it is not curative. Several factors are known to support CML LICs, but purification of LICs and a thorough understanding of their niche signals have not yet been achieved. Using a CML-like mouse model of myeloproliferative disease, we demonstrate that CML LICs can be divided into CD25(+)FcεRIα(-) Lineage marker (Lin)(-) Sca-1(+)c-Kit(+) (F(-)LSK) cells and CD25(-)F(-)LSK cells. The CD25(+)F(-)LSK cells had multilineage differentiation capacity, with a preference toward cytokine-producing mast cell commitment. Although cells interconverted between CD25(-)F(-)LSK and CD25(+)F(-)LSK status, the CD25(+)F(-)LSK cells exhibited higher LIC capacity. Our findings suggest that interleukin-2 derived from the microenvironment and CD25 expressed on CML LICs constitute a novel signaling axis. The high levels of CD25 expression in the CD34(+)CD38(-) fraction of human CML cells indicate that CD25(+) LICs constitute an "LIC-derived niche" that could be preferentially targeted in therapy for CML.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-2/fisiología , Interleucina-2/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Animales , Proliferación Celular , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Transducción de Señal/fisiología , Células Th2/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
19.
Biochim Biophys Acta ; 1830(2): 2404-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22967757

RESUMEN

BACKGROUND: Hematopoietic stem cell (HSC) niche of the BM provides a specialized microenvironment for the regulation of HSCs. The strict control of HSCs by the niche coordinates the balance between the proliferation and the differentiation of HSCs for the homeostasis of the blood system in steady states and during stress hematopoiesis. The osteoblastic and vascular niches are the classically identified constituents of the BM niche. SCOPE OF REVIEW: Recent research broadens our understanding of the BM niche as an assembly of multiple niche cells within the BM. We provide an overview of the HSC niche aiming to delineate the defined and possible niche cell interactions which collectively modulate the HSC integrity. MAJOR CONCLUSIONS: Multiple cells in the BM, including osteoblasts, vascular endothelia, perivascular mesenchymal cells and HSC progeny cells, function conjunctively as niche cells to regulate HSCs. GENERAL SIGNIFICANCE: The study of HSC niche cells and their functions provides insights into stem cell biology and also may be extrapolated into the study of cancer stem cells. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Humanos
20.
Biochem Biophys Res Commun ; 454(2): 353-7, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25451253

RESUMEN

Tissue homeostasis demands regulatory feedback, suggesting that hematopoietic stem cell (HSC) activity is controlled in part by HSC progeny. Yet, cell extrinsic HSC regulation has been well characterized only in niche cells of non-hematopoietic origin. Here we identify feedback regulation of HSCs by megakaryocytes (Mks), which are mature hematopoietic cells, through production of thrombopoietin (Thpo), a cytokine pertinent for HSC maintenance. Induced ablation of Mk cell population in mice perturbed quiescent HSCs in bone marrow (BM). The ablation of Mks resulted in decreased intra-BM Thpo concentration presumably due to Thpo production by Mks. Thpo administration Mk ablated mice restored HSC functions. Overall, our study establishes Mk as an essential cellular component of the HSC niche and delineates cytokine-oriented regulation of HSCs by their own progeny.


Asunto(s)
Células Madre Hematopoyéticas/citología , Megacariocitos/metabolismo , Trombopoyetina/metabolismo , Animales , Trasplante de Médula Ósea , Ciclo Celular , Diferenciación Celular , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/citología , Ratones , Ratones Endogámicos C57BL , Nicho de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA