Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
2.
Cell Stem Cell ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38772377

RESUMO

Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.

3.
Elife ; 122024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38573813

RESUMO

Metabolic pathways are plastic and rapidly change in response to stress or perturbation. Current metabolic profiling techniques require lysis of many cells, complicating the tracking of metabolic changes over time after stress in rare cells such as hematopoietic stem cells (HSCs). Here, we aimed to identify the key metabolic enzymes that define differences in glycolytic metabolism between steady-state and stress conditions in murine HSCs and elucidate their regulatory mechanisms. Through quantitative 13C metabolic flux analysis of glucose metabolism using high-sensitivity glucose tracing and mathematical modeling, we found that HSCs activate the glycolytic rate-limiting enzyme phosphofructokinase (PFK) during proliferation and oxidative phosphorylation (OXPHOS) inhibition. Real-time measurement of ATP levels in single HSCs demonstrated that proliferative stress or OXPHOS inhibition led to accelerated glycolysis via increased activity of PFKFB3, the enzyme regulating an allosteric PFK activator, within seconds to meet ATP requirements. Furthermore, varying stresses differentially activated PFKFB3 via PRMT1-dependent methylation during proliferative stress and via AMPK-dependent phosphorylation during OXPHOS inhibition. Overexpression of Pfkfb3 induced HSC proliferation and promoted differentiated cell production, whereas inhibition or loss of Pfkfb3 suppressed them. This study reveals the flexible and multilayered regulation of HSC glycolytic metabolism to sustain hematopoiesis under stress and provides techniques to better understand the physiological metabolism of rare hematopoietic cells.


Assuntos
Glicólise , Fosfofrutoquinase-2 , Animais , Camundongos , Trifosfato de Adenosina/metabolismo , Anaerobiose , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Fosforilação Oxidativa , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo
4.
Exp Hematol ; 135: 104191, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38493949

RESUMO

Erythropoiesis in the adult bone marrow relies on mitochondrial membrane transporters to facilitate heme and hemoglobin production. Erythrocytes in the bone marrow are produced although the differentiation of erythroid progenitor cells that originate from hematopoietic stem cells (HSCs). Whether and how mitochondria transporters potentiate HSCs and affect their differentiation toward erythroid lineage remains unclear. Here, we show that the ATP-binding cassette (ABC) transporter 10 (Abcb10), located on the inner mitochondrial membrane, is essential for HSC maintenance and erythroid-lineage differentiation. Induced deletion of Abcb10 in adult mice significantly increased erythroid progenitor cell and decreased HSC number within the bone marrow (BM). Functionally, Abcb10-deficient HSCs exhibited significant decreases in stem cell potential but with a skew toward erythroid-lineage differentiation. Mechanistically, deletion of Abcb10 rendered HSCs with excess mitochondrial iron accumulation and oxidative stress yet without alteration in mitochondrial bioenergetic function. However, impaired hematopoiesis could not be rescued through the in vivo administration of a mitochondrial iron chelator or antioxidant to Abcb10-deficient mice. Abcb10-mediated mitochondrial iron transfer is thus pivotal for the regulation of physiologic HSC potential and erythroid-lineage differentiation.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Diferenciação Celular , Eritropoese , Células-Tronco Hematopoéticas , Camundongos Knockout , Mitocôndrias , Animais , Camundongos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Mitocôndrias/metabolismo , Eritropoese/genética , Ferro/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Estresse Oxidativo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citologia , Camundongos Endogâmicos C57BL
5.
Proc Natl Acad Sci U S A ; 120(32): e2206860120, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37523546

RESUMO

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.


Assuntos
Medula Óssea , Células-Tronco Hematopoéticas , Animais , Camundongos , Ciclo Celular/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/metabolismo
6.
Exp Hematol ; 124: 56-67, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37339713

RESUMO

In physiological conditions, most adult hematopoietic stem cells (HSCs) maintain a quiescent state. Glycolysis is a metabolic process that can be divided into preparatory and payoff phases. Although the payoff phase maintains HSC function and properties, the role of the preparatory phase remains unknown. In this study, we aimed to investigate whether the preparatory or payoff phases of glycolysis were required for maintenance of quiescent and proliferative HSCs. We used glucose-6-phosphate isomerase (Gpi1) as a representative gene for the preparatory phase and glyceraldehyde-3-phosphate dehydrogenase (Gapdh) as a representative gene for the payoff phase of glycolysis. First, we identified that stem cell function and survival were impaired in Gapdh-edited proliferative HSCs. Contrastingly, cell survival was maintained in quiescent Gapdh- and Gpi1-edited HSCs. Gapdh- and Gpi1-defective quiescent HSCs maintained adenosine-triphosphate (ATP) levels by increasing mitochondrial oxidative phosphorylation (OXPHOS), whereas ATP levels were decreased in Gapdh-edited proliferative HSCs. Interestingly, Gpi1-edited proliferative HSCs maintained ATP levels independent of increased OXPHOS. Oxythiamine, a transketolase inhibitor, impaired proliferation of Gpi1-edited HSCs, suggesting that the nonoxidative pentose phosphate pathway (PPP) is an alternative means to maintain glycolytic flux in Gpi1-defective HSCs. Our findings suggest that OXPHOS compensated for glycolytic deficiencies in quiescent HSCs, and that in proliferative HSCs, nonoxidative PPP compensated for defects in the preparatory phase of glycolysis but not for defects in the payoff phase. These findings provide new insights into regulation of HSC metabolism, which could have implications for development of novel therapies for hematologic disorders.


Assuntos
Glicólise , Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Glicólise/genética , Fosforilação Oxidativa , Via de Pentose Fosfato/genética , Trifosfato de Adenosina/metabolismo
7.
Front Oncol ; 13: 1108430, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37007148

RESUMO

Fanconi Anemia (FA) is an inherited bone marrow (BM) failure disorder commonly diagnosed during school age. However, in murine models, disrupted function of FA genes leads to a much earlier decline in fetal liver hematopoietic stem cell (FL HSC) number that is associated with increased replication stress (RS). Recent reports have shown mitochondrial metabolism and clearance are essential for long-term BM HSC function. Intriguingly, impaired mitophagy has been reported in FA cells. We hypothesized that RS in FL HSC impacts mitochondrial metabolism to investigate fetal FA pathophysiology. Results show that experimentally induced RS in adult murine BM HSCs evoked a significant increase in mitochondrial metabolism and mitophagy. Reflecting the physiological RS during development in FA, increase mitochondria metabolism and mitophagy were observed in FANCD2-deficient FL HSCs, whereas BM HSCs from adult FANCD2-deficient mice exhibited a significant decrease in mitophagy. These data suggest that RS activates mitochondrial metabolism and mitophagy in HSC.

8.
Cell Rep Methods ; 2(12): 100354, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36590688

RESUMO

Other than genetically engineered mice, few reliable platforms are available for the study of hematopoietic stem cell (HSC) quiescence. Here we present a platform to analyze HSC cell cycle quiescence by combining culture conditions that maintain quiescence with a CRISPR-Cas9 genome editing system optimized for HSCs. We demonstrate that preculture of HSCs enhances editing efficiency by facilitating nuclear transport of ribonucleoprotein complexes. For post-editing culture, mouse and human HSCs edited based on non-homologous end joining and cultured under low-cytokine, low-oxygen, and high-albumin conditions retain their phenotypes and quiescence better than those cultured under the proliferative conditions. Using this approach, HSCs regain quiescence even after editing by homology-directed repair. Our results show that low-cytokine culture conditions for gene-edited HSCs are a useful approach for investigating HSC quiescence ex vivo.


Assuntos
Edição de Genes , Células-Tronco Hematopoéticas , Animais , Camundongos , Humanos , Edição de Genes/métodos , Citocinas/metabolismo
9.
Rinsho Ketsueki ; 62(5): 521-527, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34248130

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas , Trombopoetina , Ciclo Celular , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Mitocôndrias , Receptores de Trombopoetina
10.
Br J Haematol ; 193(6): 1260-1274, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34036571

RESUMO

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.


Assuntos
Diferenciação Celular , Eritroblastos/metabolismo , Macrófagos/metabolismo , Mitocôndrias/transplante , Nanotubos/química , Nicho de Células-Tronco , Animais , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo
11.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33924874

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Lisossomos/metabolismo , Renovação Mitocondrial , Anaerobiose , Animais , Diferenciação Celular , Estrona/metabolismo , Glicólise , Humanos , Tamanho Mitocondrial , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo
12.
Blood Adv ; 5(6): 1594-1604, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33710340

RESUMO

Hematopoietic stem cells (HSCs) undergo self-renewal or differentiation to sustain lifelong hematopoiesis. HSCs are preserved in quiescence with low mitochondrial activity. Recent studies indicate that autophagy contributes to HSC quiescence through suppressing mitochondrial metabolism. However, it remains unclear whether autophagy is involved in the regulation of neonatal HSCs, which proliferate actively. In this study, we clarified the role of autophagy in neonatal HSCs using 2 types of autophagy-related gene 7 (Atg7)-conditional knockout mice: Mx1-Cre inducible system and Vav-Cre system. Atg7-deficient HSCs exhibited excess cell divisions with enhanced mitochondrial metabolism, leading to bone marrow failure at adult stage. However, Atg7 deficiency minimally affected hematopoiesis and metabolic state in HSCs at neonatal stage. In addition, Atg7-deficient neonatal HSCs exhibited long-term reconstructing activity, equivalent to wild-type neonatal HSCs. Taken together, autophagy is dispensable for stem cell function and hematopoietic homeostasis in neonates and provide a novel aspect into the role of autophagy in the HSC regulation.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Animais , Autofagia , Transtornos da Insuficiência da Medula Óssea , Diferenciação Celular , Camundongos
13.
Blood ; 137(19): 2609-2620, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33657206

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/citologia , Trombopoetina/deficiência , Animais , Apoptose , Ciclo Celular , Autorrenovação Celular , Metabolismo Energético/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Receptores Fc , Receptores de Trombopoetina/agonistas , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais , Trombopoetina/genética , Trombopoetina/farmacologia , Transcriptoma
14.
Haematologica ; 106(7): 1883-1891, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32527954

RESUMO

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.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Trombopoetina , Animais , Contagem de Células , Hematopoese , Células-Tronco Hematopoéticas , Megacariócitos , Camundongos , Trombopoetina/farmacologia
15.
Rinsho Ketsueki ; 61(10): 1449-1458, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-33162439

RESUMO

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.


Assuntos
Trombopoetina/uso terapêutico , Medula Óssea , Diferenciação Celular , Células-Tronco Hematopoéticas , Humanos , Megacariócitos , Receptores de Trombopoetina
16.
Trends Cell Biol ; 30(11): 835-836, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32921525

RESUMO

The transplantation of healthy hematopoietic stem cells (HSCs) contained in the bone marrow is a frontline treatment option for hematopoietic diseases. Detailed analysis of post-transplant HSC kinetics is crucial as the initial engraftment of HSCs influences prognosis. Dong et al. have explored the dynamic change in HSC cell fate upon bone marrow transplantation through the utilization of single-cell transcriptomic analysis.


Assuntos
Transplante de Medula Óssea , Transplante de Células-Tronco Hematopoéticas , Diferenciação Celular , Células-Tronco Hematopoéticas , Humanos , Transcriptoma
17.
Dev Cell ; 54(2): 239-255, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32693057

RESUMO

Cellular metabolism in hematopoietic stem cells (HSCs) is an area of intense research interest, but the metabolic requirements of HSCs and their adaptations to their niches during development have remained largely unaddressed. Distinctive from other tissue stem cells, HSCs transition through multiple hematopoietic sites during development. This transition requires drastic metabolic shifts, insinuating the capacity of HSCs to meet the physiological demand of hematopoiesis. In this review, we highlight how mitochondrial metabolism determines HSC fate, and especially focus on the links between mitochondria, endoplasmic reticulum (ER), and lysosomes in HSC metabolism.


Assuntos
Envelhecimento , Diferenciação Celular/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Envelhecimento/fisiologia , Animais , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Humanos
18.
Blood ; 136(17): 1919-1932, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32573733

RESUMO

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.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Leucemia/genética , Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional/genética , Animais , Apoptose/genética , Sobrevivência Celular/genética , Família , Feminino , Predisposição Genética para Doença , Genótipo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patologia , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Síndromes Mielodisplásicas/sangue , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Linhagem
19.
Cell Rep ; 30(6): 1823-1834.e5, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32049013

RESUMO

The tumor suppressor folliculin (FLCN) suppresses nuclear translocation of TFE3, a master transcription factor for lysosomal biogenesis, via regulation of amino-acid-sensing Rag GTPases. However, the importance of this lysosomal regulation in mammalian physiology remains unclear. Following hematopoietic-lineage-specific Flcn deletion in mice, we found expansion of vacuolated phagocytes that accumulate glycogen in their cytoplasm, phenotypes reminiscent of lysosomal storage disorder (LSD). We report that TFE3 acts in a feedback loop to transcriptionally activate FLCN expression, and FLCN loss disrupts this loop, augmenting TFE3 activity. Tfe3 deletion in Flcn knockout mice reduces the number of phagocytes and ameliorates LSD-like phenotypes. We further reveal that TFE3 stimulates glycogenesis by promoting the expression of glycogenesis genes, including Gys1 and Gyg, upon loss of Flcn. Taken together, we propose that the FLCN-TFE3 feedback loop acts as a rheostat to control lysosome activity and prevents excessive glycogenesis and LSD-like phagocyte activation.


Assuntos
Lisossomos/metabolismo , Fagócitos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Humanos , Camundongos , Camundongos Knockout
20.
Ann N Y Acad Sci ; 1466(1): 51-58, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31292976

RESUMO

Thrombopoietin (Thpo) and its receptor myeloid proliferative leukemia (Mpl) were initially identified as the cytokine signaling that stimulates megakaryopoiesis and platelet production. However, Thpo-Mpl signaling has also been widely characterized as one of the few cytokine systems that directly regulates hematopoietic stem and progenitor cells. The ability of Thpo signaling to stimulate hematopoietic stem cell (HSC) self-renewal has led to the development and utilization of Thpo mimetic drugs to treat hematopoietic diseases with restricted function of HSCs, such as aplastic anemia. This review will cover the mechanisms by which Thpo-Mpl signaling regulates HSCs.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Trombopoetina/fisiologia , Animais , Autorrenovação Celular/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Receptores de Trombopoetina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Trombopoese/fisiologia , Trombopoetina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA