Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31442401

RESUMO

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Assuntos
Linfócitos B/fisiologia , Imunidade nas Mucosas , Animais , Antígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Jejum , Regulação da Expressão Gênica , Glicólise , Imunoglobulina A/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estado Nutricional , Ovalbumina/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/patologia , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Transdução de Sinais , Células Estromais/citologia , Células Estromais/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
Nature ; 595(7866): 266-271, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34163066

RESUMO

Obesity is a worldwide epidemic that predisposes individuals to many age-associated diseases, but its exact effects on organ dysfunction are largely unknown1. Hair follicles-mini-epithelial organs that grow hair-are miniaturized by ageing to cause hair loss through the depletion of hair follicle stem cells (HFSCs)2. Here we report that obesity-induced stress, such as that induced by a high-fat diet (HFD), targets HFSCs to accelerate hair thinning. Chronological gene expression analysis revealed that HFD feeding for four consecutive days in young mice directed activated HFSCs towards epidermal keratinization by generating excess reactive oxygen species, but did not reduce the pool of HFSCs. Integrative analysis using stem cell fate tracing, epigenetics and reverse genetics showed that further feeding with an HFD subsequently induced lipid droplets and NF-κB activation within HFSCs via autocrine and/or paracrine IL-1R signalling. These integrated factors converge on the marked inhibition of Sonic hedgehog (SHH) signal transduction in HFSCs, thereby further depleting lipid-laden HFSCs through their aberrant differentiation and inducing hair follicle miniaturization and eventual hair loss. Conversely, transgenic or pharmacological activation of SHH rescued HFD-induced hair loss. These data collectively demonstrate that stem cell inflammatory signals induced by obesity robustly represses organ regeneration signals to accelerate the miniaturization of mini-organs, and suggests the importance of daily prevention of organ dysfunction.


Assuntos
Alopecia/patologia , Alopecia/fisiopatologia , Folículo Piloso/patologia , Obesidade/fisiopatologia , Células-Tronco/patologia , Animais , Comunicação Autócrina , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Senescência Celular , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Proteínas Hedgehog/metabolismo , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , Estresse Oxidativo , Comunicação Parácrina , Receptores de Interleucina-1/metabolismo
3.
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
4.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34385317

RESUMO

The amino acid and oligopeptide transporter Solute carrier family 15 member A4 (SLC15A4), which resides in lysosomes and is preferentially expressed in immune cells, plays critical roles in the pathogenesis of lupus and colitis in murine models. Toll-like receptor (TLR)7/9- and nucleotide-binding oligomerization domain-containing protein 1 (NOD1)-mediated inflammatory responses require SLC15A4 function for regulating the mechanistic target of rapamycin complex 1 (mTORC1) or transporting L-Ala-γ-D-Glu-meso-diaminopimelic acid, IL-12: interleukin-12 (Tri-DAP), respectively. Here, we further investigated the mechanism of how SLC15A4 directs inflammatory responses. Proximity-dependent biotin identification revealed glycolysis as highly enriched gene ontology terms. Fluxome analyses in macrophages indicated that SLC15A4 loss causes insufficient biotransformation of pyruvate to the tricarboxylic acid cycle, while increasing glutaminolysis to the cycle. Furthermore, SLC15A4 was required for M1-prone metabolic change and inflammatory IL-12 cytokine productions after TLR9 stimulation. SLC15A4 could be in close proximity to AMP-activated protein kinase (AMPK) and mTOR, and SLC15A4 deficiency impaired TLR-mediated AMPK activation. Interestingly, SLC15A4-intact but not SLC15A4-deficient macrophages became resistant to fluctuations in environmental nutrient levels by limiting the use of the glutamine source; thus, SLC15A4 was critical for macrophage's respiratory homeostasis. Our findings reveal a mechanism of metabolic regulation in which an amino acid transporter acts as a gatekeeper that protects immune cells' ability to acquire an M1-prone metabolic phenotype in inflammatory tissues by mitigating metabolic stress.


Assuntos
Regulação da Expressão Gênica/fisiologia , Macrófagos/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Tecido Nervoso/metabolismo , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Células Dendríticas/metabolismo , Desoxiglucose/análogos & derivados , Desoxiglucose/metabolismo , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Macrófagos/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Oligodesoxirribonucleotídeos/farmacologia
5.
Blood ; 137(7): 908-922, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33174606

RESUMO

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Assuntos
Envelhecimento/genética , Regulação da Expressão Gênica/genética , Hematopoese/genética , Sistema Hematopoético/fisiologia , Código das Histonas/genética , Histona Desmetilases/fisiologia , Animais , Senescência Celular/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Feminino , Predisposição Genética para Doença , Hematopoese Extramedular , Histona Desmetilases/deficiência , Histona Desmetilases/genética , Reconstituição Imune , Histona Desmetilases com o Domínio Jumonji/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/virologia , Masculino , Camundongos , Camundongos Knockout , Vírus da Leucemia Murina de Moloney/fisiologia , Células Mieloides/patologia , Quimera por Radiação , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/metabolismo , Integração Viral
6.
PLoS Biol ; 18(9): e3000813, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32991574

RESUMO

Short-chain fatty acids (SCFAs) produced by gastrointestinal microbiota regulate immune responses, but host molecular mechanisms remain unknown. Unbiased screening using SCFA-conjugated affinity nanobeads identified apoptosis-associated speck-like protein (ASC), an adaptor protein of inflammasome complex, as a noncanonical SCFA receptor besides GPRs. SCFAs promoted inflammasome activation in macrophages by binding to its ASC PYRIN domain. Activated inflammasome suppressed survival of Salmonella enterica serovar Typhimurium (S. Typhimurium) in macrophages by pyroptosis and facilitated neutrophil recruitment to promote bacterial elimination and thus inhibit systemic dissemination in the host. Administration of SCFAs or dietary fibers, which are fermented to SCFAs by gut bacteria, significantly prolonged the survival of S. Typhimurium-infected mice through ASC-mediated inflammasome activation. SCFAs penetrated into the inflammatory region of the infected gut mucosa to protect against infection. This study provided evidence that SCFAs suppress Salmonella infection via inflammasome activation, shedding new light on the therapeutic activity of dietary fiber.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Ácidos Graxos Voláteis/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Infecções por Salmonella/prevenção & controle , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Feminino , Microbioma Gastrointestinal/imunologia , Células HEK293 , Humanos , Imunidade Inata/fisiologia , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Receptores Acoplados a Proteínas G/genética , Infecções por Salmonella/genética , Infecções por Salmonella/imunologia , Infecções por Salmonella/metabolismo , Salmonella typhimurium/imunologia , Células U937
7.
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
8.
J Biol Chem ; 296: 100563, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33745970

RESUMO

Hematopoietic stem cells (HSCs) and their progeny sustain lifetime hematopoiesis. Aging alters HSC function, number, and composition and increases risk of hematological malignancies, but how these changes occur in HSCs remains unclear. Signaling via p38 mitogen-activated kinase (p38MAPK) has been proposed as a candidate mechanism underlying induction of HSC aging. Here, using genetic models of both chronological and premature aging, we describe a multimodal role for p38α, the major p38MAPK isozyme in hematopoiesis, in HSC aging. We report that p38α regulates differentiation bias and sustains transplantation capacity of HSCs in the early phase of chronological aging. However, p38α decreased HSC transplantation capacity in the late progression phase of chronological aging. Furthermore, codeletion of p38α in mice deficient in ataxia-telangiectasia mutated, a model of premature aging, exacerbated aging-related HSC phenotypes seen in ataxia-telangiectasia mutated single-mutant mice. Overall, these studies provide new insight into multiple functions of p38MAPK, which both promotes and suppresses HSC aging context dependently.


Assuntos
Envelhecimento/patologia , Diferenciação Celular , Senescência Celular , Células-Tronco Hematopoéticas/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Envelhecimento/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Proliferação de Células , Feminino , Hematopoese , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Espécies Reativas de Oxigênio/metabolismo
9.
Biochem Biophys Res Commun ; 596: 29-35, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35108651

RESUMO

Thymic dendritic cells (DCs) promote immune tolerance by regulating negative selection of autoreactive T cells in the thymus. How DC homing to the thymus is transcriptionally regulated is still unclear. Microphthalmia-associated transcription factor (Mitf) is broadly expressed and plays essential roles in the hematopoietic system. Here, we used Mitf-mutated mice (Mitfvit/vit) and found enlargement of the thymus and expansion of CD4/CD8 double-positive T cells. Mitf was highly expressed in a subset of thymic DCs among the hematopoietic system. Genetic mutation or pharmacological inhibition of Mitf in DCs decreased the expression levels of Itga4, which are critical molecules for the homing of DCs to the thymus. Further, inhibition of Mitf decreased thymic DC number. These results suggest a pivotal role of Mitf in the maintenance of T cell differentiation by regulating the homing of DC subsets within the thymus.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Fator de Transcrição Associado à Microftalmia/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Células Dendríticas/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Hiperplasia , Integrina alfa4/genética , Integrina alfa4/imunologia , Integrina alfa4/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Timo/metabolismo , Timo/patologia
10.
Biochem Biophys Res Commun ; 514(1): 287-294, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31030941

RESUMO

Hematopoietic stem cells (HSCs) are quiescent cells in the bone marrow niche and are relatively dependent on glycolytic ATP production. On the other hand, differentiated cells, including hematopoietic progenitor cells (HPCs), preferentially generate ATP via oxidative phosphorylation. However, it is unclear how cellular differentiation and the cell cycle status affect nutritional requirements and ATP production in HSCs and HPCs. Using a newly developed culture system, we demonstrated that survival of HPCs was strongly dependent on glucose, whereas quiescent HSCs survived for a certain duration without glucose. Among HPCs, granulocyte/monocyte progenitors (GMPs) were particularly dependent on glucose during proliferation. By monitoring the ATP concentration in live cells, we demonstrated that the ATP level was maintained for a short duration without glucose in HSCs, possibly due to their metabolic flexibility. In addition, HSCs exhibited low ATP turnover, whereas HPCs including GMPs demonstrated high ATP turnover and required efficient ATP production from glucose. These findings show that ATP turnover and nutritional requirements differ between HSCs and HPCs according to the cell cycle and differentiation status.


Assuntos
Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacologia , Feminino , Transferência Ressonante de Energia de Fluorescência , Glicólise/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Oxigênio/metabolismo
11.
Blood ; 129(15): 2148-2160, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28209720

RESUMO

Chronic myelomonocytic leukemia (CMML) is a hematological malignancy characterized by uncontrolled proliferation of dysplastic myelomonocytes and frequent progression to acute myeloid leukemia (AML). We identified mutations in the Cbl gene, which encodes a negative regulator of cytokine signaling, in a subset of CMML patients. To investigate the contribution of mutant Cbl in CMML pathogenesis, we generated conditional knockin mice for Cbl that express wild-type Cbl in a steady state and inducibly express CblQ367P , a CMML-associated Cbl mutant. CblQ367P mice exhibited sustained proliferation of myelomonocytes, multilineage dysplasia, and splenomegaly, which are the hallmarks of CMML. The phosphatidylinositol 3-kinase (PI3K)-AKT and JAK-STAT pathways were constitutively activated in CblQ367P hematopoietic stem cells, which promoted cell cycle progression and enhanced chemokine-chemokine receptor activity. Gem, a gene encoding a GTPase that is upregulated by CblQ367P , enhanced hematopoietic stem cell activity and induced myeloid cell proliferation. In addition, Evi1, a gene encoding a transcription factor, was found to cooperate with CblQ367P and progress CMML to AML. Furthermore, targeted inhibition for the PI3K-AKT and JAK-STAT pathways efficiently suppressed the proliferative activity of CblQ367P -bearing CMML cells. Our findings provide insights into the molecular mechanisms underlying mutant Cbl-induced CMML and propose a possible molecular targeting therapy for mutant Cbl-carrying CMML patients.


Assuntos
Ciclo Celular , Células-Tronco Hematopoéticas , Leucemia Mielogênica Crônica BCR-ABL Positiva , Mutação de Sentido Incorreto , Mielopoese , Proteínas Proto-Oncogênicas c-cbl , Regulação para Cima , Substituição de Aminoácidos , Animais , Regulação Enzimológica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/patologia , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Proteínas Monoméricas de Ligação ao GTP/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-cbl/biossíntese , Proteínas Proto-Oncogênicas c-cbl/genética , Transdução de Sinais
12.
J Biol Chem ; 292(29): 12065-12076, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28578315

RESUMO

Docosahexaenoic acid (DHA) is one of the essential ω-3 polyunsaturated fatty acids with a wide range of physiological roles important for human health. For example, DHA renders cell membranes more flexible and is therefore important for cellular function, but information on the mechanisms that control DHA levels in membranes is limited. Specifically, it is unclear which factors determine DHA incorporation into cell membranes and how DHA exerts biological effects. We found that lysophosphatidic acid acyltransferase 3 (LPAAT3) is required for producing DHA-containing phospholipids in various tissues, such as the testes and retina. In this study, we report that LPAAT3-KO mice display severe male infertility with abnormal sperm morphology. During germ cell differentiation, the expression of LPAAT3 was induced, and germ cells obtained more DHA-containing phospholipids. Loss of LPAAT3 caused drastic reduction of DHA-containing phospholipids in spermatids that led to excess cytoplasm around its head, which is normally removed by surrounding Sertoli cells via endocytosis at the final stage of spermatogenesis. In vitro liposome filtration assay raised the possibility that DHA in phospholipids promotes membrane deformation that is required for the rapid endocytosis. These data suggest that decreased membrane flexibility in LPAAT3-KO sperm impaired the efficient removal of sperm content through endocytosis. We conclude that LPAAT3-mediated enrichment of cell membranes with DHA-containing phospholipids endows these membranes with physicochemical properties needed for normal cellular processes, as exemplified by spermatogenesis.


Assuntos
Aciltransferases/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Infertilidade Masculina/enzimologia , Espermatogênese , Espermatozoides/metabolismo , Testículo/metabolismo , Aciltransferases/genética , Animais , Ácidos Docosa-Hexaenoicos/análise , Ácidos Docosa-Hexaenoicos/química , Endocitose , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Lipossomos , Masculino , Fluidez de Membrana , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Cabeça do Espermatozoide/metabolismo , Cabeça do Espermatozoide/patologia , Cabeça do Espermatozoide/ultraestrutura , Espermátides/metabolismo , Espermátides/patologia , Espermátides/ultraestrutura , Espermatozoides/patologia , Espermatozoides/ultraestrutura , Testículo/patologia , Testículo/ultraestrutura
13.
Blood ; 128(5): 638-49, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27301860

RESUMO

Setdb1, also known as Eset, is a methyltransferase that catalyzes trimethylation of H3K9 (H3K9me3) and plays an essential role in the silencing of endogenous retroviral elements (ERVs) in the developing embryo and embryonic stem cells (ESCs). Its role in somatic stem cells, however, remains unclear because of the early death of Setdb1-deficient embryos. We demonstrate here that Setdb1 is the first H3K9 methyltransferase shown to be essential for the maintenance of hematopoietic stem and progenitor cells (HSPCs) in mice. The deletion of Setdb1 caused the rapid depletion of hematopoietic stem and progenitor cells (HSPCs), as well as leukemic stem cells. In contrast to ESCs, ERVs were largely repressed in Setdb1-deficient HSPCs. A list of nonhematopoietic genes was instead ectopically activated in HSPCs after reductions in H3K9me3 levels, including key gluconeogenic enzyme genes fructose-1,6-bisphosphatase 1 (Fbp1) and Fbp2 The ectopic activation of gluconeogenic enzymes antagonized glycolysis and impaired ATP production, resulting in a compromised repopulating capacity of HSPCs. Our results demonstrate that Setdb1 maintains HSPCs by restricting the ectopic activation of nonhematopoietic genes detrimental to their function and uncover that the gluconeogenic pathway is one of the critical targets of Setdb1 in HSPCs.


Assuntos
Regulação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Medula Óssea/patologia , Retrovirus Endógenos/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Deleção de Genes , Inativação Gênica , Gluconeogênese/genética , Homeostase/genética , Leucemia/genética , Leucemia/patologia , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia
14.
J Clin Biochem Nutr ; 63(1): 70-79, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30087547

RESUMO

Carbon monoxide-generating heme oxygenase-2 is expressed in neurons and plays a crucial role for regulating hypoxic vasodilation through mechanisms unlocking carbon monoxide-dependent inhibition of H2S-generating cystathionine ß-synthase expressed in astrocytes. This study aims to examine whether heme oxygenase-2 plays a protective role in mice against stroke. Focal ischemia was induced by middle cerebral artery occlusion. Regional differences in metabolites among ipsilateral and contralateral hemispheres were analysed by quantitative imaging mass spectrometry equipped with an image-processing platform to optimize comparison of local metabolite contents among different animals. Under normoxia, blood flow velocity in precapillary arterioles were significantly elevated in heme oxygenase-2-null mice vs controls, while metabolic intermediates of central carbon metabolism and glutamate synthesis were elevated in the brain of heme oxygenase-2-null mice, suggesting greater metabolic demands to induce hyperemia in these mice. In response to focal ischemia, heme oxygenase-2-null mice exhibited greater regions of ischemic core that coincide with notable decreases in energy metabolism in the contralateral hemisphere as well as in penumbra. In conclusion, these findings suggest that heme oxygenase-2 is involved in mechanisms by which not only protects against compromised energy metabolism of the ipsilateral hemisphere but also ameliorates transhemispheric diaschisis of the contralateral hemisphere in ischemic brain.

15.
J Biol Chem ; 291(14): 7373-85, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26839315

RESUMO

Choroidal neovascularization (CNV) is a pathogenic process of age-related macular degeneration, a vision-threatening disease. The retinal pigment epithelium and macrophages both influence CNV development. However, the underlying mechanisms remain obscure. Here, we focus on Angptl2 (angiopoietin-like protein 2), a cytokine involved in age-related systemic diseases. Angptl2 was originally identified as an adipocytokine and is also expressed in the eye. Using a laser-induced CNV model, we found thatAngptl2KO mice exhibited suppressed CNV development with reduced macrophage recruitment and inflammatory mediator induction. The mediators monocyte chemotactic protein-1, interleukin-1ß (Il-1ß),Il-6, matrix metalloprotease-9 (Mmp-9), and transforming growth factor-ß1 (Tgf-ß1) that were up-regulated during CNV development were all suppressed in the retinal pigment epithelium-choroid of CNV models generated in theAngptl2KO mice. Bone marrow transplantation using wild-type and KO mice suggested that both bone marrow-derived and host-derived Angptl2 were responsible for macrophage recruitment and CNV development. Peritoneal macrophages derived fromAngptl2KO mice expressed lower levels of the inflammatory mediators. In the wild-type peritoneal macrophages and RAW264.7 cells, Angptl2 induced the mediators via integrins α4 and ß2, followed by the downstream activation of NF-κB and ERK. The activation of NF-κB and ERK by Angptl2 also promoted macrophage migration. Therefore, Angptl2 from focal tissue might trigger macrophage recruitment, and that from recruited macrophages might promote expression of inflammatory mediators including Angptl2 in an autocrine and/or paracrine fashion to facilitate CNV development. Angptl2 might therefore represent a multistep regulator of CNV pathogenesis and serve as a new therapeutic target for age-related macular degeneration.


Assuntos
Angiopoietinas/biossíntese , Neovascularização de Coroide/metabolismo , Macrófagos/metabolismo , Degeneração Macular/metabolismo , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Animais , Antígenos CD18/genética , Antígenos CD18/metabolismo , Linhagem Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Neovascularização de Coroide/genética , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Integrina alfa4/genética , Integrina alfa4/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/patologia , Degeneração Macular/genética , Degeneração Macular/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
16.
Blood ; 125(22): 3437-46, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25872778

RESUMO

We previously reported that deficiency for Samd9L, which was cloned as a candidate gene for -7/7q- syndrome, accelerated leukemia cooperatively with enhanced expression of a histone demethylase: F-box and leucine-rich repeat protein 10 (Fbxl10, also known as Jhdm1b, Kdm2b, and Ndy1). To further investigate the role of Fbxl10 in leukemogenesis, we generated transgenic (Tg) mice that overexpress Fbxl10 in hematopoietic stem cells (HSCs). Interestingly, Fbxl10 Tg mice developed myeloid or B-lymphoid leukemia with complete penetrance. HSCs from the Tg mice exhibited an accelerated G0/G1-to-S transition with a normal G0 to G1 entry, resulting in pleiotropic progenitor cell expansion. Fbxl10 Tg HSCs displayed enhanced expression of neuron-specific gene family member 2 (Nsg2), and forced expression of Nsg2 in primary bone marrow cells resulted in expansion of immature cells. In addition, the genes involved in mitochondrial oxidative phosphorylation were markedly enriched in Fbxl10 Tg HSCs, coupled with increased cellular adenosine 5'-triphosphate levels. Moreover, chromatin immunoprecipitation followed by sequencing analysis demonstrated that Fbxl10 directly binds to the regulatory regions of Nsg2 and oxidative phosphorylation genes. These findings define Fbxl10 as a bona fide oncogene, whose deregulated expression contributes to the development of leukemia involving metabolic proliferative advantage and Nsg2-mediated impaired differentiation.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas F-Box/genética , Células-Tronco Hematopoéticas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Leucemia/genética , Leucemia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Linfócitos B/patologia , Proteínas de Transporte/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Proteínas F-Box/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/patologia , Proteínas do Tecido Nervoso/genética , Oncogenes , Regulação para Cima/genética
17.
Stem Cells ; 34(4): 1068-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27095138

RESUMO

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.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Diferenciação Celular/genética , Estrona/genética , Células-Tronco Hematopoéticas/patologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Células da Medula Óssea/patologia , Linhagem da Célula/genética , Proliferação de Células/genética , Desenvolvimento Embrionário/genética , Células-Tronco Hematopoéticas/metabolismo , Homeostase/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout
18.
Rinsho Ketsueki ; 58(10): 1844-1850, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28978823

RESUMO

During steady-state conditions, hematopoietic stem cells (HSCs) maintain a quiescent status in the cell cycle. Upon infection or inflammation, bone marrow HSCs begin proliferating and generating differentiated hematopoietic cells via multi-lineage differentiation and self-renewal; this effect is partially due to the alteration of their surrounding microenvironment or niche. In addition, recent studies have revealed that the bone marrow niche critically contributes to abnormal hematopoiesis, including leukemogenesis. In this review, we discuss the recent advances in our understanding of HSC/niche functions and the regulatory machineries employed during homeostasis, stress hematopoiesis, or disease conditions.


Assuntos
Hematopoese , Nicho de Células-Tronco , Animais , Células-Tronco Hematopoéticas , Humanos , Leucemia , Neovascularização Patológica
19.
Clin Calcium ; 27(6): 823-828, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28536320

RESUMO

At steady state, hematopoietic stem cells(HSCs), the most undifferentiated cells in the hematological system, are kept quiescent in the cell cycle. Upon hematological stresses, including radiation, anti-cancer medication, infection, and transplantation, bone marrow HSCs enter the cell cycle and robustly repopulate the entire hematopoietic system via multi-lineage differentiation and self-renewal, partly due to the alteration of their surrounding microenvironment or niche. Such hematological repopulation activity is termed "stress hematopoiesis," an activity essential for homeostatic maintenance of blood production. Understanding stress hematopoiesis is also critical to establish efficient methods enabling expansion in vitro of HSCs, since forced activation of the cell cycle by cytokine supplementation induces a stress-like cellular state.


Assuntos
Células-Tronco Hematopoéticas/citologia , Estresse Fisiológico , Animais , Proliferação de Células , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Humanos , Nicho de Células-Tronco
20.
Pflugers Arch ; 468(1): 13-22, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26490456

RESUMO

Bone marrow, the site of hematopoiesis throughout adulthood, is a physiologically hypoxic organ. Thus, various biological oxygen sensors and their signaling cascades play a pivotal role in hematopoietic systems in the bone marrow under both physiologic and pathologic conditions. Hypoxia-inducible factors (HIFs) are hypoxic stress sensor proteins that are stabilized under homeostatic or stress-induced hypoxia. In the hypoxic bone marrow, HIFs play crucial roles in hematopoietic stem cells (HSCs) and in the cells of the HSC niche. The signals downstream of the HIFs maintain HSC quiescence, survival, and metabolic homeostasis through both cell-autonomous and non-cell-autonomous mechanisms. Leukemic stem cells (LSCs) hijack these delicate hypoxia-sensing mechanisms to sustain their self-renewal potential, promoting disease progression and drug resistance even under normoxic conditions. This review focuses on HIF-mediated oxygen-sensing mechanisms of adult HSCs and LSCs and their niche cells in the hypoxic bone marrow.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA