Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 159(5): 1070-1085, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25416946

RESUMO

Hematopoietic stem cells (HSCs) underlie the production of blood and immune cells for the lifetime of an organism. In vertebrate embryos, HSCs arise from the unique transdifferentiation of hemogenic endothelium comprising the floor of the dorsal aorta during a brief developmental window. To date, this process has not been replicated in vitro from pluripotent precursors, partly because the full complement of required signaling inputs remains to be determined. Here, we show that TNFR2 via TNF? activates the Notch and NF-?B signaling pathways to establish HSC fate, indicating a requirement for inflammatory signaling in HSC generation. We determine that primitive neutrophils are the major source of TNF?, assigning a role for transient innate immune cells in establishing the HSC program. These results demonstrate that proinflammatory signaling, in the absence of infection, is utilized by the developing embryo to generate the lineal precursors of the adult hematopoietic system.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , Animais , Embrião não Mamífero/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , NF-kappa B/metabolismo , Neutrófilos/metabolismo , Receptores Notch/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra/metabolismo
2.
Development ; 142(6): 1050-61, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25758220

RESUMO

The adult blood system is established by hematopoietic stem cells (HSCs), which arise during development from an endothelial-to-hematopoietic transition of cells comprising the floor of the dorsal aorta. Expression of aortic runx1 has served as an early marker of HSC commitment in the zebrafish embryo, but recent studies have suggested that HSC specification begins during the convergence of posterior lateral plate mesoderm (PLM), well before aorta formation and runx1 transcription. Further understanding of the earliest stages of HSC specification necessitates an earlier marker of hemogenic endothelium. Studies in mice have suggested that GATA2 might function at early stages within hemogenic endothelium. Two orthologs of Gata2 exist in zebrafish: gata2a and gata2b. Here, we report that gata2b expression initiates during the convergence of PLM, becoming restricted to emerging HSCs. We observe Notch-dependent gata2b expression within the hemogenic subcompartment of the dorsal aorta that is in turn required to initiate runx1 expression. Our results indicate that Gata2b functions within hemogenic endothelium from an early stage, whereas Gata2a functions more broadly throughout the vascular system.


Assuntos
Padronização Corporal/fisiologia , Fator de Transcrição GATA2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hemangioblastos/fisiologia , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Animais , Aorta/citologia , Aorta/embriologia , Proteínas de Bactérias , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hibridização In Situ , Proteínas Luminescentes , Mesoderma/embriologia , Oligonucleotídeos Antissenso/genética , Reação em Cadeia da Polimerase em Tempo Real , Imagem com Lapso de Tempo , Proteínas de Peixe-Zebra/metabolismo , Proteína Vermelha Fluorescente
3.
PLoS Pathog ; 12(6): e1005699, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27351838

RESUMO

TNFα is a pleiotropic pro-inflammatory cytokine with a key role in the activation of the immune system to fight viral infections. Despite its antiviral role, a few viruses might utilize the host produced TNFα to their benefit. Some recent reports have shown that anti-TNFα therapies could be utilized to treat certain viral infections. However, the underlying mechanisms by which TNFα can favor virus replication have not been identified. Here, a rhabdoviral infection model in zebrafish allowed us to identify the mechanism of action by which Tnfa has a deleterious role for the host to combat certain viral infections. Our results demonstrate that Tnfa signals through its receptor Tnfr2 to enhance viral replication. Mechanistically, Tnfa does not affect viral adhesion and delivery from endosomes to the cytosol. In addition, the host interferon response was also unaffected by Tnfa levels. However, Tnfa blocks the host autophagic response, which is required for viral clearance. This mechanism of action provides new therapeutic targets for the treatment of SVCV-infected fish, and advances our understanding of the previously enigmatic deleterious role of TNFα in certain viral infections.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Infecções por Rhabdoviridae/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Autofagia/fisiologia , Western Blotting , Modelos Animais de Doenças , Imunofluorescência , Organismos Geneticamente Modificados , Reação em Cadeia da Polimerase , Rhabdoviridae/imunologia , Replicação Viral/fisiologia , Peixe-Zebra
4.
PLoS Biol ; 12(5): e1001855, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24802997

RESUMO

TNFα overexpression has been associated with several chronic inflammatory diseases, including psoriasis, lichen planus, rheumatoid arthritis, and inflammatory bowel disease. Paradoxically, numerous studies have reported new-onset psoriasis and lichen planus following TNFα antagonist therapy. Here, we show that genetic inhibition of Tnfa and Tnfr2 in zebrafish results in the mobilization of neutrophils to the skin. Using combinations of fluorescent reporter transgenes, fluorescence microscopy, and flow cytometry, we identified the local production of dual oxidase 1 (Duox1)-derived H2O2 by Tnfa- and Tnfr2-deficient keratinocytes as a trigger for the activation of the master inflammation transcription factor NF-κB, which then promotes the induction of genes encoding pro-inflammatory molecules. In addition, pharmacological inhibition of Duox1 completely abrogated skin inflammation, placing Duox1-derived H2O2 upstream of this positive feedback inflammatory loop. Strikingly, DUOX1 was drastically induced in the skin lesions of psoriasis and lichen planus patients. These results reveal a crucial role for TNFα/TNFR2 axis in the protection of the skin against DUOX1-mediated oxidative stress and could establish new therapeutic targets for skin inflammatory disorders.


Assuntos
Líquen Plano/metabolismo , NADPH Oxidases/metabolismo , Psoríase/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Pele/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Embrião não Mamífero , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Queratinócitos/metabolismo , Queratinócitos/patologia , Líquen Plano/genética , Líquen Plano/patologia , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Infiltração de Neutrófilos , Estresse Oxidativo , Psoríase/genética , Psoríase/patologia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Transdução de Sinais , Pele/patologia , Fator de Necrose Tumoral alfa/genética , Peixe-Zebra
5.
Blood ; 122(24): 3918-28, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24128862

RESUMO

Granulocyte colony-stimulating factor (Gcsf) drives the proliferation and differentiation of granulocytes, monocytes, and macrophages (mφs) from hematopoietic stem and progenitor cells (HSPCs). Analysis of the zebrafish genome indicates the presence of 2 Gcsf ligands, likely resulting from a duplication event in teleost evolution. Although Gcsfa and Gcsfb share low sequence conservation, they share significant similarity in their predicted ligand/receptor interaction sites and structure. Each ligand displays differential temporal expression patterns during embryogenesis and spatial expression patterns in adult animals. To determine the functions of each ligand, we performed loss- and gain-of-function experiments. Both ligands signal through the Gcsf receptor to expand primitive neutrophils and mφs, as well as definitive granulocytes. To further address their functions, we generated recombinant versions and tested them in clonal progenitor assays. These sensitive in vitro techniques indicated similar functional attributes in supporting HSPC growth and differentiation. Finally, in addition to supporting myeloid differentiation, zebrafish Gcsf is required for the specification and proliferation of hematopoietic stem cells, suggesting that Gcsf represents an ancestral cytokine responsible for the broad support of HSPCs. These findings may inform how hematopoietic cytokines evolved following the diversification of teleosts and mammals from a common ancestor.


Assuntos
Fator Estimulador de Colônias de Granulócitos/genética , Hematopoese/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Fator Estimulador de Colônias de Granulócitos/metabolismo , Sistema Hematopoético/embriologia , Sistema Hematopoético/metabolismo , Hibridização In Situ , Ligantes , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Microscopia Confocal , Mielopoese/genética , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo
6.
Nat Commun ; 15(1): 7787, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39242546

RESUMO

Most gene functions have been discovered through phenotypic observations under loss of function experiments that lack temporal control. However, cell signaling relies on limited transcriptional effectors, having to be re-used temporally and spatially within the organism. Despite that, the dynamic nature of signaling pathways have been overlooked due to the difficulty on their assessment, resulting in important bottlenecks. Here, we have utilized the rapid and synchronized developmental transitions occurring within the zebrafish embryo, in conjunction with custom NF-kB reporter embryos driving destabilized fluorophores that report signaling dynamics in real time. We reveal that NF-kB signaling works as a clock that controls the developmental progression of hematopoietic stem and progenitor cells (HSPCs) by two p65 activity waves that inhibit cell cycle. Temporal disruption of each wave results in contrasting phenotypic outcomes: loss of HSPCs due to impaired specification versus proliferative expansion and failure to delaminate from their niche. We also show functional conservation during human hematopoietic development using iPSC models. Our work identifies p65 as a previously unrecognized contributor to cell cycle regulation, revealing why and when pro-inflammatory signaling is required during HSPC development. It highlights the importance of considering and leveraging cell signaling as a temporally dynamic entity.


Assuntos
Ciclo Celular , Células-Tronco Hematopoéticas , Transdução de Sinais , Peixe-Zebra , Animais , Humanos , Diferenciação Celular , Proliferação de Células , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Fator de Transcrição RelA/metabolismo , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
7.
Nat Commun ; 14(1): 7668, 2023 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-37996457

RESUMO

Uncovering the mechanisms regulating hematopoietic specification not only would overcome current limitations related to hematopoietic stem and progenitor cell (HSPC) transplantation, but also advance cellular immunotherapies. However, generating functional human induced pluripotent stem cell (hiPSC)-derived HSPCs and their derivatives has been elusive, necessitating a better understanding of the developmental mechanisms that trigger HSPC specification. Here, we reveal that early activation of the Nod1-Ripk2-NF-kB inflammatory pathway in endothelial cells (ECs) primes them to switch fate towards definitive hemogenic endothelium, a pre-requisite to specify HSPCs. Our genetic and chemical embryonic models show that HSPCs fail to specify in the absence of Nod1 and its downstream kinase Ripk2 due to a failure on hemogenic endothelial (HE) programming, and that small Rho GTPases coordinate the activation of this pathway. Manipulation of NOD1 in a human system of definitive hematopoietic differentiation indicates functional conservation. This work establishes the RAC1-NOD1-RIPK2-NF-kB axis as a critical intrinsic inductor that primes ECs prior to HE fate switch and HSPC specification. Manipulation of this pathway could help derive a competent HE amenable to specify functional patient specific HSPCs and their derivatives for the treatment of blood disorders.


Assuntos
Hemangioblastos , Células-Tronco Pluripotentes Induzidas , Proteínas Monoméricas de Ligação ao GTP , Humanos , Diferenciação Celular , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , NF-kappa B/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
8.
Zebrafish ; 19(3): 104-108, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35704898

RESUMO

Cleavage Under Targets and Release Using Nuclease (CUT&RUN) has emerged as a chromatin profiling strategy that excels traditional methods. Although CUT&RUN has been widely utilized in mammalian cells, its use in the zebrafish is at its early stages. In this study, we have developed a protocol to successfully perform CUT&RUN to map transcription factor (TF) binding sites in embryonic, adult tissues, and FACS-sorted zebrafish cells. We also provide a detailed workflow for the identification of predicted TF binding sites that can be utilized in any animal species. Altogether, our strategy will expand this invaluable tool to the zebrafish community, improving the epigenetic resolution that can be achieved in this model organism.


Assuntos
Cromatina , Peixe-Zebra , Animais , Sítios de Ligação , Endonucleases/metabolismo , Mamíferos/metabolismo , Ligação Proteica , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo
9.
Blood Adv ; 5(3): 796-811, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33560393

RESUMO

Granulin is a pleiotropic protein involved in inflammation, wound healing, neurodegenerative disease, and tumorigenesis. These roles in human health have prompted research efforts to use granulin to treat rheumatoid arthritis and frontotemporal dementia and to enhance wound healing. But how granulin contributes to each of these diverse biological functions remains largely unknown. Here, we have uncovered a new role for granulin during myeloid cell differentiation. We have taken advantage of the tissue-specific segregation of the zebrafish granulin paralogues to assess the functional role of granulin in hematopoiesis without perturbing other tissues. By using our zebrafish model of granulin deficiency, we revealed that during normal and emergency myelopoiesis, myeloid progenitors are unable to terminally differentiate into neutrophils and macrophages in the absence of granulin a (grna), failing to express the myeloid-specific genes cebpa, rgs2, lyz, mpx, mpeg1, mfap4, and apoeb. Functionally, macrophages fail to recruit to the wound, resulting in abnormal healing. Our CUT&RUN experiments identify Pu.1, which together with Irf8, positively regulates grna expression. In vivo imaging and RNA sequencing experiments show that grna inhibits the expression of gata1, leading to the repression of the erythroid program. Importantly, we demonstrated functional conservation between the mammalian granulin and the zebrafish ortholog grna. Our findings uncover a previously unrecognized role for granulin during myeloid cell differentiation, which opens a new field of study that can potentially have an impact on different aspects of human health and expand the therapeutic options for treating myeloid disorders such as neutropenia or myeloid leukemia.


Assuntos
Doenças Neurodegenerativas , Peixe-Zebra , Animais , Proteínas de Transporte , Proteínas da Matriz Extracelular , Glicoproteínas , Granulinas , Hematopoese , Humanos , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Trends Cell Biol ; 28(1): 58-66, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28882414

RESUMO

Hematopoietic stem cells (HSCs) have the extraordinary ability to both self-renew and generate all mature blood cell lineages. The ability to produce or expand patient-derived HSCs in vitro would greatly improve the outcome for patients with blood disorders that are currently treated with allogeneic HSC transplantation. Many laboratories have been working to identify the signals required for HSC emergence in their native environments to apply this knowledge in vitro. Recently, several signals traditionally known to underlie classical inflammation have emerged as essential regulators of HSC development. In this review we synthesize the findings that have established inflammatory cues as key regulators of HSC development.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Inflamação/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula/fisiologia , Citocinas/metabolismo , Hematopoese Extramedular/fisiologia , Humanos , Transdução de Sinais
11.
Exp Hematol ; 44(7): 519-27, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27132652

RESUMO

The nuclear factor-κB (NF-κB) family is a crucial transcription factor group known mainly for its role in the regulation of the immune system and its response to infection in vertebrates. The signaling pathway leading to NF-κB activation and translocation to the nucleus to exert its function as a transcription factor is well conserved among Kingdom Animalia, which has helped to elucidate other roles that NF-κB plays in other biological contexts such as developmental biology. The manipulation of NF-κB members in a diverse range of animal models results in severe developmental defects during embryogenesis, very often leading to embryonic lethality. Defects include dorsal-ventral patterning and limb, liver, skin, lung, neural, notochord, muscle, skeletal, and hematopoietic defects. Here, we recapitulate the research that has been done to address the role that NF-κB plays during embryonic development, in particular to emphasize its recently discovered role in the specification of hematopoietic stem cells (HSCs), the foundation of the hematopoietic system in vertebrates.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/metabolismo , NF-kappa B/metabolismo , Animais , Padronização Corporal/genética , Desenvolvimento Embrionário/genética , Hematopoese , Humanos , NF-kappa B/genética , Organogênese/genética
12.
Dev Comp Immunol ; 50(2): 155-65, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25681741

RESUMO

TLR4 was the first TLR family member identified in mammals and is responsible for the activation of the immune response by bacterial LPS. Later, MD1 and RP105 were shown to form complexes that directly interact with the MD2-TLR4 complex, acting as physiological negative regulators of LPS signaling. Despite the general conservation of various TLR families from fish to mammals, several differences can be appreciated, such as the high tolerance of fish to LPS, the absence of the crucial accessory molecules Md2 and Cd14 for Tlr4 signaling in fish, the absence of Tlr4 in some fish species, and the confirmation that LPS does not signal through Tlr4 in zebrafish. The present study has identified the Rp105 and Md1 homologs in zebrafish, confirming (i) Rp105 and Tlr4 evolved from a common ancestor before the divergence between fish and tetrapods and (ii) the presence of Md1 in teleost fish and the lack of Md2, suggesting that the divergence of these accessory molecules occurred in the tetrapod lineage. Biochemical and functional studies indicate that Md1 binds both Rp105 and Tlr4 in zebrafish. Genetic inhibition of zebrafish Md1 and Rp105 reveals that Md1 or Rp105 deficiency impairs the expression of genes encoding pro-inflammatory and antiviral molecules, leading to increased susceptibility to viral infection. These results shed light on the evolutionary history of Md1 and Rp105 and uncover a previously unappreciated function of these molecules in the regulation of innate immunity.


Assuntos
Antígenos de Superfície/imunologia , Doenças dos Peixes/imunologia , Receptor 4 Toll-Like/imunologia , Proteínas de Peixe-Zebra/imunologia , Peixe-Zebra/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Superfície/genética , Doenças dos Peixes/virologia , Imunidade Inata/imunologia , Inflamação/genética , Inflamação/imunologia , Antígeno 96 de Linfócito/imunologia , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Transdução de Sinais/imunologia , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA