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.
Proc Natl Acad Sci U S A ; 120(18): e2217862120, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37094122

RESUMO

Hematopoietic stem and progenitor cells maintain blood cell homeostasis by integrating various cues provided by specialized microenvironments or niches. Biomechanical forces are emerging as key regulators of hematopoiesis. Here, we report that mechanical stimuli provided by blood flow in the vascular niche control Drosophila hematopoiesis. In vascular niche cells, the mechanosensitive channel Piezo transduces mechanical forces through intracellular calcium upregulation, leading to Notch activation and repression of FGF ligand transcription, known to regulate hematopoietic progenitor maintenance. Our results provide insight into how the vascular niche integrates mechanical stimuli to regulate hematopoiesis.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Hematopoese/fisiologia , Células Sanguíneas , Células-Tronco/metabolismo , Nicho de Células-Tronco , Canais Iônicos
2.
Blood ; 136(7): 831-844, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32457985

RESUMO

The defined location of a stem cell within a niche regulates its fate, behavior, and molecular identity via a complex extrinsic regulation that is far from being fully elucidated. To explore the molecular characteristics and key components of the aortic microenvironment, where the first hematopoietic stem cells are generated during development, we performed genome-wide RNA tomography sequencing on zebrafish, chicken, mouse, and human embryos. The resulting anterior-posterior and dorsal-ventral transcriptional maps provided a powerful resource for exploring genes and regulatory pathways active in the aortic microenvironment. By performing interspecies comparative RNA sequencing analyses and functional assays, we explored the complexity of the aortic microenvironment landscape and the fine-tuning of various factors interacting to control hematopoietic stem cell generation, both in time and space in vivo, including the ligand-receptor couple ADM-RAMP2 and SVEP1. Understanding the regulatory function of the local environment will pave the way for improved stem cell production in vitro and clinical cell therapy.


Assuntos
Aorta/embriologia , Células-Tronco Hematopoéticas/citologia , RNA/análise , Nicho de Células-Tronco/genética , Tomografia , Animais , Animais Geneticamente Modificados , Aorta/citologia , Rastreamento de Células/métodos , Embrião de Galinha , Embrião de Mamíferos , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , RNA/genética , Análise de Sequência de RNA/métodos , Análise de Célula Única , Especificidade da Espécie , Tomografia/métodos , Tomografia/veterinária , Peixe-Zebra/embriologia , Peixe-Zebra/genética
3.
Proc Natl Acad Sci U S A ; 109(9): 3389-94, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22331866

RESUMO

The Drosophila melanogaster larval hematopoietic organ, the lymph gland, is a model to study in vivo the function of the hematopoietic niche. A small cluster of cells in the lymph gland, the posterior signaling center (PSC), maintains the balance between hematopoietic progenitors (prohemocytes) and their differentiation into specialized blood cells (hemocytes). Here, we show that Decapentaplegic/bone morphogenetic protein (Dpp/BMP) signaling activity in PSC cells controls niche size. In the absence of BMP signaling, the number of PSC cells increases. Correlatively, no hemocytes differentiate. Controlling PSC size is, thus, essential for normal blood cell homeostasis. Activation of BMP signaling in the PSC requires expression of the Dally-like heparan-sulfate proteoglycan, under the control of the Collier/early B-cell factor (EBF) transcription factor. A Dpp > dpp autoregulatory loop maintains BMP signaling, which limits PSC cell proliferation by repressing the protooncogene dmyc. Dpp antagonizes activity of wingless (Wg)/Wnt signaling, which positively regulates the number of PSC cells via the control of Dmyc expression. Together, our data show that Collier controls hemocyte homeostasis via coordinate regulation of PSC cell number and PSC signaling to prohemocytes. In mouse, EBF2, BMP, and Wnt signaling in osteoblasts is required for the proper number of niche and hematopoietic stem cells. Our findings bring insights to niche size control and draw parallels between Drosophila and mammalian hematopoiesis.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Hematopoese/fisiologia , Hemócitos/citologia , Nicho de Células-Tronco , Fatores de Transcrição/fisiologia , Animais , Contagem de Células , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Genes myc , Hemócitos/metabolismo , Larva , Camundongos , Índice Mitótico , Proteoglicanas/genética , Proteoglicanas/fisiologia , Transdução de Sinais/fisiologia , Especificidade da Espécie , Fatores de Transcrição/genética , Vertebrados/fisiologia , Proteína Wnt1/genética , Proteína Wnt1/fisiologia
4.
Med Sci (Paris) ; 30(1): 76-81, 2014 Jan.
Artigo em Francês | MEDLINE | ID: mdl-24472463

RESUMO

Stem cells are required for both tissue renewal and repair in response to injury. The maintenance and function of stem cells is controlled by their specific cellular microenvironment called "niche". Hematopoietic stem cells (HSC) that give rise to all blood cell types have been extensively studied in mammals. Genetic and molecular analyses performed in mice identified several signaling pathways involved in the cellular communications between HSC and their niche. However, hematopoietic niche plasticity remains poorly understood. The discovery of a Drosophila hematopoietic niche, called PSC, established a new model to decipher the niche function in vivo. Size control of the PSC is essential to maintain hematopoietic tissue homeostasis and a molecular cascade controlling the PSC cell number has been characterized. Novel parallels between Drosophila and mammalian hematopoietic niches open new perspectives for studies of HSC biology in human.


Assuntos
Drosophila , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Drosophila/citologia , Drosophila/fisiologia , Hematopoese/fisiologia , Humanos , Mamíferos , Camundongos , Modelos Biológicos
5.
Front Cell Dev Biol ; 10: 834720, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237606

RESUMO

The Drosophila lymph gland is the larval hematopoietic organ and is aligned along the anterior part of the cardiovascular system, composed of cardiac cells, that form the cardiac tube and its associated pericardial cells or nephrocytes. By the end of embryogenesis the lymph gland is composed of a single pair of lobes. Two additional pairs of posterior lobes develop during larval development to contribute to the mature lymph gland. In this study we describe the ontogeny of lymph gland posterior lobes during larval development and identify the genetic basis of the process. By lineage tracing we show here that each posterior lobe originates from three embryonic pericardial cells, thus establishing a bivalent blood cell/nephrocyte potential for a subset of embryonic pericardial cells. The posterior lobes of L3 larvae posterior lobes are composed of heterogeneous blood progenitors and their diversity is progressively built during larval development. We further establish that in larvae, homeotic genes and the transcription factor Klf15 regulate the choice between blood cell and nephrocyte fates. Our data underline the sequential production of blood cell progenitors during larval development.

6.
Front Immunol ; 12: 719349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484226

RESUMO

In adult mammals, blood cells are formed from hematopoietic stem progenitor cells, which are controlled by a complex cellular microenvironment called "niche". Drosophila melanogaster is a powerful model organism to decipher the mechanisms controlling hematopoiesis, due both to its limited number of blood cell lineages and to the conservation of genes and signaling pathways throughout bilaterian evolution. Insect blood cells or hemocytes are similar to the mammalian myeloid lineage that ensures innate immunity functions. Like in vertebrates, two waves of hematopoiesis occur in Drosophila. The first wave takes place during embryogenesis. The second wave occurs at larval stages, where two distinct hematopoietic sites are identified: subcuticular hematopoietic pockets and a specialized hematopoietic organ called the lymph gland. In both sites, hematopoiesis is regulated by distinct niches. In hematopoietic pockets, sensory neurons of the peripheral nervous system provide a microenvironment that promotes embryonic hemocyte expansion and differentiation. In the lymph gland blood cells are produced from hematopoietic progenitors. A small cluster of cells called Posterior Signaling Centre (PSC) and the vascular system, along which the lymph gland develops, act collectively as a niche, under homeostatic conditions, to control the balance between maintenance and differentiation of lymph gland progenitors. In response to an immune stress such as wasp parasitism, lymph gland hematopoiesis is drastically modified and shifts towards emergency hematopoiesis, leading to increased progenitor proliferation and their differentiation into lamellocyte, a specific blood cell type which will neutralize the parasite. The PSC is essential to control this emergency response. In this review, we summarize Drosophila cellular and molecular mechanisms involved in the communication between the niche and hematopoietic progenitors, both under homeostatic and stress conditions. Finally, we discuss similarities between mechanisms by which niches regulate hematopoietic stem/progenitor cells in Drosophila and mammals.


Assuntos
Comunicação Celular , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Nicho de Células-Tronco , Estresse Fisiológico , Animais , Microambiente Celular , Drosophila , Hemócitos/citologia , Hemócitos/metabolismo , Larva , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Nicho de Células-Tronco/imunologia , Estresse Fisiológico/imunologia
7.
Elife ; 102021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33395389

RESUMO

In adult mammals, hematopoiesis, the production of blood cells from hematopoietic stem and progenitor cells (HSPCs), is tightly regulated by extrinsic signals from the microenvironment called 'niche'. Bone marrow HSPCs are heterogeneous and controlled by both endosteal and vascular niches. The Drosophila hematopoietic lymph gland is located along the cardiac tube which corresponds to the vascular system. In the lymph gland, the niche called Posterior Signaling Center controls only a subset of the heterogeneous hematopoietic progenitor population indicating that additional signals are necessary. Here we report that the vascular system acts as a second niche to control lymph gland homeostasis. The FGF ligand Branchless produced by vascular cells activates the FGF pathway in hematopoietic progenitors. By regulating intracellular calcium levels, FGF signaling maintains progenitor pools and prevents blood cell differentiation. This study reveals that two niches contribute to the control ofDrosophila blood cell homeostasis through their differential regulation of progenitors.


Assuntos
Drosophila/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Hematopoese/fisiologia , Transdução de Sinais , Animais
8.
Elife ; 92020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33357377

RESUMO

Organisms rely on inducible and constitutive immune defences to combat infection. Constitutive immunity enables a rapid response to infection but may carry a cost for uninfected individuals, leading to the prediction that it will be favoured when infection rates are high. When we exposed populations of Drosophila melanogaster to intense parasitism by the parasitoid wasp Leptopilina boulardi, they evolved resistance by developing a more reactive cellular immune response. Using single-cell RNA sequencing, we found that immune-inducible genes had become constitutively upregulated. This was the result of resistant larvae differentiating precursors of specialized immune cells called lamellocytes that were previously only produced after infection. Therefore, populations evolved resistance by genetically hard-wiring the first steps of an induced immune response to become constitutive.


Assuntos
Evolução Biológica , Resistência à Doença/imunologia , Drosophila melanogaster/imunologia , Imunidade Celular/imunologia , Infecções/imunologia , Animais , Resistência à Doença/genética , Drosophila melanogaster/parasitologia , Feminino , Regulação da Expressão Gênica , Hemócitos/imunologia , Larva/imunologia , Masculino , Vespas
9.
Elife ; 62017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29091025

RESUMO

Hematopoietic stem/progenitor cells in the adult mammalian bone marrow ensure blood cell renewal. Their cellular microenvironment, called 'niche', regulates hematopoiesis both under homeostatic and immune stress conditions. In the Drosophila hematopoietic organ, the lymph gland, the posterior signaling center (PSC) acts as a niche to regulate the hematopoietic response to immune stress such as wasp parasitism. This response relies on the differentiation of lamellocytes, a cryptic cell type, dedicated to pathogen encapsulation and killing. Here, we establish that Toll/NF-κB pathway activation in the PSC in response to wasp parasitism non-cell autonomously induces the lymph gland immune response. Our data further establish a regulatory network where co-activation of Toll/NF-κB and EGFR signaling by ROS levels in the PSC/niche controls lymph gland hematopoiesis under parasitism. Whether a similar regulatory network operates in mammals to control emergency hematopoiesis is an open question.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/imunologia , Receptores ErbB/metabolismo , Hematopoese , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Receptores Toll-Like/metabolismo , Vespas/imunologia , Animais , Drosophila/parasitologia , Interações Hospedeiro-Parasita , Imunidade Inata
10.
Nat Commun ; 7: 11634, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27193394

RESUMO

Self-renewal and differentiation of mammalian haematopoietic stem cells (HSCs) are controlled by a specialized microenvironment called 'the niche'. In the bone marrow, HSCs receive signals from both the endosteal and vascular niches. The posterior signalling centre (PSC) of the larval Drosophila haematopoietic organ, the lymph gland, regulates blood cell differentiation under normal conditions and also plays a key role in controlling haematopoiesis under immune challenge. Here we report that the Drosophila vascular system also contributes to the lymph gland homoeostasis. Vascular cells produce Slit that activates Robo receptors in the PSC. Robo activation controls proliferation and clustering of PSC cells by regulating Myc, and small GTPase and DE-cadherin activity, respectively. These findings reveal that signals from the vascular system contribute to regulating the rate of blood cell differentiation via the regulation of PSC morphology.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Hematopoese , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Animais , Caderinas/metabolismo , Sistema Cardiovascular/metabolismo , Drosophila/citologia , Larva/citologia , Larva/fisiologia , Proteínas do Tecido Nervoso/genética , Proteoglicanas/metabolismo , Receptores Imunológicos/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Roundabout
11.
PLoS One ; 11(2): e0148978, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866694

RESUMO

Blood cell production in the Drosophila hematopoietic organ, the lymph gland, is controlled by intrinsic factors and extrinsic signals. Initial analysis of Collier/Early B Cell Factor function in the lymph gland revealed the role of the Posterior Signaling Center (PSC) in mounting a dedicated cellular immune response to wasp parasitism. Further, premature blood cell differentiation when PSC specification or signaling was impaired, led to assigning the PSC a role equivalent to the vertebrate hematopoietic niche. We report here that Collier is expressed in a core population of lymph gland progenitors and cell autonomously maintains this population. The PSC contributes to lymph gland homeostasis by regulating blood cell differentiation, rather than by maintaining core progenitors. In addition to PSC signaling, switching off Collier expression in progenitors is required for efficient immune response to parasitism. Our data show that two independent sites of Collier/Early B Cell Factor expression, hematopoietic progenitors and the PSC, achieve control of hematopoiesis.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Hemócitos/citologia , Linfa/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Hematopoese , Homeostase , Interações Hospedeiro-Parasita , Sistema Imunitário , Hibridização In Situ , Mitose , Interferência de RNA , Transdução de Sinais , Nicho de Células-Tronco , Células-Tronco , Vespas
12.
JAKSTAT ; 2(3): e25700, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24069567

RESUMO

Genetic alterations affecting the JAK-STAT signaling pathway are linked to several malignancies and hematological disorders in humans. Despite being one of the most extensively studied pathways, there remain many gaps to fill. JAK-STAT components are widely conserved during evolution. Here, we review the known roles of the JAK-STAT pathway in Drosophila immunity: controlling the different steps of hematopoiesis, both under physiological conditions and in response to immune challenge, and contributing to antiviral responses. We then summarize what is currently known about JAK-STAT signaling in renewal of the adult intestine, under physiological conditions or in response to ingestion of pathogenic bacteria.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA