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











Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 12(8): 729, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294700

RESUMO

Bone morphogenetic protein (Bmp) signaling is critical for organismal development and homeostasis. To elucidate Bmp2 function in the vascular/hematopoietic lineages we generated a new transgenic mouse line in which ectopic Bmp2 expression is controlled by the Tie2 promoter. Tie2CRE/+;Bmp2tg/tg mice develop aortic valve dysfunction postnatally, accompanied by pre-calcific lesion formation in valve leaflets. Remarkably, Tie2CRE/+;Bmp2tg/tg mice develop extensive soft tissue bone formation typical of acquired forms of heterotopic ossification (HO) and genetic bone disorders, such as Fibrodysplasia Ossificans Progressiva (FOP). Ectopic ossification in Tie2CRE/+;Bmp2tg/tg transgenic animals is accompanied by increased bone marrow hematopoietic, fibroblast and osteoblast precursors and circulating pro-inflammatory cells. Transplanting wild-type bone marrow hematopoietic stem cells into lethally irradiated Tie2CRE/+;Bmp2tg/tg mice significantly delays HO onset but does not prevent it. Moreover, transplanting Bmp2-transgenic bone marrow into wild-type recipients does not result in HO, but hematopoietic progenitors contribute to inflammation and ectopic bone marrow colonization rather than to endochondral ossification. Conversely, aberrant Bmp2 signaling activity is associated with fibroblast accumulation, skeletal muscle fiber damage, and expansion of a Tie2+ fibro-adipogenic precursor cell population, suggesting that ectopic bone derives from a skeletal muscle resident osteoprogenitor cell origin. Thus, Tie2CRE/+;Bmp2tg/tg mice recapitulate HO pathophysiology, and might represent a useful model to investigate therapies seeking to mitigate disorders associated with aberrant extra-skeletal bone formation.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Linhagem da Célula , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Receptor TIE-2/metabolismo , Animais , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/patologia , Valva Aórtica/fisiopatologia , Transplante de Medula Óssea , Proteína Morfogenética Óssea 2/sangue , Calcinose/diagnóstico por imagem , Calcinose/patologia , Calcinose/fisiopatologia , Condrogênese , Células Endoteliais/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Estimativa de Kaplan-Meier , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Musculares/patologia , Ossificação Heterotópica/sangue , Ossificação Heterotópica/diagnóstico por imagem , Osteogênese , Tomografia Computadorizada por Raios X
2.
Cell Stem Cell ; 25(3): 407-418.e6, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31303548

RESUMO

Hematopoietic stem cells (HSCs) residing in the bone marrow (BM) accumulate during aging but are functionally impaired. However, the role of HSC-intrinsic and -extrinsic aging mechanisms remains debated. Megakaryocytes promote quiescence of neighboring HSCs. Nonetheless, whether megakaryocyte-HSC interactions change during pathological/natural aging is unclear. Premature aging in Hutchinson-Gilford progeria syndrome recapitulates physiological aging features, but whether these arise from altered stem or niche cells is unknown. Here, we show that the BM microenvironment promotes myelopoiesis in premature/physiological aging. During physiological aging, HSC-supporting niches decrease near bone but expand further from bone. Increased BM noradrenergic innervation promotes ß2-adrenergic-receptor(AR)-interleukin-6-dependent megakaryopoiesis. Reduced ß3-AR-Nos1 activity correlates with decreased endosteal niches and megakaryocyte apposition to sinusoids. However, chronic treatment of progeroid mice with ß3-AR agonist decreases premature myeloid and HSC expansion and restores the proximal association of HSCs to megakaryocytes. Therefore, normal/premature aging of BM niches promotes myeloid expansion and can be improved by targeting the microenvironment.


Assuntos
Senilidade Prematura/patologia , Envelhecimento/fisiologia , Medula Óssea/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Megacariócitos/fisiologia , Células Mieloides/fisiologia , Progéria/patologia , Agonistas Adrenérgicos/administração & dosagem , Envelhecimento/metabolismo , Senilidade Prematura/metabolismo , Animais , Diferenciação Celular , Encapsulamento de Células , Proliferação de Células , Modelos Animais de Doenças , Humanos , Interleucina-6/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo I/metabolismo , Progéria/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Nicho de Células-Tronco
3.
Front Physiol ; 10: 159, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30881310

RESUMO

Calcium is an important second messenger required not only for the excitation-contraction coupling of the heart but also critical for the activation of cell signaling pathways involved in the adverse cardiac remodeling and consequently for the heart failure. Sustained neurohumoral activation, pressure-overload, or myocardial injury can cause pathologic hypertrophic growth of the heart followed by interstitial fibrosis. The consequent heart's structural and molecular adaptation might elevate the risk of developing heart failure and malignant arrhythmia. Compelling evidences have demonstrated that Ca2+ entry through TRP channels might play pivotal roles in cardiac function and pathology. TRP proteins are classified into six subfamilies: TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), TRPML (mucolipin), and TRPP (polycystin), which are activated by numerous physical and/or chemical stimuli. TRP channels participate to the handling of the intracellular Ca2+ concentration in cardiac myocytes and are mediators of different cardiovascular alterations. This review provides an overview of the current knowledge of TRP proteins implication in the pathologic process of some frequent cardiac diseases associated with the adverse cardiac remodeling such as cardiac hypertrophy, fibrosis, and conduction alteration.

4.
Oncotarget ; 8(4): 6700-6717, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28036268

RESUMO

The hypoxia-inducible factor 1α (HIF-1α) and its microRNA target, miR-210, are candidate tumor-drivers of metabolic reprogramming in cancer. Neuroendocrine neoplasms such as paragangliomas (PGLs) are particularly appealing for understanding the cancer metabolic adjustments because of their associations with deregulations of metabolic enzymes, such as succinate dehydrogenase (SDH), and the von Hippel Lindau (VHL) gene involved in HIF-1α stabilization. However, the role of miR-210 in the pathogenesis of SDH-related tumors remains an unmet challenge. Herein is described an in vivo genetic analysis of the role of VHL, HIF1A and SDH on miR-210 by using knockout murine models, siRNA gene silencing, and analyses of human tumors. HIF-1α knockout abolished hypoxia-induced miR-210 expression in vivo but did not alter its constitutive expression in paraganglia. Normoxic miR-210 levels substantially increased by complete, but not partial, VHL silencing in paraganglia of knockout VHL-mice and by over-expression of p76del-mutated pVHL. Similarly, VHL-mutated PGLs, not those with decreased VHL-gene/mRNA dosage, over-expressed miR-210 and accumulate HIF-1α in most tumor cells. Ablation of SDH activity in SDHD-null cell lines or reduction of the SDHD or SDHB protein levels elicited by siRNA-induced gene silencing did not induce miR-210 whereas the presence of SDH mutations in PGLs and tumor-derived cell lines was associated with mild increase of miR-210 and the presence of a heterogeneous, HIF-1α-positive and HIF-1α-negative, tumor cell population. Thus, activation of HIF-1α is likely an early event in VHL-defective PGLs directly linked to VHL mutations, but it is a late event favored but not directly triggered by SDHx mutations. This combined analysis provides insights into the mechanisms of HIF-1α/miR-210 regulation in normal and tumor tissues potentially useful for understanding the pathogenesis of cancer and other diseases sharing similar underpinnings.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/genética , Paraganglioma/genética , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Hipóxia Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Adulto , Animais , Linhagem Celular Tumoral , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos Knockout , MicroRNAs/metabolismo , Mutação , Paraganglioma/enzimologia , Paraganglioma/patologia , Fenótipo , Estabilidade Proteica , Interferência de RNA , Transdução de Sinais , Transfecção , Microambiente Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
5.
J Clin Invest ; 124(7): 3230-40, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24937433

RESUMO

Autonomic sympathetic nerves innervate peripheral resistance arteries, thereby regulating vascular tone and controlling blood supply to organs. Despite the fundamental importance of blood flow control, how sympathetic arterial innervation develops remains largely unknown. Here, we identified the axon guidance cue netrin-1 as an essential factor required for development of arterial innervation in mice. Netrin-1 was produced by arterial smooth muscle cells (SMCs) at the onset of innervation, and arterial innervation required the interaction of netrin-1 with its receptor, deleted in colorectal cancer (DCC), on sympathetic growth cones. Function-blocking approaches, including cell type-specific deletion of the genes encoding Ntn1 in SMCs and Dcc in sympathetic neurons, led to severe and selective reduction of sympathetic innervation and to defective vasoconstriction in resistance arteries. These findings indicate that netrin-1 and DCC are critical for the control of arterial innervation and blood flow regulation in peripheral organs.


Assuntos
Artérias Mesentéricas/inervação , Fatores de Crescimento Neural/fisiologia , Sistema Nervoso Simpático/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Animais Recém-Nascidos , Receptor DCC , Feminino , Cones de Crescimento/fisiologia , Masculino , Artérias Mesentéricas/crescimento & desenvolvimento , Artérias Mesentéricas/fisiologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Modelos Neurológicos , Miócitos de Músculo Liso/fisiologia , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Netrina-1 , Gravidez , Receptores de Superfície Celular/fisiologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Vasoconstrição/fisiologia
7.
Cell Rep ; 3(5): 1714-24, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23623496

RESUMO

Strategies for expanding hematopoietic stem cells (HSCs) include coculture with cells that recapitulate their natural microenvironment, such as bone marrow stromal stem/progenitor cells (BMSCs). Plastic-adherent BMSCs may be insufficient to preserve primitive HSCs. Here, we describe a method of isolating and culturing human BMSCs as nonadherent mesenchymal spheres. Human mesenspheres were derived from CD45- CD31- CD71- CD146+ CD105+ nestin+ cells but could also be simply grown from fetal and adult BM CD45--enriched cells. Human mesenspheres robustly differentiated into mesenchymal lineages. In culture conditions where they displayed a relatively undifferentiated phenotype, with decreased adherence to plastic and increased self-renewal, they promoted enhanced expansion of cord blood CD34+ cells through secreted soluble factors. Expanded HSCs were serially transplantable in immunodeficient mice and significantly increased long-term human hematopoietic engraftment. These results pave the way for culture techniques that preserve the self-renewal of human BMSCs and their ability to support functional HSCs.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD/metabolismo , Células da Medula Óssea/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Técnicas de Cocultura , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Nestina/metabolismo
8.
Dev Cell ; 22(3): 489-500, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22421041

RESUMO

Activin receptor-like kinase 1 (ALK1) is an endothelial-specific member of the TGF-ß/BMP receptor family that is inactivated in patients with hereditary hemorrhagic telangiectasia (HHT). How ALK1 signaling regulates angiogenesis remains incompletely understood. Here we show that ALK1 inhibits angiogenesis by cooperating with the Notch pathway. Blocking Alk1 signaling during postnatal development in mice leads to retinal hypervascularization and the appearance of arteriovenous malformations (AVMs). Combined blockade of Alk1 and Notch signaling further exacerbates hypervascularization, whereas activation of Alk1 by its high-affinity ligand BMP9 rescues hypersprouting induced by Notch inhibition. Mechanistically, ALK1-dependent SMAD signaling synergizes with activated Notch in stalk cells to induce expression of the Notch targets HEY1 and HEY2, thereby repressing VEGF signaling, tip cell formation, and endothelial sprouting. Taken together, these results uncover a direct link between ALK1 and Notch signaling during vascular morphogenesis that may be relevant to the pathogenesis of HHT vascular lesions.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Malformações Arteriovenosas/enzimologia , Neovascularização Fisiológica/fisiologia , Receptores Notch/fisiologia , Receptores de Ativinas Tipo I/antagonistas & inibidores , Receptores de Activinas Tipo II , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Ciclo Celular/fisiologia , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Repressoras/fisiologia , Retina/crescimento & desenvolvimento , Retina/patologia , Transdução de Sinais , Proteínas Smad/fisiologia , Telangiectasia Hemorrágica Hereditária/tratamento farmacológico , Telangiectasia Hemorrágica Hereditária/fisiopatologia , Fatores de Crescimento do Endotélio Vascular/fisiologia
9.
Dev Cell ; 20(1): 33-46, 2011 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21238923

RESUMO

Robo4 is an endothelial cell-specific member of the Roundabout axon guidance receptor family. To identify Robo4 binding partners, we performed a protein-protein interaction screen with the Robo4 extracellular domain. We find that Robo4 specifically binds to UNC5B, a vascular Netrin receptor, revealing unexpected interactions between two endothelial guidance receptors. We show that Robo4 maintains vessel integrity by activating UNC5B, which inhibits signaling downstream of vascular endothelial growth factor (VEGF). Function-blocking monoclonal antibodies against Robo4 and UNC5B increase angiogenesis and disrupt vessel integrity. Soluble Robo4 protein inhibits VEGF-induced vessel permeability and rescues barrier defects in Robo4(-/-) mice, but not in mice treated with anti-UNC5B. Thus, Robo4-UNC5B signaling maintains vascular integrity by counteracting VEGF signaling in endothelial cells, identifying a novel function of guidance receptor interactions in the vasculature.


Assuntos
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Neovascularização Patológica/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/enzimologia , Permeabilidade Capilar/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Ligantes , Camundongos , Modelos Biológicos , Receptores de Netrina , Ligação Proteica/efeitos dos fármacos , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Transdução de Sinais/efeitos dos fármacos , Sus scrofa , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
10.
Blood ; 116(19): 4025-33, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20705756

RESUMO

Sprouting of developing blood vessels is mediated by specialized motile endothelial cells localized at the tips of growing capillaries. Following behind the tip cells, endothelial stalk cells form the capillary lumen and proliferate. Expression of the Notch ligand Delta-like-4 (Dll4) in tip cells suppresses tip cell fate in neighboring stalk cells via Notch signaling. In DLL4(+/-) mouse mutants, most retinal endothelial cells display morphologic features of tip cells. We hypothesized that these mouse mutants could be used to isolate tip cells and so to determine their genetic repertoire. Using transcriptome analysis of retinal endothelial cells isolated from DLL4(+/-) and wild-type mice, we identified 3 clusters of tip cell-enriched genes, encoding extracellular matrix degrading enzymes, basement membrane components, and secreted molecules. Secreted molecules endothelial-specific molecule 1, angiopoietin 2, and apelin bind to cognate receptors on endothelial stalk cells. Knockout mice and zebrafish morpholino knockdown of apelin showed delayed angiogenesis and reduced proliferation of stalk cells expressing the apelin receptor APJ. Thus, tip cells may regulate angiogenesis via matrix remodeling, production of basement membrane, and release of secreted molecules, some of which regulate stalk cell behavior.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica/genética , Proteínas Adaptadoras de Transdução de Sinal , Adipocinas , Animais , Apelina , Receptores de Apelina , Proteínas de Ligação ao Cálcio , Capilares/citologia , Capilares/crescimento & desenvolvimento , Capilares/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Haploinsuficiência , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Vasos Retinianos/citologia , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Peixe-Zebra
11.
Genes Dev ; 21(19): 2433-47, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908930

RESUMO

Netrins are secreted molecules with roles in axonal growth and angiogenesis. The Netrin receptor UNC5B is required during embryonic development for vascular patterning, suggesting that it may also contribute to postnatal and pathological angiogenesis. Here we show that unc5b is down-regulated in quiescent adult vasculature, but re-expressed during sprouting angiogenesis in matrigel and tumor implants. Stimulation of UNC5B-expressing neovessels with an agonist (Netrin-1) inhibits sprouting angiogenesis. Genetic loss of function of unc5b reduces Netrin-1-mediated angiogenesis inhibition. Expression of UNC5B full-length receptor also triggers endothelial cell repulsion in response to Netrin-1 in vitro, whereas a truncated UNC5B lacking the intracellular signaling domain fails to induce repulsion. These data show that UNC5B activation inhibits sprouting angiogenesis, thus identifying UNC5B as a potential anti-angiogenic target.


Assuntos
Neovascularização Patológica/metabolismo , Fatores de Crescimento Neural/farmacologia , Receptores de Superfície Celular/agonistas , Proteínas Supressoras de Tumor/farmacologia , Animais , Colágeno/metabolismo , Combinação de Medicamentos , Endotélio Vascular/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Mutantes , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Receptores de Netrina , Netrina-1 , Proteoglicanas/metabolismo , Pseudópodes/efeitos dos fármacos , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
12.
J Biol Chem ; 278(25): 22316-24, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12679337

RESUMO

Cellular responses to hypoxia can be acute or chronic. Acute responses mainly depend on oxygen-sensitive ion channels, whereas chronic responses rely on the hypoxia-inducible transcription factors (HIFs), which up-regulate the expression of enzymes, transporters, and growth factors. It is unknown whether the expression of genes coding for ion channels is also influenced by hypoxia. We report here that the alpha1H gene of T-type voltage-gated calcium channels is highly induced by lowering oxygen tension in PC12 cells. Accumulation of alpha1H mRNA in response to hypoxia is time- and dose-dependent and paralleled by an increase in the density of T-type calcium channel current recorded in patch clamped cells. HIF appears to be involved in the response to hypoxia, since cobalt chloride, desferrioxamine, and dimethyloxalylglycine, compounds that mimic HIF-regulated gene expression, replicate the hypoxic effect. Moreover, functional inhibition of HIF-2alpha protein accumulation using antisense HIF-2alpha oligonucleotides reverses the effect of hypoxia on T-type Ca2+ channel expression. Importantly, regulation by oxygen tension is specific for T-type calcium channels, since it is not observed with the L-, N-, and P/Q-channel types. These findings show for the first time that hypoxia induces an ion channel gene via a HIF-dependent mechanism and define a new role for the T-type calcium channels as regulators of cellular excitability and calcium influx under chronic hypoxia.


Assuntos
Canais de Cálcio Tipo T/genética , Hipóxia Celular/fisiologia , Animais , Sequência de Bases , Encéfalo/fisiologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/fisiologia , Primers do DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Células PC12 , Feocromocitoma , Ratos , Proteínas Recombinantes/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tionucleotídeos/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA