Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Alzheimers Dis Rep ; 7(1): 469-473, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37313494

RESUMO

We describe a 52-year-old patient with a progressive visuospatial disorder and apraxia. Neuropsychological assessment, neuroradiological findings, and Alzheimer's disease (AD) core biomarker assay on cerebrospinal fluid led to a diagnosis of posterior cortical atrophy due to AD. We performed a next generation sequencing dementia-gene panel and found the c.1301 C>T p.(Ala434Val) variant in the Presenilin1 (PSEN1) gene. The missense change affects the PAL (Pro433-Ala434-Leu435) motif critical for catalytic activity of the macromolecular γ-secretase complex. Evolutionary and integrated bioinformatic tools predicted a deleterious effect of the variant supporting its role in the AD pathogenesis.

2.
Arterioscler Thromb Vasc Biol ; 30(8): 1562-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20489169

RESUMO

OBJECTIVE: Inflammatory stimuli released into atherosclerotic plaque microenvironment regulate vessel formation by modulating gene expression and translation. microRNAs are a class of short noncoding RNAs, acting as posttranscriptional regulators of protein-coding genes involved in various biological processes, including vascular cell biology. Among them, microRNA-221/222 (miR-221/222) seem to negatively modulate vascular remodeling by targeting different target genes. Here, we investigated their potential contribution to inflammation-mediated neovessel formation. METHODS AND RESULTS: We used quantitative real-time RT-PCR amplification to analyze expression of 7 microRNAs previously linked to vascular biology, such as miR-17-5p, miR-21, miR-126, miR-210, miR-221, miR-222, and miR-296 and found high levels of expression for all of them in quiescent endothelial cells. However, miR-126, miR-221, miR-222, and miR-296 turned out to be down-modulated in endothelial cells exposed to inflammatory stimuli. Applying a gain-of-function approach, we demonstrated that, among them, only miR-222 was involved in inflammation-mediated vascular remodeling. In addition, we identified signal transducer and activator of transcription 5A (STAT5A) as a bona fide target of miR-222 and observed that miR-222 negatively correlated with STAT5A expression in human endothelial cells from advanced neovascularized atherosclerotic lesions. CONCLUSIONS: We identified STAT5A as a novel miR-222 target, and this finding opens up new perspectives for treatment of vascular diseases.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , MicroRNAs/metabolismo , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica , Fator de Transcrição STAT5/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regiões 3' não Traduzidas , Animais , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Células Cultivadas , Regulação para Baixo , Células Endoteliais/patologia , Células Endoteliais/transplante , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Interleucina-3/metabolismo , Camundongos , Camundongos SCID , MicroRNAs/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/genética , Interferência de RNA , Fator de Transcrição STAT5/genética , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/genética
3.
Blood ; 112(2): 350-61, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18460645

RESUMO

Interleukin-3 (IL-3) released by infiltrating inflammatory cells in different pathologic settings contributes to organ and tumor angiogenesis. Here we demonstrate that IL-3 expands a subset of CD45+ circulating angiogenic cells clonally derived from the hemopoietic progenitors. Moreover, CD45+ cells exposed to IL-3 acquire arterial specification and contribute to the formation of vessels in vivo. Depletion of signal transducer and activator of transcription 5 (STAT5) provides evidence that IL-3-mediated cell expansion and arterial morphogenesis rely on STAT5 activation. In addition, by means of Tie2-transgenic mice, we demonstrate that STAT5 also regulates IL-3-induced expansion and arterial specification of bone marrow-derived CD45+ cells. Thus, our data provide the first evidence that, in inflammatory microenvironments containing IL-3, angiogenic cells derived from hemopoietic precursors can act as adult vasculogenic cells. Moreover, the characterization of the signaling pathway regulating these events provides the rationale for therapeutically targeting STAT5 in these pathologic settings.


Assuntos
Artérias/crescimento & desenvolvimento , Sistema Hematopoético/citologia , Interleucina-3/fisiologia , Antígenos Comuns de Leucócito , Neovascularização Fisiológica , Fator de Transcrição STAT5/metabolismo , Animais , Células da Medula Óssea , Proliferação de Células , Humanos , Camundongos , Células-Tronco
4.
J Cell Biol ; 168(7): 1099-108, 2005 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-15795318

RESUMO

We previously demonstrated that integrin-dependent adhesion activates STAT5A, a well known target of IL-3-mediated signaling. Here, we show that in endothelial cells the active beta1 integrin constitutively associates with the unphosphorylated IL-3 receptor (IL-3R) beta common subunit. This association is not sufficient for activating downstream signals. Indeed, only upon fibronectin adhesion is Janus Kinase 2 (JAK2) recruited to the beta1 integrin-IL-3R complex and triggers IL-3R beta common phosphorylation, leading to the formation of docking sites for activated STAT5A. These events are IL-3 independent but require the integrity of the IL-3R beta common. IL-3 treatment increases JAK2 activation and STAT5A and STAT5B tyrosine and serine phosphorylation and leads to cell cycle progression in adherent cells. Expression of an inactive STAT5A inhibits cell cycle progression upon IL-3 treatment, identifying integrin-dependent STAT5A activation as a priming event for IL-3-mediated S phase entry. Consistently, overexpression of a constitutive active STAT5A leads to anchorage-independent cell cycle progression. Therefore, these data provide strong evidence that integrin-dependent STAT5A activation controls IL-3-mediated proliferation.


Assuntos
Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Proteínas do Leite/metabolismo , Receptores de Interleucina-3/metabolismo , Transativadores/metabolismo , Adesão Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Fibronectinas/metabolismo , Humanos , Interleucina-3/metabolismo , Interleucina-3/farmacologia , Janus Quinase 2 , Proteínas do Leite/genética , Fosforilação/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fase S/fisiologia , Fator de Transcrição STAT5 , Transdução de Sinais/fisiologia , Transativadores/genética , Ativação Transcricional/fisiologia , Proteínas Supressoras de Tumor
5.
Arterioscler Thromb Vasc Biol ; 29(1): 114-20, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18927468

RESUMO

OBJECTIVE: Circulating angiogenic cells (CACs) expansion is a multistage process requiring sequential activation of transcriptional factors, including STAT5. STAT5, in concert with peroxisome proliferator-activated receptors (PPARs), seems to induce discrete biological responses in different tissues. In the present study we investigated the role of STAT5 and PPARgamma in regulating CAC expansion in normal and diabetic settings. METHODS AND RESULTS: Normal and diabetic CACs were used. siRNA technology, EMSA, and chromatin immunoprecipitation (ChIP) assay as well as site-directed mutagenesis of the STAT5 response element in the PPARgamma promoter enabled us to demonstrate that STAT5 transcriptional activity controls PPARgamma expression. Moreover, FACS analysis, coimmunoprecipitation experiments, and ChIP assay revealed that a STAT5/PPARgamma transcriptional complex controls cyclin D1 expression and CAC progression into the cell-cycle. Conversely, PPARgamma agonists, by preventing the expression of STAT5 and the formation of the STAT5/PPARgamma heterodimeric complex failed to promote CAC expansion. Finally, we demonstrated that diabetic CAC functional capability can be recovered by molecules able to activate the STAT5/PPARgamma transcriptional complex. CONCLUSIONS: Our data identify the STAT5/PPARgamma heterodimers as landmark of CAC expansion and provide evidences for a mechanism that partially rescues CAC bioavailability in diabetic setting.


Assuntos
Diabetes Mellitus Tipo 2/genética , Angiopatias Diabéticas/genética , Neovascularização Patológica/genética , PPAR gama/fisiologia , Fator de Transcrição STAT5/genética , Transcrição Gênica , Animais , Células da Medula Óssea/fisiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/fisiopatologia , Angiopatias Diabéticas/fisiopatologia , Feminino , Genes Reporter , Humanos , Luciferases/genética , Masculino , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , PPAR gama/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Fator de Transcrição STAT5/fisiologia
6.
BMJ Case Rep ; 13(12)2020 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-33318253

RESUMO

A 4-year-old girl was referred to the geneticist with a history of ataxia associated with intention tremor of the hands, strabismus and hypermetropy. Her symptoms presented about 2 years earlier with inability to walk unaided and lower limbs hypotonia. Cognitive functions were normal. Brain MRI showed a cerebellar and vermian hypoplasia with enlargement of both the cerebrospinal fluid spaces and the IV brain ventricle. Family history was unremarkable. A genetic screening using a 42-gene panel for hereditary ataxia/spastic paraparesis identified a de novo c.1438C>T - p.(Arg480Trp) missense change in the SPTBN2 gene (NM_006946.2). This variant is reported to be associated with congenital ataxia, later evolving into ataxia and intellectual disability. This case further supports the existence of a specific SPTBN2 p.(Arg480Trp)-associated phenotype, with a de novo recurrence of this variant in the heterozygous state.


Assuntos
Encéfalo/patologia , Espectrina/genética , Ataxias Espinocerebelares/genética , Pré-Escolar , Feminino , Humanos , Deficiência Intelectual , Imageamento por Ressonância Magnética , Mutação de Sentido Incorreto , Fenótipo , Espectrina/metabolismo
7.
ScientificWorldJournal ; 8: 1111-5, 2008 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-18979051

RESUMO

The manuscript deals with the effects exerted by the inflammatory microenvironment on angiogenic cell expansion and arterial specification. Data reported demonstrated that the inflammatory cytokine IL-3 expands a subset of hemopoietic-derived cells, which directly contribute to neovessel formation. In addition, the identification of STAT5 as a molecular signature of the vasculogenetic process provides the rationale for a novel therapeutic strategy to manipulate vascular growth.


Assuntos
Artérias/crescimento & desenvolvimento , Inflamação/patologia , Neovascularização Patológica/fisiopatologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Interleucina-3/fisiologia , Camundongos , Camundongos SCID , Fator de Transcrição STAT5/fisiologia
8.
Oncogene ; 37(9): 1175-1191, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29238040

RESUMO

The proangiogenic cytokine Interleukin-3 (IL-3) is released by inflammatory cells in breast and ovarian cancer tissue microenvironments and also acts as an autocrine factor for human breast and kidney tumor-derived endothelial cells (TECs). We have previously shown that IL-3-treated endothelial cells (ECs) release extracellular vesicles (EVs), which serve as a paracrine mechanism for neighboring ECs, by transferring active molecules. The impact of an anti-IL-3R-alpha blocking antibody on the proangiogenic effect of EVs released from TECs (anti-IL-3R-EVs) has therefore been investigated in this study. We have found that anti-IL-3R-EV treatment prevented neovessel formation and, more importantly, also induced the regression of in vivo TEC-derived neovessels. Two miRs that target the canonical wingless (Wnt)/ß-catenin pathway, at different levels, were found to be differentially regulated when comparing the miR-cargo of naive TEC-derived EVs (EVs) and anti-IL-3R-EVs. miR-214-3p, which directly targets ß-catenin, was found to be upregulated, whereas miR-24-3p, which targets adenomatous polyposis coli (APC) and glycogen synthase kinase-3ß (GSK3ß), was found to be downregulated. In fact, upon their transfer into the cell, low ß-catenin content and high levels of the two members of the "ß-catenin destruction complex" were detected. Moreover, c-myc downregulation was found in TECs treated with anti-IL-3R-EVs, pre-miR-214-3p-EVs and antago-miR-24-3p-EVs, which is consistent with network analyses of miR-214-3p and miR-24-3p gene targeting. Finally, in vivo studies have demonstrated the impaired growth of vessels in pre-miR-214-3p-EV- and antago-miR-24-3p-EV-treated animals. These effects became much more evident when combo treatment was applied. The results of the present study identify the canonical Wnt/ß-catenin pathway as a relevant mechanism of TEC-derived EV proangiogenic action. Furthermore, we herein provide evidence that IL-3R blockade may yield some significant advantages, than miR targeting, in inhibiting the proangiogenic effects of naive TEC-derived EVs by changing TEC-EV-miR cargo.


Assuntos
Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Células Endoteliais/patologia , Vesículas Extracelulares/patologia , Subunidade alfa de Receptor de Interleucina-3/antagonistas & inibidores , beta Catenina/antagonistas & inibidores , Animais , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-3/genética , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Camundongos , Camundongos SCID , MicroRNAs/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética , beta Catenina/metabolismo
9.
Diabetes ; 67(4): 704-716, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29386225

RESUMO

Endothelial cell-derived extracellular vesicles (CD31EVs) constitute a new entity for therapeutic/prognostic purposes. The roles of CD31EVs as mediators of vascular smooth muscle cell (VSMC) dysfunction in type 2 diabetes (T2D) are investigated herein. We demonstrated that, unlike serum-derived extracellular vesicles in individuals without diabetes, those in individuals with diabetes (D CD31EVs) boosted apoptosis resistance of VSMCs cultured in hyperglycemic condition. Biochemical analysis revealed that this effect relies on changes in the balance between antiapoptotic and proapoptotic signals: increase of bcl-2 and decrease of bak/bax. D CD31EV cargo analysis demonstrated that D CD31EVs are enriched in membrane-bound platelet-derived growth factor-BB (mbPDGF-BB). Thus, we postulated that mbPDGF-BB transfer by D CD31EVs could account for VSMC resistance to apoptosis. By depleting CD31EVs of platelet-derived growth factor-BB (PDGF-BB) or blocking the PDGF receptor ß on VSMCs, we demonstrated that mbPDGF-BB contributes to D CD31EV-mediated bak/bax and bcl-2 levels. Moreover, we found that bak expression is under the control of PDGF-BB-mediated microRNA (miR)-296-5p expression. In fact, while PDGF-BB treatment recapitulated D CD31EV-mediated antiapoptotic program and VSMC resistance to apoptosis, PDGF-BB-depleted CD31EVs failed. D CD31EVs also increased VSMC migration and recruitment to neovessels by means of PDGF-BB. Finally, we found that VSMCs, from human atherosclerotic arteries of individuals with T2D, express low bak/bax and high bcl-2 and miR-296-5p levels. This study identifies the mbPDGF-BB in D CD31EVs as a relevant mediator of diabetes-associated VSMC resistance to apoptosis.


Assuntos
Apoptose , Aterosclerose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Vesículas Extracelulares/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Idoso , Idoso de 80 Anos ou mais , Becaplermina , Estudos de Casos e Controles , Feminino , Humanos , Masculino , MicroRNAs/metabolismo , Músculo Liso Vascular/citologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
10.
J Clin Invest ; 109(1): 111-9, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11781356

RESUMO

Modified LDL is a major cause of injury to the endothelium in diabetes. In the present study, we analyzed the effects on endothelial cells of LDL recovered from type 2 diabetic patients (dm-LDL) or from nondiabetic subjects (n-LDL). Treatment of human umbilical vein endothelial cells with dm-LDL, but not n-LDL, led to the accumulation of cells in G1. To dissect the molecular mechanisms of this effect, we analyzed the expression and function of the cyclin-dependent kinase inhibitor p21(waf), a cell cycle regulator known to be a target of the signal transducers and activators of transcription (STATs). dm-LDL led to transient STAT5 phosphorylation and the formation of a STAT5-containing complex and activated p21(waf) expression at the transcriptional level. Expression of the dominant-negative form of STAT5B, but not of STAT5A, significantly decreased both p21(waf) expression and the fraction of cells in G1. Finally, immunofluorescence analysis demonstrated that activated STAT5 is expressed in newly formed intraplaque vessels and in endothelial cells lining the luminal side of the plaque. Similarly, p21(waf) immunoreactivity was found in the neointimal vasculature. Our results suggest a role of STAT5B as a regulator of gene expression in diabetes-associated vascular disease.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular/efeitos dos fármacos , Ciclinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/sangue , Lipoproteínas LDL/sangue , Lipoproteínas LDL/farmacologia , Proteínas do Leite , Transativadores/metabolismo , Idoso , Arteriosclerose/sangue , Arteriosclerose/etiologia , Arteriosclerose/patologia , Estudos de Casos e Controles , Ciclo Celular/fisiologia , Células Cultivadas , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Angiopatias Diabéticas/sangue , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipoproteínas LDL/isolamento & purificação , Masculino , Pessoa de Meia-Idade , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT5 , Transativadores/genética , Proteínas Supressoras de Tumor
11.
Oncogene ; 24(42): 6394-405, 2005 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-16007196

RESUMO

As vascular endothelial growth factor (VEGF), interleukin-3 (IL-3), released into the tumor microenvironment stimulates motogenic and mitogenic activity of normal and transformed cells. In the present study, we investigate the effects of IL-3 and VEGF on neoplastic vascular growth. Engagement of IL-3 receptor beta common (IL-3R beta c) contributes to both IL-3- and VEGF-induced Rac1 activation, cell migration and in vitro tube-like structure formation as shown by the expression of the dominant-negative IL-3R beta c construct (Delta455). In normal and transformed endothelial cells (EC) as well as in HEK 293 cells expressing KDR and IL-3R, VEGF and IL-3 treatment induces the formation of a KDR/IL-3R beta c complex. Moreover, as shown by the IL-3R Delta455 mutant or by the kinase dead KDR, functional receptors are required for this interaction. Consistent with the contribution of IL-3R beta c in both IL-3- and VEGF-mediated angiogenic signal, a reduced number of vessels inside tumors are found in mice injected with cells expressing the IL-3R Delta455 mutant. Thus, these findings provide a novel mechanism through which IL-3 and VEGF support cell survival and tumor neovascularization.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica , Receptores de Interleucina-3/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Sequência de Bases , Western Blotting , Linhagem Celular , Primers do DNA , Humanos , Imunoprecipitação , Interleucina-3/fisiologia , Invasividade Neoplásica , Fator A de Crescimento do Endotélio Vascular/fisiologia
12.
PLoS One ; 11(9): e0162417, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27611075

RESUMO

Extracellular vesicles (EVs) that are derived from stem cells are proving to be promising therapeutic options. We herein investigate the therapeutic potential of EVs that have been derived from different stem cell sources, bone-marrow (MSC) and human liver (HLSC), on mesangial cells (MCs) exposed to hyperglycaemia. By expressing a dominant negative STAT5 construct (ΔNSTAT5) in HG-cultured MCs, we have demonstrated that miR-21 expression is under the control of STAT5, which translates into Transforming Growth Factor beta (TGFß) expression and collagen production. A number of approaches have been used to show that both MSC- and HLSC-derived EVs protect MCs from HG-induced damage via the transfer of miR-222. This resulted in STAT5 down-regulation and a decrease in miR-21 content, TGFß expression and matrix protein synthesis within MCs. Moreover, we demonstrate that changes in the balance between miR-21 and miR-100 in the recipient cell, which are caused by the transfer of EV cargo, further contribute to providing beneficial effects. Interestingly, these effects were only detected in HG-cultured cells. Finally, it was found that HG reduced the expression of the nuclear encoded mitochondrial electron transport chain (ETC) components, CoxIV. It is worth noting that EV administration can rescue CoxIV expression in HG-cultured MCs. These results thus demonstrate that both MSC- and HLSC-derived EVs transfer the machinery needed to preserve MCs from HG-mediated damage. This occurs via the horizontal transfer of functional miR-222 which directly interferes with damaging cues. Moreover, our data indicate that the release of EV cargo into recipient cells provides additional therapeutic advantages against harmful mitochondrial signals.


Assuntos
Colágeno/biossíntese , Vesículas Extracelulares/metabolismo , Hiperglicemia/metabolismo , Células Mesangiais/metabolismo , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo IV/metabolismo , Glucose/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Modelos Biológicos , Processamento Pós-Transcricional do RNA , Fator de Transcrição STAT5/metabolismo
13.
Sci Rep ; 6: 25689, 2016 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-27157262

RESUMO

Soluble factors and cell-derived extracellular vesicles (EVs) control vascular cell fate during inflammation. The present study investigates the impact of Interleukin 3 (IL-3) on EV release by endothelial cells (ECs), the mechanisms involved in EV release and paracrine actions. We found that IL-3 increases EV release, which is prevented by IL-3Ralpha blockade. EVs released upon IL-3 stimulation were able to induce pro-angiogenic signals as shown by chromatin immunoprecipitation (ChIP) assay performed on the promoter region of cyclin D1 and tridimensional tube-like structure formation. We herein demonstrate that these effects rely on the transfer of miR-126-3p, pre-miR-126 and, more importantly, of activated signal transduction and activator of transcription 5 (pSTAT5) from IL-3-EV cargo into recipient ECs. We show, using the dominant negative form (ΔN)STAT5 and an activated STAT5 (1*6STAT5) constructs, that STAT5 drives IL-3-mediated EV release, miR-126-3p and pSTAT5 content. Finally, using EVs recovered from ΔNSTAT5 expressing ECs, we provide evidence that miR-126-3p and pSTAT5 trafficking is relevant for IL-3-mediated paracrine pro-angiogenic signals. These results indicate that IL-3 regulates EC-EV release, cargo and IL-3 angiogenic paracrine action via STAT5. Moreover, these results provide evidence that EC-derived IL-3-EVs can serve as pro-angiogenic clinical delivery wound healing devices.


Assuntos
Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Interleucina-3/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Fator de Transcrição STAT5/metabolismo , Extratos Celulares , Ciclina D1/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vesículas Extracelulares/ultraestrutura , Humanos , MicroRNAs/metabolismo , Modelos Biológicos , Fosforilação , Transporte Proteico
14.
Oncogene ; 23(9): 1681-92, 2004 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-14755254

RESUMO

Interleukin-3 (IL-3) expression by tumor-infiltrating lymphocytes (TILs) and its effects on vessel assembly were evaluated. TILs from 'in situ' human breast cancers expressed CD4/CD25 antigens and IL-3. An injection of Matrigel containing SMC and IL-3 or basic-fibroblast growth factor (bFGF) into SCID mice confirmed the neoangiogenetic effect of both factors. However, in response to IL-3, but not to bFGF, only few SMC became incorporated into the nascent vessels. To evaluate the possibility that signals emanated by the nascent vasculature in the presence of IL-3 may negatively regulate SMC recruitment, conditioned media (CM) from IL-3-treated endothelial cells (EC) or SMC were tested for their biological effects on SMC and EC. CM from IL-3-treated SMC stimulated the migration of EC. In contrast, the migration of SMC was not affected by CM from IL-3-stimulated EC; however, it was greatly enhanced by blocking transforming growth factor beta (TGF beta) activity. TGF beta immunoenzymatic assay demonstrated the following: (i) the absence of TGF beta activity in CM from IL-3-stimulated EC; (ii) a barely detectable TGF beta activity in CM from IL-3-stimulated SMC; and (iii) the presence of TGF beta activity in the supernatants of SMC stimulated with CM from IL-3-, but not from bFGF-stimulated EC. Increased TGF beta mRNA expression was only detected in SMC stimulated with CM from IL-3-treated EC. Finally, the inhibitory signals induced by IL-3 in vivo were abrogated by the addition of the neutralizing TGF beta antibody. Thus, the positive immunostaining for IL-3 by TILs in 'in situ' breast cancers sustains the possibility that early in tumor development, IL-3 can contribute to the chronic immaturity of these vessels.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Interleucina-3/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Neovascularização Patológica , Fator de Crescimento Transformador beta/metabolismo , Animais , Neoplasias da Mama/imunologia , Células Cultivadas , Colágeno , Meios de Cultivo Condicionados/farmacologia , Combinação de Medicamentos , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Interleucina-3/metabolismo , Laminina , Linfócitos/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos SCID , Mitógenos/farmacologia , Modelos Biológicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Proteoglicanas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Diabetes ; 51(11): 3311-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12401724

RESUMO

Advanced glycation end products (AGEs) have been implicated in the accelerated vascular injury occurring in diabetes. We recently reported that LDL prepared from type 2 diabetic patients (dm-LDL), but not normal LDL (n-LDL) triggered signal transducers and activators of transcription STAT5 activation and p21(waf) expression in endothelial cells (ECs). The aims of the present study were to investigate the role of LDL glycation in dm-LDL- mediated signals and to analyze the molecular mechanisms leading to STAT5 activation. We found that glycated LDL (gly-LDL) triggered STAT5 activation, the formation of a prolactin inducible element (PIE)-binding complex containing STAT5, and increased p21(waf) expression through the activation of the receptor for AGE (RAGE). We also demonstrated that dm-LDL and gly-LDL, but not n-LDL treatment induced the formation of a stable complex containing the activated STAT5 and RAGE. Moreover, gly-LDL triggered src but not JAK2 kinase activity. Pretreatment with the src kinase inhibitor PP1 abrogated both STAT5 activation and the expression of p21(waf) induced by gly-LDL. Consistently, gly-LDL failed to activate STAT5 in src(-/-) fibroblasts. Collectively, our results provide evidence for the role of glycation in dm-LDL-mediated effects and for a specific role of src kinase in STAT5-dependent p21(waf) expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus/sangue , Produtos Finais de Glicação Avançada/metabolismo , Lipoproteínas LDL/sangue , Proteínas do Leite , Transativadores/metabolismo , Quinases da Família src/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Glicosilação , Humanos , Proteínas Recombinantes/metabolismo , Valores de Referência , Fator de Transcrição STAT5 , Transfecção , Veias Umbilicais
16.
Inflamm Bowel Dis ; 11(2): 91-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15677901

RESUMO

Crohn's disease (CD) and ulcerative colitis (UC), the two major forms of chronic inflammatory bowel disease (IBD), are characterized by mucosal immune cell activation that is driven by a cytokine imbalance. Several cytokines involved in IBD act through the activation of the signal transducers and activators of transcription (STAT) family. We investigated the activation of STAT3 in the mucosa of CD and UC patients, and evaluated whether this event is specific for IBD patients. Using immunofluorescence and immunoblotting, total and phosphorylated STAT3 levels were assessed in biopsy specimens, isolated lamina propria mononuclear cells, and peripheral blood mononuclear cells from patients with CD, UC, other forms of intestinal inflammation, and control subjects. Immunoblotting revealed phosphorylated STAT3 in mucosal biopsy specimens from patients with CD, UC, celiac disease, and acute self-limited colitis, but not in the normal mucosa of control subjects. In IBD patients, STAT3 activation was confined to actively inflamed areas. Accordingly, activated STAT3 was detected in isolated lamina propria mononuclear cells from inflamed IBD tissues, but not in peripheral blood mononuclear cells from control subjects or IBD patients. Immunofluorescence demonstrated that the sources of activated STAT3 were macrophages and T lymphocytes, but not neutrophils. STAT3 activation also was detected in T cells infiltrating the duodenal mucosa of celiac disease patients. We conclude that STAT3 signaling occurs in both CD and UC, where it is strictly confined to areas of active inflammation and is limited to infiltrating macrophages and T cells. The occurrence of STAT3 signaling in other acute and chronic intestinal inflammatory conditions suggests that, rather than a specific feature of IBD, it represents a fundamental signaling pathway that is shared by multiple forms of gut inflammation.


Assuntos
Doença de Crohn/fisiopatologia , Doenças Inflamatórias Intestinais/fisiopatologia , Transdução de Sinais , Adulto , Estudos de Casos e Controles , Proteínas de Ligação a DNA , Feminino , Humanos , Inflamação , Mucosa Intestinal , Macrófagos , Masculino , Pessoa de Meia-Idade , Fator de Transcrição STAT3 , Linfócitos T , Transativadores , Ativação Transcricional
17.
FASEB J ; 18(11): 1249-51, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15180953

RESUMO

The molecular events associated with acute and chronic exposure of mesangial cells (MC) to hyperglycemia were evaluated. We found that, unlike high glucose (HG) and Amadori adducts, advanced glycation end products (AGE) and transforming growth factor-beta (TGF-beta) induced p21waf expression and accumulation of MC in G0/G1. TGF-beta1 blockade inhibited AGE-mediated collagen production but only partially affected AGE-induced p21waf expression and cell-cycle events, indicating that AGE by binding to AGE receptor (RAGE) per se could control MC growth. Moreover, AGE and TGF-beta treatment led to the activation of the signal transduction and activators of transcription (STAT)5 and the formation of a STAT5/p21SIE2 complex. The role of STAT5 in AGE- and TGF-beta-mediated p21waf expression and growth arrest, but not collagen production, was confirmed by the expression of the dominant negative STAT5 (DeltaSTAT5) or the constitutively activated STAT5 (1*6-STAT5) constructs. Finally, in p21waf-/- fibroblasts both AGE and TGF-beta failed to inhibit cell-cycle progression. A potential in vivo role of these mechanisms was sustained by the increasing immunoreactivity for the activated STAT5 and p21(waf) in kidney biopsies from early to advanced stage of diabetic nephropathy. Our data indicate that AGE- and TGF-beta-mediated signals, by converging on STAT5 activation and p21waf expression, may regulate MC growth.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Nefropatias Diabéticas/metabolismo , Mesângio Glomerular/efeitos dos fármacos , Produtos Finais de Glicação Avançada/farmacologia , Receptores Imunológicos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Albuminúria/metabolismo , Albuminúria/patologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Colágeno/biossíntese , Colágeno/genética , Inibidor de Quinase Dependente de Ciclina p21 , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Nefropatias Diabéticas/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Mesângio Glomerular/citologia , Humanos , Hipertrofia , Proteínas do Leite/genética , Síndrome Nefrótica/metabolismo , Síndrome Nefrótica/patologia , Proteinúria/metabolismo , Proteinúria/patologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/efeitos dos fármacos , Fator de Transcrição STAT5 , Transativadores/deficiência , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
18.
Arterioscler Thromb Vasc Biol ; 24(3): 511-8, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14726409

RESUMO

OBJECTIVE: To characterize the molecules and the mechanisms regulating the neoangiogenetic process in advanced atherosclerotic plaques. METHODS AND RESULTS: Western blot and immunofluorescence analysis of atherosclerotic specimens demonstrated that unlike neovessels from early lesions that expressed vascular endothelial growth factor (VEGF) and angiopoietin1 (Angio1), vessels from advanced lesions expressed VEGF and angiopoietin 2 (Angio2). Moreover, only few neovessels from advanced lesions showed a positive immunostaining for proliferating cell nuclear antigen. Angio1-elicited and Angio2-elicited intracellular events in endothelial cells (EC) demonstrated that while Angio1 triggered Erk1/Erk2 mitogen activated protein kinases (MAPK) and Akt activation, Angio2 (50 ng/mL) induced STAT5 activation and p21waf expression and increased the fraction of cells in G1. Both Angio2-mediated events were abrogated by expressing a dominant negative STAT5 construct (DeltaSTAT5). Consistent with the expression of Angio2 in neovessels of advanced lesions a transcriptionally active STAT5 was detected. Moreover, co-immunoprecipitation experiments revealed the presence of a STAT5/Tie2 molecular complex in neointima vessels from advanced, but not from early, lesions. CONCLUSIONS: In advanced lesions, the activation of the Tie2-mediated STAT5 signaling pathway may negatively regulate vessel growth.


Assuntos
Angiopoietina-2/fisiologia , Arteriosclerose/patologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Células Endoteliais/efeitos dos fármacos , Receptor TIE-2/fisiologia , Transativadores/fisiologia , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Angiopoietina-1/fisiologia , Angiopoietina-2/biossíntese , Angiopoietina-2/genética , Angiopoietina-2/farmacologia , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral/metabolismo , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Doença da Artéria Coronariana/patologia , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/genética , Progressão da Doença , Células Endoteliais/citologia , Endotélio Vascular/patologia , Fase G1/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Substâncias Macromoleculares , Proteínas do Leite/genética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Antígeno Nuclear de Célula em Proliferação/biossíntese , Antígeno Nuclear de Célula em Proliferação/genética , Fator de Transcrição STAT5 , Transdução de Sinais/efeitos dos fármacos , Transativadores/genética , Transcrição Gênica , Transfecção , Túnica Íntima/patologia
19.
J Diabetes Res ; 2015: 373708, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26064979

RESUMO

Vascular complications are major causes of morbidity and mortality in type 2 diabetes patients. Mitochondrial reactive oxygen species (ROS) generation and a lack of efficient antioxidant machinery, a result of hyperglycaemia, mainly contribute to this problem. Although advances in therapy have significantly reduced both morbidity and mortality in diabetic individuals, diabetes-associated vascular complications are still one of the most challenging health problems worldwide. New healing options are urgently needed as current therapeutics are failing to improve long-term outcomes. Particular effort has recently been devoted to understanding the functional relationship between chromatin structure regulation and the persistent change in gene expression which is driven by hyperglycaemia and which accounts for long-lasting diabetic complications. A detailed investigation into epigenetic chromatin modifications in type 2 diabetes is underway. This will be particularly useful in the design of mechanism-based therapeutics which interfere with long-lasting activating epigenetics and improve patient outcomes. We herein provide an overview of the most relevant mechanisms that account for hyperglycaemia-induced changes in chromatin structure; the most relevant mechanism is called "metabolic memory."


Assuntos
Diabetes Mellitus Tipo 2/genética , Angiopatias Diabéticas/terapia , Epigênese Genética , Hiperglicemia/genética , Montagem e Desmontagem da Cromatina/genética , Angiopatias Diabéticas/genética , Humanos
20.
Diabetes ; 64(4): 1370-82, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25368096

RESUMO

Reactive oxygen species (ROS) are crucial in long-term diabetes complications, including peripheral artery disease (PAD). In this study, we have investigated the potential clinical impact of unacylated ghrelin (UnAG) in a glucose intolerance and PAD mouse model. We demonstrate that UnAG is able to protect skeletal muscle and endothelial cells (ECs) from ROS imbalance in hind limb ischemia-subjected ob/ob mice. This effect translates into reductions in hind limb functional impairment. We show that UnAG rescues sirtuin 1 (SIRT1) activity and superoxide dismutase-2 (SOD-2) expression in ECs. This leads to SIRT1-mediated p53 and histone 3 lysate 56 deacetylation and results in reduced EC senescence in vivo. We demonstrate, using small interfering RNA technology, that SIRT1 is also crucial for SOD-2 expression. UnAG also renews micro-RNA (miR)-126 expression, resulting in the posttranscriptional regulation of vascular cell adhesion molecule 1 expression and a reduced number of infiltrating inflammatory cells in vivo. Loss-of-function experiments that target miR-126 demonstrate that miR-126 also controls SIRT1 and SOD-2 expression, thus confirming its role in driving UnAG-mediated EC protection against ROS imbalance. These results indicate that UnAG protects vessels from ROS imbalance in ob/ob mice by rescuing miR-126 expression, thus emphasizing its potential clinical impact in avoiding limb loss in PAD.


Assuntos
Células Endoteliais/efeitos dos fármacos , Grelina/farmacologia , Intolerância à Glucose/metabolismo , MicroRNAs/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Doença Arterial Periférica/metabolismo , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/metabolismo , Masculino , Camundongos , MicroRNAs/genética , Sirtuína 1/metabolismo , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA