Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Oncoimmunology ; 3(1): e27259, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24711955

RESUMO

We have recently demonstrated that a DNA vaccine targeting membrane-bound KIT ligand (KITL) inhibits tumor growth by interfering with vessel stabilization/permeability and by disrupting the recruitment of inflammatory cells and regulatory T cells, the latter being an essential mechanism by which tumors resist available treatments. Combining KITL-targeting vaccines with conventional chemotherapy might avert drug resistance and improve the efficacy of standard-of-care therapeutic interventions.

10.
Cell Cycle ; 13(11): 1811-26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24736554

RESUMO

α6ß4 integrin is an adhesion molecule for laminin receptors involved in tumor progression. We present a link between ß4 integrin expression and miR-221/222 in the most prevalent human mammary tumor: luminal invasive carcinomas (Lum-ICs). Using human primary tumors that display different ß4 integrin expression and grade, we show that miR-221/222 expression inversely correlates with tumor proliferating index, Ki67. Interestingly, most high-grade tumors express ß4 integrin and low miR-221/222 levels. We ectopically transfected miR-221/222 into a human-derived mammary tumor cell line that recapitulates the luminal subtype to investigate whether miR-221/222 regulates ß4 expression. We demonstrate that miR-221/222 overexpression results in ß4 expression downregulation, breast cancer cell proliferation, and invasion inhibition. The role of miR-221/222 in driving ß4 integrin expression is also confirmed via mutating the miR-221/222 seed sequence for ß4 integrin 3'UTR. Furthermore, we show that these 2 miRNAs are also key breast cancer cell proliferation and invasion regulators, via the post-transcriptional regulation of signal transducer and activator of transcription 5A (STAT5A) and of a disintegrin and metalloprotease-17 (ADAM-17). We further confirm these data by silencing ADAM-17, using a dominant-negative or an activated STAT5A form. miR-221/222-driven ß4 integrin, STAT5A, and ADAM-17 did not occur in MCF-10A cells, denoted "normal" breast epithelial cells, indicating that the mechanism is cancer cell-specific.   These results provide the first evidence of a post-transcriptional mechanism that regulates ß4 integrin, STAT5A, and ADAM-17 expression, thus controlling breast cancer cell proliferation and invasion. Pre-miR-221/222 use in the aggressive luminal subtype may be a powerful therapeutic anti-cancer strategy.


Assuntos
Neoplasias da Mama/fisiopatologia , Carcinoma Ductal de Mama/fisiopatologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Integrina beta4/metabolismo , MicroRNAs/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Análise de Variância , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Primers do DNA/genética , Progressão da Doença , Feminino , Inativação Gênica , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Luciferases , Invasividade Neoplásica/genética , RNA Interferente Pequeno/genética , Fator de Transcrição STAT5/metabolismo , Proteínas Supressoras de Tumor/metabolismo
11.
PLoS One ; 9(1): e84859, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24400121

RESUMO

MicroRNAs are single-stranded non-coding RNAs that simultaneously down-modulate the expression of multiple genes post-transcriptionally by binding to the 3'UTRs of target mRNAs. Here we used computational methods to predict microRNAs relevant in breast cancer progression. Specifically, we applied different microRNA target prediction algorithms to various groups of differentially expressed protein-coding genes obtained from four breast cancer datasets. Six potential candidates were identified, among them miR-223, previously described to be highly expressed in the tumor microenvironment and known to be actively transferred into breast cancer cells. To investigate the function of miR-223 in tumorigenesis and to define its molecular mechanism, we overexpressed miR-223 in breast cancer cells in a transient or stable manner. Alternatively we overexpressed miR-223 in mouse embryonic fibroblasts or HEK293 cells and used their conditioned medium to treat tumor cells. With both approaches, we obtained elevated levels of miR-223 in tumor cells and observed decreased migration, increased cell death in anoikis conditions and augmented sensitivity to chemotherapy but no effect on adhesion and proliferation. The analysis of miR-223 predicted targets revealed enrichment in cell death and survival-related genes and in pathways frequently altered in breast cancer. Among these genes, we showed that protein levels for STAT5A, ITGA3 and NRAS were modulated by miR-223. In addition, we proved that STAT5A is a direct miR-223 target and highlighted a possible correlation between miR-223 and STAT5A in migration and chemotherapy response. Our investigation revealed that a computational analysis of cancer gene expression datasets can be a relevant tool to identify microRNAs involved in cancer progression and that miR-223 has a prominent role in breast malignancy that could potentially be exploited therapeutically.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , MicroRNAs/genética , Regiões 3' não Traduzidas , Anoikis/genética , Antineoplásicos/farmacologia , Pareamento de Bases , Sequência de Bases , Neoplasias da Mama/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Biologia Computacional , Bases de Dados Factuais , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/química , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Eur J Cancer ; 50(1): 234-46, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24144734

RESUMO

A functional c-Kit/Kit ligand (KitL) signalling network is required for tumour angiogenesis and growth, and therefore the c-Kit/KitL system might well be a suitable target for the cancer immunotherapy approach. We herein describe a strategy that targets membrane-bound KitL (mbKitL) via DNA vaccination. The vaccination procedure generated antibodies which are able to detect mbKitL on human tumour endothelial cells (TECs) and on the breast cancer cell line: TSA. DNA vaccination, interferes with tumour vessel formation and transplanted tumour growth in vivo. Histological analysis demonstrates that, while tumour cell proliferation and vessel stabilisation are impaired, vessel permeability is increased in mice that produce mbKitL-targeting antibodies. We also demonstrate that vessel stabilisation and tumour growth require Akt activation in endothelial cells but not in pericytes. Moreover, we found that regulatory T cells (Treg) and tumour infiltrating inflammatory cells, involved in tumour growth and angiogenesis, were reduced in number in the tumour microenvironment of mice that generate anti-mbKitL antibodies. These data provide evidence that mbKitL targeted vaccination is an effective means of inhibiting tumour angiogenesis and growth.


Assuntos
Neoplasias Hepáticas Experimentais/terapia , Proteínas Proto-Oncogênicas c-kit/imunologia , Neoplasias Esplênicas/terapia , Vacinas de DNA/administração & dosagem , Animais , Células CHO , Processos de Crescimento Celular/imunologia , Cricetulus , Humanos , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Neoplasias Hepáticas Experimentais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias Esplênicas/irrigação sanguínea , Neoplasias Esplênicas/imunologia , Vacinas de DNA/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
J Am Heart Assoc ; 2(6): e000376, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24308935

RESUMO

BACKGROUND: Surgical treatment of peripheral artery disease, even if successful, does not prevent reoccurrence. Under these conditions, increased oxidative stress is a crucial determinant of tissue damage. Given its reported antioxidant effects, we investigated the potential of unacylated-ghrelin (UnAG) to reduce ischemia-induced tissue damage in a mouse model of peripheral artery disease. METHODS AND RESULTS: We show that UnAG but not acylated ghrelin (AG) induces skeletal muscle regeneration in response to ischemia via canonical p38/mitogen-actived protein kinase signaling UnAG protected against reactive oxygen species-induced cell injuries by inducing the expression of superoxide dismutase-2 (SOD-2) in satellite cells. This led to a reduced number of infiltrating CD68(+) cells and was followed by induction of the myogenic process and a reduction in functional impairment. Moreover, we found that miR-221/222, previously linked to muscle regeneration processes, was up-regulated and negatively correlated with p57(Kip2) expression in UnAG-treated mice. UnAG, unlike AG, promoted cell-cycle entry in satellite cells of mice lacking the genes for ghrelin and its receptor (GHSR1a). UnAG-induced p38/mitogen-actived protein kinase phosphorylation, leading to activation of the myogenic process, was prevented in SOD-2-depleted SCs. By siRNA technology, we also demonstrated that SOD-2 is the antioxidant enzyme involved in the control of miR-221/222-driven posttranscriptional p57(Kip2) regulation. Loss-of-function experiments targeting miR-221/222 and local pre-miR-221/222 injection in vivo confirmed a role for miR-221/222 in driving skeletal muscle regeneration after ischemia. CONCLUSIONS: These results indicate that UnAG-induced skeletal muscle regeneration after ischemia depends on SOD-2-induced miR-221/222 expression and highlight its clinical potential for the treatment of reactive oxygen species-mediated skeletal muscle damage.


Assuntos
Antioxidantes/farmacologia , Grelina/farmacologia , Isquemia/tratamento farmacológico , MicroRNAs/metabolismo , Músculo Esquelético/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Modelos Animais de Doenças , Grelina/análogos & derivados , Grelina/deficiência , Grelina/genética , Membro Posterior , Isquemia/enzimologia , Isquemia/genética , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Proteína MyoD/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fator de Transcrição PAX7/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/enzimologia , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/genética , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Diabetes ; 62(4): 1245-57, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23223023

RESUMO

Metabolic profiling of plasma nonesterified fatty acids discovered that palmitic acid (PA), a natural peroxisome proliferator-activated receptor γ (PPARγ) ligand, is a reliable type 2 diabetes biomarker. We investigated whether and how PA diabetic (d-PA) concentrations affected endothelial progenitor cell (EPC) and bone marrow-derived hematopoietic cell (BM-HC) biology. PA physiologic (n-PA) and d-PA concentrations were used. Proliferating cell nuclear antigen content and signal transducer and activator of transcription 5 (STAT5), PPARγ, cyclin D1, and p21(Waf) expression were evaluated. Small interfering RNA technology, gene reporter luciferase assay, electrophoretic mobility shift assay, chromatin immunoprecipitation assay, and coimmunoprecipitation were exploited. In vivo studies and migration assays were also performed. d-PA, unlike n-PA or physiological and diabetic oleic and stearic acid concentrations, impaired EPC migration and EPC/BM-HC proliferation through a PPARγ-mediated STAT5 transcription inhibition. This event did not prevent the formation of a STAT5/PPARγ transcriptional complex but was crucial for gene targeting, as p21(Waf) gene promoter, unlike cyclin D1, was the STAT5/PPARγ transcriptional target. Similar molecular events could be detected in EPCs isolated from type 2 diabetic patients. By expressing a constitutively activated STAT5 form, we demonstrated that STAT5 content is crucial for gene targeting and EPC fate. Finally, we also provide in vivo data that d-PA-mediated EPC dysfunction could be rescued by PPARγ blockade. These data provide first insights on how mechanistically d-PA drives EPC/BM-HC dysfunction in diabetes.


Assuntos
Células da Medula Óssea/classificação , Células Endoteliais/classificação , PPAR gama/metabolismo , Ácido Palmítico/metabolismo , Fator de Transcrição STAT5/metabolismo , Células-Tronco/classificação , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Movimento Celular , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hematopoese/fisiologia , Humanos , PPAR gama/genética , Ácido Palmítico/sangue , Ácido Palmítico/toxicidade , Fator de Transcrição STAT5/genética , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
15.
Stem Cells Dev ; 2012 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22272916

RESUMO

NADPH-oxidase (NOX)-dependent reactive oxygen species (ROS) production is involved in self-renewal of stem and progenitor cells. Herein, we investigated whether high glucose (25 mM/L) (HG)-dependent NOX-mediated ROS generation is involved in self-renewal of visceral adipose tissue-derived stem cells (ASCs) as well. To this end ASCs cultured in HG or normal glucose (5 mM/L) used as control, were evaluated for their stem cell identity. We demonstrated that freshly isolated ASCs are pluripotent as they differentiate into adipocytes in-vitro and form neovessels in-vivo. However, only HG-cultured ASCs expressed octamer-binding transcription factor 4 (Oct4) and Nanog and formed spheroids. The assembly of p47phox and p67phox subunits is crucial for NOX-enzymatic activity. By knock-down of p47phox the role of NOX-generated ROS in driving ASC de-differentiation has been provided. siRNA technology was also applied to demonstrate the role of Akt activity in mediating HG-induced Oct-4 and Nanog expression as well as spheroid formation. Additionally, by knock-down of Oct4 we provided further evidence that Oct4 is essential for HG-mediated stem cell identity. Soluble factors released by ASCs are key elements in their mechanism of action. We found that NOX and Akt activity are required for cytokine production by "spheroids". Finally, as HG-cultured ASCs, diabetic patient-derived ASCs expressed higher levels of Oct-4 and Nanog than ASCs derived from healthy subjects and engaged ROS and Akt activity to turn on their secretion program. Thereby, our data indicate that HG via NOX-dependent Akt activity induces ASC de-differentiation, and suggest that HG pre-conditioning might be exploited for ASC ex-vivo expansion.

16.
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
17.
Diabetes ; 59(4): 1016-25, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20068135

RESUMO

OBJECTIVE: Acylated ghrelin (AG) is a diabetogenic and orexigenic gastric polypeptide. These properties are not shared by the most abundant circulating form, which is unacylated (UAG). An altered UAG/AG profile together with an impairment of circulating endothelial progenitor cell (EPC) bioavailability were found in diabetes. Based on previous evidence for the beneficial cardiovascular effects of AG and UAG, we investigated their potential to revert diabetes-associated defects. RESEARCH DESIGN AND METHODS: Healthy human subjects, individuals with type 2 diabetes, and ob/ob mice were AG or UAG infused. EPC mobilization in patients and mice was evaluated, and the underlying molecular mechanisms were investigated in bone marrow stromal cells. Recovered EPCs were also evaluated for the activity of senescence regulatory pathways and for NADPH oxidase activation by knocking down p47(phox) and Rac1. Finally, UAG modulation of human EPC vasculogenic potential was investigated in an in vivo mouse model. RESULTS: Neither AG nor UAG had any effect in healthy subjects. However, systemic administration of UAG, but not AG, prevented diabetes-induced EPC damage by modulating the NADPH oxidase regulatory protein Rac1 and improved the vasculogenic potential both in individuals with type 2 diabetes and in ob/ob mice. In addition, unlike AG, UAG facilitated the recovery of bone marrow EPC mobilization. Crucial to EPC mobilization by UAG was the rescue of endothelial NO synthase (eNOS) phosphorylation by Akt, as UAG treatment was ineffective in eNOS knockout mice. Consistently, EPCs expressed specific UAG-binding sites, not recognized by AG. CONCLUSIONS: These data provide the rationale for clinical applications of UAG in pathologic settings where AG fails.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Endotélio Vascular/fisiopatologia , Grelina/uso terapêutico , Células-Tronco/fisiologia , Acilação , Idoso , Animais , Doadores de Sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Feminino , Humanos , Lipídeos/sangue , Masculino , Camundongos , Camundongos Obesos , Pessoa de Meia-Idade , Estresse Oxidativo/efeitos dos fármacos , Valores de Referência , Células-Tronco/efeitos dos fármacos
18.
Acta Diabetol ; 47(2): 105-11, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19404565

RESUMO

Pericyte survival in diabetic retinopathy depends also on interactions with extracellular matrix (ECM) proteins, which are degraded by matrix metalloproteinases (MMP). Elevated glucose influences ECM turnover, through expression of MMP and their tissue inhibitors, TIMP. We reported on reduced pericyte adhesion to high glucose-conditioned ECM and correction by thiamine. We aimed at verifying the effects of thiamine and benfotiamine on MMP-2, MMP-9 and TIMP expression and activity in human vascular cells with high glucose. In HRP, MMP-2 activity, though not expression, increased with high glucose and decreased with thiamine and benfotiamine; TIMP-1 expression increased with high glucose plus thiamine and benfotiamine; MMP-9 was not expressed. In EC, MMP-9 and MMP-2 expression and activity increased with high glucose, but thiamine and benfotiamine had no effects; TIMP-1 expression was unchanged. Neither glucose nor thiamine modified TIMP-2 and TIMP-3 expression. TIMP-1 concentrations did not change in either HRP or EC. High glucose imbalances MMP/TIMP regulation, leading to increased ECM turnover. Thiamine and benfotiamine correct the increase in MMP-2 activity due to high glucose in HRP, while increasing TIMP-1.


Assuntos
Glucose/farmacologia , Metaloproteinases da Matriz/metabolismo , Tiamina/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Células Cultivadas , Primers do DNA , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/efeitos dos fármacos , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/enzimologia , RNA Mensageiro/genética , Retina/enzimologia , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidores Teciduais de Metaloproteinases/efeitos dos fármacos
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...