Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 13(1): 12240, 2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37507448

RESUMO

Rejuvenation of an old organism was achieved in heterochronic parabiosis experiments, implicating different soluble factors in this effect. Extracellular vesicles (EVs) are the secretory effectors of many cells, including cardiosphere-derived cells (CDCs) with demonstrated anti-senescent effect. 1. To determine the role of EVs (versus other blood fractions) on the rejuvenating effect of the young blood. 2. To evaluate the anti-aging properties of therapeutically administered EVs secreted by young-CDCs in an old organism. Neonatal blood fractioned in 4 components (whole blood, serum, EV-depleted serum and purified EVs) was used to treat old human cardiac stromal cells (CSPCs). CDCs were generated from neonatal rat hearts and the secreted CDC-EVs were purified. CDC-EVs were then tested in naturally-aged rats, using monthly injections over 4-months period. For validation in human samples, pediatric CDC-EVs were tested in aged human CSPCs and progeric fibroblasts. While the purified EVs reproduced the rejuvenating effects of the whole blood, CSPCs treated with EV-depleted serum exhibited the highest degree of senescence. Treatment with young CDC-EVs induce structural and functional improvements in the heart, lungs, skeletal muscle, and kidneys of old rats, while favorably modulating glucose metabolism and anti-senescence pathways. Lifespan was prolonged. EVs secreted by young CDCs exert broad-ranging anti-aging effects in aged rodents and in cellular models of human senescence. Our work not only identifies CDC-EVs as possible therapeutic candidates for a wide range of age-related pathologies, but also raises the question of whether EVs function as endogenous modulators of senescence.


Assuntos
Vesículas Extracelulares , Humanos , Ratos , Animais , Criança , Idoso , Vesículas Extracelulares/metabolismo , Envelhecimento , Coração , Fibroblastos , Pulmão , Senescência Celular/fisiologia
3.
Sci Adv ; 6(38)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32938681

RESUMO

Extracellular vesicles (EVs) including plasma membrane-derived microvesicles and endosomal-derived exosomes aggregate by unknown mechanisms, forming microcalcifications that promote cardiovascular disease, the leading cause of death worldwide. Here, we show a framework for assessing cell-independent EV mechanisms in disease by suggesting that annexin A1 (ANXA1)-dependent tethering induces EV aggregation and microcalcification. We present single-EV microarray, a method to distinguish microvesicles from exosomes and assess heterogeneity at a single-EV level. Single-EV microarray and proteomics revealed increased ANXA1 primarily on aggregating and calcifying microvesicles. ANXA1 vesicle aggregation was suppressed by calcium chelation, altering pH, or ANXA1 neutralizing antibody. ANXA1 knockdown attenuated EV aggregation and microcalcification formation in human cardiovascular cells and acellular three-dimensional collagen hydrogels. Our findings explain why microcalcifications are more prone to form in vulnerable regions of plaque, regulating critical cardiovascular pathology, and likely extend to other EV-associated diseases, including autoimmune and neurodegenerative diseases and cancer.

4.
Arterioscler Thromb Vasc Biol ; 39(10): 2082-2096, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31434491

RESUMO

OBJECTIVE: Extracellular vesicles secreted by cardiosphere-derived cells (CDCev) polarize macrophages toward a distinctive phenotype with enhanced phagocytic capacity (MCDCev). These changes underlie cardioprotection by CDCev and by the parent CDCs, notably attenuating the no-reflow phenomenon following myocardial infarction, but the mechanisms are unclear. Here, we tested the hypothesis that MCDCev are especially effective at scavenging debris from dying cells (ie, efferocytosis) to attenuate irreversible damage post-myocardial infarction. Approach and Results: In vitro efferocytosis assays with bone marrow-derived macrophages, and in vivo transgenic rodent models of myocardial infarction, demonstrate enhanced apoptotic cell clearance with MCDCev. CDCev exposure induces sustained MerTK expression in MCDCev through extracellular vesicle transfer of microRNA-26a (via suppression of Adam17); the cardioprotective response is lost in animals deficient in MerTK. Single-cell RNA-sequencing revealed phagocytic pathway activation in MCDCev, with increased expression of complement factor C1qa, a phagocytosis facilitator. CONCLUSIONS: Together, these data demonstrate that extracellular vesicle modulation of MerTK and C1qa expression leads to enhanced macrophage efferocytosis and cardioprotection.


Assuntos
Proteína ADAM17/genética , Regulação da Expressão Gênica , Glicoproteínas de Membrana/genética , Infarto do Miocárdio/patologia , Fagocitose/genética , Receptores de Complemento/genética , c-Mer Tirosina Quinase/genética , Análise de Variância , Animais , Apoptose/genética , Biópsia por Agulha , Células Cultivadas , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Proteína Tirosina Quinases/metabolismo , Análise de Sequência de RNA
5.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282820

RESUMO

Sudden death is the most common mode of exodus in patients with heart failure and preserved ejection fraction (HFpEF). Cardiosphere-derived cells (CDCs) reduce inflammation and fibrosis in a rat model of HFpEF, improving diastolic function and prolonging survival. We tested the hypothesis that CDCs decrease ventricular arrhythmias (VAs) and thereby possibly contribute to prolonged survival. Dahl salt-sensitive rats were fed a high-salt diet to induce HFpEF. Allogeneic rat CDCs (or phosphate-buffered saline as placebo) were injected in rats with echo-verified HFpEF. CDC-injected HFpEF rats were less prone to VA induction by programmed electrical stimulation. Action potential duration (APD) was shortened, and APD homogeneity was increased by CDC injection. Transient outward potassium current density was upregulated in cardiomyocytes from CDC rats relative to placebo, as were the underlying transcript (Kcnd3) and protein (Kv4.3) levels. Fibrosis was attenuated in CDC-treated hearts, and survival was increased. Sudden death risk also trended down, albeit nonsignificantly. CDC therapy decreased VA in HFpEF rats by shortening APD, improving APD homogeneity, and decreasing fibrosis. Unlike other stem/progenitor cells, which often exacerbate arrhythmias, CDCs reverse electrical remodeling and suppress arrhythmogenesis in HFpEF.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/prevenção & controle , Morte Súbita Cardíaca/prevenção & controle , Insuficiência Cardíaca/mortalidade , Mioblastos Cardíacos/transplante , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/mortalidade , Morte Súbita Cardíaca/etiologia , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Insuficiência Cardíaca/etiologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos Dahl , Canais de Potássio Shal/metabolismo , Sódio na Dieta/efeitos adversos , Volume Sistólico , Transplante Homólogo , Remodelação Ventricular
6.
J Nanobiotechnology ; 16(1): 61, 2018 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-30165851

RESUMO

BACKGROUND: Extracellular vesicles (EVs) and exosomes are nano-sized, membrane-bound vesicles shed by most eukaryotic cells studied to date. EVs play key signaling roles in cellular development, cancer metastasis, immune modulation and tissue regeneration. Attempts to modify exosomes to increase their targeting efficiency to specific tissue types are still in their infancy. Here we describe an EV membrane anchoring platform termed "cloaking" to directly embed tissue-specific antibodies or homing peptides on EV membrane surfaces ex vivo for enhanced vesicle uptake in cells of interest. The cloaking system consists of three components: DMPE phospholipid membrane anchor, polyethylene glycol spacer and a conjugated streptavidin platform molecule, to which any biotinylated molecule can be coupled for EV decoration. RESULTS: We demonstrate the utility of membrane surface engineering and biodistribution tracking with this technology along with targeting EVs for enhanced uptake in cardiac fibroblasts, myoblasts and ischemic myocardium using combinations of fluorescent tags, tissue-targeting antibodies and homing peptide surface cloaks. We compare cloaking to a complementary approach, surface display, in which parental cells are engineered to secrete EVs with fusion surface targeting proteins. CONCLUSIONS: EV targeting can be enhanced both by cloaking and by surface display; the former entails chemical modification of preformed EVs, while the latter requires genetic modification of the parent cells. Reduction to practice of the cloaking approach, using several different EV surface modifications to target distinct cells and tissues, supports the notion of cloaking as a platform technology.


Assuntos
Exossomos/química , Vesículas Extracelulares/metabolismo , Corantes Fluorescentes/química , Terapia de Alvo Molecular/métodos , Nanopartículas/química , Animais , Anticorpos/química , Anticorpos/metabolismo , Transporte Biológico , Linhagem Celular , Feminino , Humanos , Imagem Óptica , Tamanho da Partícula , Peptídeos/química , Peptídeos/metabolismo , Fosfolipídeos/química , Polietilenoglicóis/química , Pontos Quânticos/química , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Propriedades de Superfície , Distribuição Tecidual/efeitos dos fármacos
7.
J Extracell Vesicles ; 7(1): 1456888, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29696078

RESUMO

Newts can regenerate amputated limbs and cardiac tissue, unlike mammals which lack broad regenerative capacity. Several signaling pathways involved in cell proliferation, differentiation and survival during newt tissue regeneration have been elucidated, however the factors that coordinate signaling between cells, as well as the conservation of these factors in other animals, are not well defined. Here we report that media conditioned by newt limb explant cells (A1 cells) protect mammalian cardiomyocytes from oxidative stress-induced apoptosis. The cytoprotective effect of A1-conditioned media was negated by exposing A1 cells to GW4869, which suppresses the generation of extracellular vesicles (EVs). A1-EVs are similar in diameter (~100-150 nm), structure, and share several membrane surface and cargo proteins with mammalian exosomes. However, isolated A1-EVs contain significantly higher levels of both RNA and protein per particle than mammalian EVs. Additionally, numerous cargo RNAs and proteins are unique to A1-EVs. Of particular note, A1-EVs contain numerous mRNAs encoding nuclear receptors, membrane ligands, as well as transcription factors. Mammalian cardiomyocytes treated with A1-EVs showed increased expression of genes in the PI3K/AKT pathway, a pivotal player in survival signaling. We conclude that newt cells secrete EVs with diverse, distinctive RNA and protein contents. Despite ~300 million years of evolutionary divergence between newts and mammals, newt EVs confer cytoprotective effects on mammalian cardiomyocytes.

8.
J Mol Cell Cardiol ; 119: 125-137, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29698635

RESUMO

Though experimental, stem cell transplantation has the potential to improve the condition of the heart after myocardial infarction. It does so by reducing infarct size and inducing repair of heart muscle and its blood supply. Mesenchymal stem cells (MSC) have been found to be effective in pre-clinical animal models and clinical trials, but the mechanisms by which they induce cardioprotection and repair are still not fully understood. Small extracellular vesicles known as exosomes are now recognized to be key mediators of beneficial MSC paracrine effects, and the concept that they transfer miRNA to change gene expression in recipient cells is of current therapeutic interest. We present complete deep miRNA sequencing of MSC exosome cargo, and found that of several cardioprotective miRNAs, miR-21a-5p was the most abundant. Because miR-21a-5p is a well-known cardioprotective miRNA, we investigated the hypothesis that MSC exosomes can cardioprotect the heart by increasing the level of miR-21a-5p in recipient cardiac cells, thereby downregulating expression of the pro-apoptotic gene products PDCD4, PTEN, Peli1 and FasL in the myocardium. Using miR-21 mimic transfection and treatment with wild type and miR-21a knockout MSC exosomes, we confirmed that exosomal miR-21a-5p is transferred into myocardium and is a major cardioprotective paracrine factor produced by MSCs acting via synergistic activity on multiple pathways. The data supports that residual cardioprotective effect may be due to other ncRNA or protein cargo. In silico analyses support that MSC exosomes may also contribute to angiogenesis, cell proliferation and other aspects of cardiac repair.


Assuntos
Exossomos/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Animais , Linhagem Celular , Proliferação de Células/genética , Exossomos/metabolismo , Técnicas de Inativação de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Ratos
9.
CNS Neurosci Ther ; 20(12): 1056-60, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25367718

RESUMO

AIMS: There is still no effective way to save a surviving healthy mind when there is critical organ failure in the body. The next frontier in CTA is allo-head and body reconstruction (AHBR), and just as animal models were key in the development of CTA, they will be crucial in establishing the procedures of AHBR for clinical translation. METHODS AND RESULTS: Our approach, pioneered in mice, involves retaining the donor brain stem and transplanting the recipient head. Our preliminary data in mice support that this allows for retention of breathing and circulatory function. Critical aspects of the current protocol include avoiding cerebral ischemia through cross-circulation (donor to recipient) and retaining the donor brain stem. Successful clinical translation of AHBR will become a milestone of medical history and potentially could save millions of people. CONCLUSIONS: This experimental study has confirmed a method to avoid cerebral ischemia during the surgery and solved an important part of the problem of how to accomplish long-term survival after transplantation and preservation of the donor brain stem.


Assuntos
Cabeça/cirurgia , Procedimentos de Cirurgia Plástica/métodos , Transplante Homólogo/métodos , Animais , Eletrocardiografia , Eletroencefalografia , Camundongos , Camundongos Endogâmicos , Modelos Animais , Sinais Vitais/fisiologia
10.
Am J Rhinol Allergy ; 28(3): 215-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24980233

RESUMO

BACKGROUND: Capsaicin, a prototypic transient receptor potential vanilloid 1 (TRPV1) agonist, has been shown to be more clinically effective in the treatment of nonallergic rhinitis (NAR) compared with other rhinitis subtypes. Azelastine has also been found to be clinically effective in the treatment of NAR but its mechanism(s) of action is still poorly elucidated. This study was designed to determine, using in vitro cell lines, whether topical therapies including azelastine have activity on TRPV1 ion channels similar to capsaicin. METHODS: The effects of capsaicin (1 µM), azelastine (30 µM), bepotastine (10 µM), olopatadine (10 µM), and fluticasone (200 µM) on TRPV1 channels using mice neuronal cells (Cath.a), as surrogates for submucosal sensory neurons, and human nasal epithelial cells (hNEC) were determined and compared. For azelastine, bepotastine, and capsaicin, which elicited an agonist effect on TRPV1, live cell [Ca(2+)] signaling in Cath.a cells and hNECs expressing TRPV1 were performed in the absence and presence of capsazepine at 10 µM (a TRPV1 antagonist) or using wild-type mouse embryonic fibroblasts (wtMEFs) that express TRPV1 ion channels and TRPV1 homozygous null mutant (TRPV1-/-) knockout MEF cells as controls to establish TRPV1 channel selectivity. As azelastine has previously been found clinically effective in NAR, additional experiments were performed to determine its ability to desensitize TRPV1 ion channels and its effect on regulating intracellular calcium homeostasis. RESULTS: Cath.a cells treated with azelastine, bepotastine, or capsaicin showed a significant increase in TRPV1-dependant (Ca(2+)) specific cytosolic fluorescence. Continuous treatment with azelastine or capsaicin resulted in desensitization of TRPV1 channels. In hNECs, azelastine stimulation resulted in Ca(2+) shifts from the cytosol to mitochondria and overexpression of hematopoietic cell-specific Lyn substrate 1-associated protein X1, which may thus be effective in cytosolic Ca(2+) homeostasis. CONCLUSION: Azelastine, similar to capsaicin, exhibits direct activity on TRPV1 ion channels that may represent a novel mechanistic pathway explaining its clinical efficacy in NAR.


Assuntos
Capsaicina/farmacologia , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ftalazinas/farmacologia , Rinite/tratamento farmacológico , Canais de Cátion TRPV/agonistas , Androstadienos/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Capsaicina/análogos & derivados , Capsaicina/uso terapêutico , Linhagem Celular , Dibenzoxepinas/farmacologia , Células Epiteliais/fisiologia , Fibroblastos/fisiologia , Fluticasona , Humanos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Mucosa Nasal/citologia , Neurônios/fisiologia , Cloridrato de Olopatadina , Ftalazinas/uso terapêutico , Piperidinas/farmacologia , Piridinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
12.
PLoS One ; 8(8): e70023, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23990893

RESUMO

OBJECTIVE: The purpose of this study was to assess the effect of collagen composition on engraftment of progenitor cells within infarcted myocardium. BACKGROUND: We previously reported that intramyocardial penetration of stem/progenitor cells in epicardial patches was enhanced when collagen was reduced in hearts overexpressing adenylyl cyclase-6 (AC6). In this study we hypothesized an alternative strategy wherein overexpression of microRNA-29b (miR-29b), inhibiting mRNAs that encode cardiac fibroblast proteins involved in fibrosis, would similarly facilitate progenitor cell migration into infarcted rat myocardium. METHODS: In vitro: A tri-cell patch (Tri-P) consisting of cardiac sodium-calcium exchanger-1 (NCX1) positive iPSC (iPSC(NCX1+)), endothelial cells (EC), and mouse embryonic fibroblasts (MEF) was created, co-cultured, and seeded on isolated peritoneum. The expression of fibrosis-related genes was analyzed in cardiac fibroblasts (CFb) by qPCR and Western blot. In vivo: Nude rat hearts were administered mimic miRNA-29b (miR-29b), miRNA-29b inhibitor (Anti-29b), or negative mimic (Ctrl) before creation of an ischemically induced regional myocardial infarction (MI). The Tri-P was placed over the infarcted region 7 days later. Angiomyogenesis was analyzed by micro-CT imaging and immunofluorescent staining. Echocardiography was performed weekly. RESULTS: The number of green fluorescent protein positive (GFP(+)) cells, capillary density, and heart function were significantly increased in hearts overexpressing miR-29b as compared with Ctrl and Anti-29b groups. Conversely, down-regulation of miR-29b with anti-29b in vitro and in vivo induced interstitial fibrosis and cardiac remodeling. CONCLUSION: Overexpression of miR-29b significantly reduced scar formation after MI and facilitated iPSC(NCX1+) penetration from the cell patch into the infarcted area, resulting in restoration of heart function after MI.


Assuntos
Colágeno/química , Células-Tronco Pluripotentes Induzidas/citologia , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Movimento Celular , Técnicas de Cocultura , Ecocardiografia , Fibroblastos/citologia , Fibrose/patologia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Lentivirus/metabolismo , Camundongos , MicroRNAs/metabolismo , Infarto do Miocárdio/patologia , Neovascularização Patológica , Ratos , Ratos Nus , Transfecção , Remodelação Ventricular/fisiologia , Microtomografia por Raio-X
13.
J Cardiovasc Pharmacol Ther ; 18(3): 280-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23241275

RESUMO

The current therapeutic options for acute decompensated heart failure are limited to afterload reducers and positive inotropes. The latter increases myocardial contractility through changes in myocyte calcium (Ca²âº) handling (mostly through stimulation of the ß-adrenergic pathways [ß-ADR]) and is associated with paradoxical effects of arrhythmias, cell death, and subsequently increased mortality. We have previously demonstrated that probenecid can increase cytosolic Ca²âº levels in the cardiomyocyte resulting in an improved inotropic response in vitro and in vivo without activating the ß-ADR system. We hypothesize that, in contrast to other commonly used inotropes, probenecid functions through a system separate from that of ß-ADR and hence will increase contractility and improve function without damaging the heart. Furthermore, our goal was to evaluate the effect of probenecid on cell death in vitro and its use in vivo as a positive inotrope in a mouse model of ischemic cardiomyopathy. Herein, we demonstrate that probenecid induced an influx of Ca²âº similar to isoproterenol, but does not induce cell death in vitro. Through a series of in vivo experiments we also demonstrate that probenecid can be used at various time points and with various methods of administration in vivo in mice with myocardial ischemia, resulting in improved contractility and no significant difference in infarct size. In conclusion, we provide novel data that probenecid, through its activity on cellular Ca²âº levels, induces an inotropic effect without causing or exacerbating injury. This discovery may be translatable if this mechanism is preserved in man.


Assuntos
Cardiotônicos/uso terapêutico , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Moduladores de Transporte de Membrana/uso terapêutico , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Probenecid/uso terapêutico , Administração Oral , Animais , Sinalização do Cálcio/efeitos dos fármacos , Cardiotônicos/administração & dosagem , Cardiotônicos/efeitos adversos , Cardiotônicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Coração/fisiopatologia , Injeções Intraperitoneais , Cinética , Masculino , Moduladores de Transporte de Membrana/administração & dosagem , Moduladores de Transporte de Membrana/efeitos adversos , Moduladores de Transporte de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Probenecid/administração & dosagem , Probenecid/efeitos adversos , Probenecid/farmacologia , Distribuição Aleatória
14.
Prog Mol Biol Transl Sci ; 111: 265-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22917235

RESUMO

Progenitor/stem cells are a viable option to replace myocytes lost subsequent to myocardial infarction (MI). The stromal cell-derived factor-1α/CXC-chemokine receptor type 4 (SDF-1α/CXCR4) axis plays an important role in numerous biological processes including hematopoiesis, cardiogenesis, vasculogenesis, and neuronal development, as well as endothelial progenitor cell trafficking. The secretion of chemoattractants such as SDF-1α at the site of injury creates an environment facilitating the homing of circulating CXCR4 positive and other stem cells (such as mast/stem cell growth factor receptor kit-positive (c-kit(+)) and c-kit(+)/GATA binding protein 4 positive (GATA4(+)) cells) for organ regeneration and tissue repair. SDF-1α is also secreted by hematopoietic progenitor/stem cells and is involved in the autocrine/paracrine regulation of their development and survival. Hypoxic preconditioning activates SDF-1α/CXCR4 signaling and upregulates several vascular/angiogenic factors that cause mobilization of progenitor cells. The SDF-1α/CXCR4 signaling pathway can thus be effectively exploited for cell-based therapy.


Assuntos
Quimiocina CXCL12/metabolismo , Engenharia Genética/métodos , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Receptores CXCR4/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Humanos
15.
Mol Endocrinol ; 25(5): 754-66, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21372145

RESUMO

Our laboratory has previously cloned and purified an ovarian protein found to be a novel 17ß-hydroxysteroid dehydrogenase type 7 enzyme (HSD17B7) (formerly prolactin receptor-associated protein) that converts the weak estrogen, estrone, to the highly potent estradiol. The regulation of this enzyme has not yet been explored. In this report, we show high expression of HSD17B7 in human ductal carcinoma and breast cancer cell lines and present evidence for a strong up-regulation of this enzyme by estradiol at the level of mRNA, protein expression, and promoter activity in MCF-7 cells. The effect of estradiol is mediated by estrogen receptor (ER)α, whereas ERß prevents this stimulation. ER antagonists, ICI 182,780 and 4-hydroxytamoxifen, prevent estradiol-induced stimulation of the endogenously expressed HSD17B7, suggesting that these inhibitors not only block estradiol action but also its production. We have identified a -185-bp region of the hsd17b7 promoter that is highly conserved among rat, mouse, and human and confers regulation by estradiol in MCF-7 cells. This region is devoid of a classical estradiol-response element but contains a nuclear factor 1 (NF1) site that is essential for estradiol action. We found that estradiol stimulates the recruitment and DNA binding of NF1 to this region of the hsd17b7 promoter. Furthermore, knockdown of NF1 family members, NF1B, NF1A, and NF1X, completely prevents induction of this gene by estradiol. In summary, our findings demonstrate that estradiol stimulates HSD17B7 transcriptional activity in breast cancer cells through a novel mechanism requiring NF1 and strongly suggest a positive feedback mechanism to increase local estradiol synthesis causing growth of estrogen-dependent breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proteínas de Ciclo Celular/genética , Estradiol/biossíntese , Proteínas S100/genética , Animais , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Retroalimentação Fisiológica , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Soros Imunes , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Camundongos , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Coelhos , Elementos de Resposta , Proteína A6 Ligante de Cálcio S100 , Proteínas S100/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA