Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1122: 187-210, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30937870

RESUMO

Mural cells known as pericytes envelop the endothelial layer of microvessels throughout the body and have been described to have tissue-specific functions. Cardiac pericytes are abundantly found in the heart, but they are relatively understudied. Currently, their importance is emerging in cardiovascular homeostasis and dysfunction due to their pleiotropism. They are known to play key roles in vascular tone and vascular integrity as well as angiogenesis. However, their dysfunctional presence and/or absence is critical in the mechanisms that lead to cardiac pathologies such as myocardial infarction, fibrosis, and thrombosis. Moreover, they are targeted as a therapeutic potential due to their mesenchymal properties that could allow them to repair and regenerate a damaged heart. They are also sought after as a cell-based therapy based on their healing potential in preclinical studies of animal models of myocardial infarction. Therefore, recognizing the importance of cardiac pericytes and understanding their biology will lead to new therapeutic concepts.


Assuntos
Coração , Microvasos/citologia , Miocárdio/citologia , Pericitos/citologia , Animais , Humanos , Neovascularização Fisiológica , Regeneração
2.
J Biol Chem ; 292(31): 12787-12800, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28637870

RESUMO

Stress-induced p38 mitogen-activated protein kinase (MAPK) activity is implicated in pathological remodeling in the heart. For example, constitutive p38 MAPK activation in cardiomyocytes induces pathological features, including myocyte hypertrophy, apoptosis, contractile dysfunction, and fetal gene expression. However, the physiological function of cardiomyocyte p38 MAPK activity in beneficial compensatory vascular remodeling is unclear. This report investigated the functional role and the underlying mechanisms of cardiomyocyte p38 MAPK activity in cardiac remodeling induced by chronic stress. Using both in vitro and in vivo model systems, we found that p38 MAPK activity is required for hypoxia-induced pro-angiogenic activity from cardiomyocytes and that p38 MAPK activation in cardiomyocyte is sufficient to promote paracrine signaling-mediated, pro-angiogenic activity. We further demonstrate that VEGF is a paracrine factor responsible for the p38 MAPK-mediated pro-angiogenic activity from cardiomyocytes and that p38 MAPK pathway activation is sufficient for inducing VEGF secretion from cardiomyocytes in an Sp1-dependent manner. More significantly, cardiomyocyte-specific inactivation of p38α in mouse heart impaired compensatory angiogenesis after pressure overload and promoted early onset of heart failure. In summary, p38αMAPK has a critical role in the cross-talk between cardiomyocytes and vasculature by regulating stress-induced VEGF expression and secretion in cardiomyocytes. We conclude that as part of a stress-induced signaling pathway, p38 MAPK activity significantly contributes to both pathological and compensatory remodeling in the heart.


Assuntos
Endotélio Vascular/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Isquemia Miocárdica/metabolismo , Revascularização Miocárdica , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Hipóxia Celular , Células Cultivadas , Cruzamentos Genéticos , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Ativação Enzimática , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 14 Ativada por Mitógeno/química , Proteína Quinase 14 Ativada por Mitógeno/genética , Isquemia Miocárdica/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/patologia , Interferência de RNA , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Fator de Transcrição Sp1/antagonistas & inibidores , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Sus scrofa , Fator A de Crescimento do Endotélio Vascular/agonistas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
J Mol Cell Cardiol ; 51(3): 337-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21684288

RESUMO

The platelet derived growth factor receptor (PDGFR) is an important target for novel anti-cancer therapeutics, but agents targeting PDGFR have been associated with cardiotoxicity. Cardiomyocyte PDGFR-ß signaling in pressure-overloaded hearts induces compensatory angiogenesis via a paracrine-signaling cascade. Tight regulation of receptor tyrosine kinases in response to ligand stimulation is a critical part of any such cascade. The objective of the present study was to characterize the early and late regulation of PDGFR-ß following ligand stimulation and define a potential role for microRNAs (miRNAs) predicted to interact with the 3'UTR of PDGFR-ß in feedback regulation. Using two in-vitro model systems (U87 glioblastoma cells and neonatal cardiomyocytes), we observed that in response to stimulation with PDGF-BB, levels of PDGFR-ß declined beginning at one hour, persisting for 48 h. PDGFR-ß mRNA levels declined beginning at 6h after receptor activation. Early, but not late activation-induced receptor downregulation was proteasome dependent. Levels of miRNA-9 (miR-9) were significantly increased in U87 cells and cardiomyocytes beginning 6h after addition of ligand. In response to pressure overload, miR-9 levels were significantly reduced in the hearts of cardiac-specific PDGFR-ß knockout mice. Luciferase reporter assays demonstrate that miR-9 directly interacts with its predicted seed in the 3'UTR of PDGFR-ß. Increasing miR-9 levels reduces levels of PDGFR-ß, resulting in a reduction in the paracrine angiogenic capacity of cardiomyocytes, consistent with the established function of cardiomyocyte PDGFR-ß. Importantly, increase of anti-miR-9 in cardiomyocytes attenuates ligand-induced PDGFR-ß downregulation. In conclusion, we have identified miR-9 as an activation-induced regulator of PDGFR-ß expression in cardiomyocytes that is part of a negative feedback loop which serves to modulate PDGFR-ß expression upon ligand-stimulation through direct interaction with the 3'UTR of PDFGR-ß. This article is part of a Special Issue entitled 'Possible Editorial'.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Regiões 3' não Traduzidas/genética , Animais , Células COS , Linhagem Celular Transformada , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetinae , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , MicroRNAs/genética , Miócitos Cardíacos/efeitos dos fármacos , Oligorribonucleotídeos/genética , Oligorribonucleotídeos/metabolismo , Comunicação Parácrina/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Inibidores de Proteínas Quinases/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
4.
FEBS Open Bio ; 11(1): 207-225, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33135334

RESUMO

Pericytes (PCs)-mural cells that envelop endothelial cells (ECs) of microvessels-regulate tissue-specific vasculature development as well as maturation and maintenance of endothelial barrier integrity. However, little is known about their tissue-specific function in the heart. Specifically, the mechanism by which cardiac PCs constrict coronary capillaries remains undetermined. To gain insights into the function of cardiac PCs at the cellular level, we isolated NG2+ PDGFRß+ CD146+ CD34- CD31- CD45- PCs for detailed characterization. Functionally, we provide evidence that these PCs increased transepithelial electrical resistance and decreased endothelial permeability. We show for the first time that this population of PCs express contractile proteins, are stimulated by adrenergic signaling, and demonstrate stereotypical contraction and relaxation. Furthermore, we also studied for the first time, the PCs in in vitro models of disease. PCs in hypoxia activated the hypoxia-inducible factor 1 alpha pathway, increased secretion of angiogenic factors, and caused cellular apoptosis. Supraphysiological levels of low-density lipoprotein decreased PC proliferation and induced lipid droplet accumulation. Elevated glucose levels triggered a proinflammatory response. Taken together, our study characterizes cardiac PCs under in vitro disease conditions and supports the hypothesis that cardiac PCs are key vasoactive cells that can regulate blood flow in the heart.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Fenômeno de não Refluxo/fisiopatologia , Pericitos/fisiologia , Animais , Permeabilidade Capilar , Hipóxia Celular/fisiologia , Células Cultivadas , Circulação Coronária/fisiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , Masculino , Camundongos , Contração Miocárdica/fisiologia , Cultura Primária de Células
5.
Am J Physiol Heart Circ Physiol ; 298(3): H1088-95, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20061537

RESUMO

Variations in circadian rhythms are evident in the incidence of cardiovascular disease, and the risk of cardiovascular events increases when rhythms are disrupted. The suprachiasmatic nucleus is the central circadian pacemaker that regulates the daily rhythm of peripheral organs. Diurnal rhythms have more recently been shown to exist in myocardial tissue and are involved in metabolism and contractile function. Thus we sought to determine whether the functional deletion of the circadian rhythm mouse periodic gene 2 (mPer2) would protect the heart against ischemic injury. Nonreperfused myocardial infarction was induced in anesthetized, ventilated C57 (n = 17) and mPer2 mutant (mPer2-M; n = 15) mice via permanent ligation of the left anterior descending coronary artery. At 4 days post-myocardial infarction, we observed a 43% reduction of infarct area in mPer2-M mice compared with wild-type mice. This is coincident with 25% less macrophage infiltration, 43% higher capillary density, 17% increase in hypertrophy, and 15% less cardiomyocyte apoptosis in the infarct zone. Also, matrix metalloproteinase-9 was expressed in inflammatory cells in both groups, but total protein was 40% higher in wild-type mice, whereas it was not elevated in mPer2-M mice in response to injury. The functional deletion of the mPer2 gene reduces the severity of myocardial infarct injury by limiting the inflammatory response, reducing apoptosis, and inducing cardiomyocyte hypertrophy, thus preserving cardiac function. These findings collectively imply that the disruption of the circadian clock gene mPer2 is protective. Understanding the interactions between circadian rhythm genes and cardiovascular disease may provide insights into potential preventative and therapeutic strategies for susceptible populations.


Assuntos
Deleção de Genes , Infarto do Miocárdio/genética , Infarto do Miocárdio/prevenção & controle , Proteínas Circadianas Period/genética , Animais , Apoptose , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Hipertrofia , Ligadura/efeitos adversos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Infarto do Miocárdio/etiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia
6.
JCI Insight ; 5(8)2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32208384

RESUMO

Heart failure (HF) remains a grievous illness with poor prognosis even with optimal care. The apelin receptor (APJ) counteracts the pressor effect of angiotensin II, attenuates ischemic injury, and has the potential to be a novel target to treat HF. Intravenous administration of apelin improves cardiac function acutely in patients with HF. However, its short half-life restricts its use to infusion therapy. To identify a longer acting APJ agonist, we conducted a medicinal chemistry campaign, leading to the discovery of potent small-molecule APJ agonists with comparable activity to apelin by mimicking the C-terminal portion of apelin-13. Acute infusion increased systolic function and reduced systemic vascular resistance in 2 rat models of impaired cardiac function. Similar results were obtained in an anesthetized but not a conscious canine HF model. Chronic oral dosing in a rat myocardial infarction model reduced myocardial collagen content and improved diastolic function to a similar extent as losartan, a RAS antagonist standard-of-care therapy, but lacked additivity with coadministration. Collectively, this work demonstrates the feasibility of developing clinical, viable, potent small-molecule agonists that mimic the endogenous APJ ligand with more favorable drug-like properties and highlights potential limitations for APJ agonism for this indication.


Assuntos
Receptores de Apelina/agonistas , Coração/efeitos dos fármacos , Animais , Cães , Descoberta de Drogas , Insuficiência Cardíaca , Peptídeos e Proteínas de Sinalização Intercelular , Ratos
7.
Biochem Biophys Res Commun ; 386(4): 612-6, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19540196

RESUMO

Previous studies have demonstrated a role for angiotensin II (AngII) and myofibroblasts (myoFb) in cardiac fibrosis. However, the role of PKC-delta in AngII mediated cardiac fibrosis is unclear. Therefore, the present study was designed to investigate the role of PKC-delta in AngII induced cardiac collagen expression and fibrosis. AngII treatment significantly (p<0.05) increased myoFb collagen expression, whereas PKC-delta siRNA treatment or rottlerin, a PKC-delta inhibitor abrogated (p<0.05) AngII induced collagen expression. MyoFb transfected with PKC-delta over expression vector showed significant increase (p<0.05) in the collagen expression as compared to control. Two weeks of chronic AngII infused rats showed significant (p<0.05) increase in collagen expression compared to sham operated rats. This increase in cardiac collagen expression was abrogated by rottlerin treatment. In conclusion, both in vitro and in vivo data strongly suggest a role for PKC-delta in AngII induced cardiac fibrosis.


Assuntos
Angiotensina II/metabolismo , Colágeno/biossíntese , Miocárdio/patologia , Proteína Quinase C-delta/metabolismo , Acetofenonas/farmacologia , Angiotensina II/farmacologia , Animais , Benzopiranos/farmacologia , Proliferação de Células , Inibidores Enzimáticos/farmacologia , Fibroblastos/enzimologia , Fibroblastos/patologia , Fibrose , Masculino , Miocárdio/enzimologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
8.
J Vis Exp ; (150)2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31475977

RESUMO

Pericytes, perivascular cells of microvessels and capillaries, are known to play a part in angiogenesis, vessel stabilization, and endothelial barrier integrity. However, their tissue-specific functions in the heart are not well understood. Moreover, there is currently no protocol utilizing readily accessible materials to isolate and purify pericytes of cardiac origin. Our protocol focuses on using the widely used mammalian model, the mouse, as our source of cells. Using the enzymatic digestion and mechanical dissociation of heart tissue, we obtained a crude cell mixture that was further purified by fluorescence activating cell sorting (FACS) by a plethora of markers. Because there is no single unequivocal marker for pericytes, we gated for cells that were CD31-CD34-CD45-CD140b+NG2+CD146+. Following purification, these primary cells were cultured and passaged multiple times without any changes in morphology and marker expression. With the ability to regularly obtain primary murine cardiac pericytes using our protocol, we hope to further understand the role of pericytes in cardiovascular physiology and their therapeutic potential.


Assuntos
Separação Celular/métodos , Miocárdio/citologia , Pericitos/fisiologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Camundongos
9.
FASEB J ; 21(3): 656-70, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17209129

RESUMO

Recent observations demonstrated that translation of mRNAs may occur in axonal processes at sites that are long distances away from the neuronal perikaria. While axonal protein synthesis has been documented in several studies, the mechanism of its regulation remains unclear. The aim of this study was to investigate whether RNA interference (RNAi) may be one of the pathways that control local protein synthesis in axons. Here we show that sciatic nerve contains Argonaute2 nuclease, fragile X mental retardation protein, p100 nuclease, and Gemin3 helicase-components of the RNA-induced silencing complex (RISC). Application of short-interfering RNAs against neuronal beta-tubulin to the sciatic nerve initiated RISC formation, causing a decrease in levels of neuronal beta-tubulin III mRNA and corresponding protein, as well as a significant reduction in retrograde labeling of lumbar motor neurons. Our observations indicate that RNAi is functional in peripheral mammalian axons and is independent from the neuronal cell body or Schwann cells. We introduce a concept of local regulation of axonal translation via RNAi.


Assuntos
Axônios/metabolismo , Nervos Periféricos/citologia , Proteínas/metabolismo , Interferência de RNA/fisiologia , Animais , Proteína DEAD-box 20 , RNA Helicases DEAD-box/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Tubulina (Proteína)/efeitos dos fármacos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
10.
Cardiovasc Res ; 74(1): 140-50, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17320065

RESUMO

OBJECTIVES: Myofibroblasts (myoFb) are the major cell types that appear at the site of myocardial infarction (MI) in response to injury and play a vital role in tissue repair/remodeling. Since vascular endothelial growth factor (VEGF) plays a crucial role in the infarcted/ischemic heart, we hypothesized that activation of the peroxisome proliferator-activated receptor (PPAR)-gamma by its agonists induces VEGF expression while simultaneously decreasing inflammation (NF-kappaB). Such an increase in myoFb VEGF expression by PPAR-gamma agonists may play a role in angiogenesis. METHODS: Rat myoFb were treated with PPAR-gamma agonists and VEGF expression was measured by ELISA. The effect of these agonists on VEGF receptors was determined by qRT-PCR and flow-cytometric analysis. VEGF produced by these cells was also used for analysis of in vitro tubule formation (Matrigel assay). RESULTS: The PPAR-gamma activators troglitazone (TZ) and 15-deoxy-prostaglandin J2 (15J2) induced the expression of VEGF and its receptors (Flt-1 and KDR) in myoFb. TZ and 15J2 elicited a significant increase in the expression of KDR (14.7+/-1.0% and 9.6+/-2.1% respectively) and Flt-1 (24.5+/-2.0%, and 14.0+/-2.2% respectively) when compared to untreated myoFb. MyoFb treated with PPAR-gamma agonists increased extracellular VEGF, augmenting tubule formation on a Matrigel. The PPAR-gamma activator 15J2 significantly decreased the NF-kappaB activity in myoFb. CONCLUSION: This study demonstrates the induction of the VEGF accompanied by a reduction of NF-kappaB activity (inflammatory signaling) by PPAR-gamma agonists in cardiac myoFb. These results may further the understanding of the beneficial effects of PPAR-gamma agonists on infarcted tissue repair and angiogenesis.


Assuntos
Cromanos/farmacologia , Miócitos Cardíacos/metabolismo , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Tiazolidinedionas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Colágeno , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica , Laminina , Masculino , Microscopia de Fluorescência , Fibras Musculares Esqueléticas/fisiologia , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Prostaglandina D2/farmacologia , Proteoglicanas , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Troglitazona , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Pharmacol Res Perspect ; 6(6): e00442, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30459952

RESUMO

Tissue Inhibitor of Metalloproteinase 3 (TIMP3) is a secreted protein that has a great utility to inhibit elevated metalloproteinase (MMP) activity in injured tissues including infarcted cardiac tissue, inflamed vessels, and joint cartilages. An imbalance between TIMP3 and active MMP levels in the local tissue area may cause worsening of disease progression. To counter balance elevated MMP levels, exogenous administration of TIMP3 appeared to be beneficial in preclinical studies. However, the current form of WT-TIMP3 molecule has a limitation to be a therapeutic candidate due to low production yield, short plasma half-life, injection site retention, and difficulty in delivery, etc. We have engineered TIMP3 molecules by adding extra glycosylation sites or fusing with albumin, Fc, and antibody to improve pharmacokinetic properties. In general, the C-terminal fusion of TIMP3 improved expression and production in mammalian cells and extended half-lives dramatically 5-20 folds. Of note, a site-specific glycosylation at K22S/F34N resulted in a higher level of expression and better cardiac function compared to other fusion proteins in the context of left ventricle ejection fraction (LVEF) changes in a rat myocardial infarction model. It appeared that cardiac efficacy depends on a high ECM binding affinity, in which K22S/F34N and N-TIMP3 showed a higher binding to the ECM compared to other engineered molecules. In conclusion, we found that the ECM binding and sustained residence of injected TIMP3 molecules are important for cardiac tissue localization and inhibition of adverse remodeling activity.


Assuntos
Proteína ADAM17/antagonistas & inibidores , Metaloproteinases da Matriz/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Inibidor Tecidual de Metaloproteinase-3/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Proteína ADAM17/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos , Glicosilação , Humanos , Infusões Intravenosas , Injeções Intralesionais , Masculino , Mutação , Infarto do Miocárdio/etiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Inibidor Tecidual de Metaloproteinase-3/química , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/uso terapêutico , Resultado do Tratamento
12.
Reprod Biol Endocrinol ; 4: 7, 2006 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-16457734

RESUMO

BACKGROUND: Beta-defensins are small cationic peptides that exhibit broad spectrum antimicrobial properties. The majority of beta-defensins identified in humans are predominantly expressed in the male reproductive tract and have roles in non-immunological processes such as sperm maturation and capacitation. Characterization of novel defensins in the male reproductive tract can lead to increased understanding of their dual roles in immunity and sperm maturation. METHODS: In silico rat genomic analyses were used to identify novel beta-defensins related to human defensins 118-123. RNAs isolated from male reproductive tract tissues of rat were reverse transcribed and PCR amplified using gene specific primers for defensins. PCR products were sequenced to confirm their identity. RT-PCR analysis was performed to analyze the tissue distribution, developmental expression and androgen regulation of these defensins. Recombinant defensins were tested against E. coli in a colony forming unit assay to analyze their antimicrobial activities. RESULTS: Novel beta-defensins, Defb21, Defb24, Defb27, Defb30 and Defb36 were identified in the rat male reproductive tract. Defb30 and Defb36 were the most restricted in expression, whereas the others were expressed in a variety of tissues including the female reproductive tract. Early onset of defensin expression was observed in the epididymides of 10-60 day old rats. Defb21-Defb36 expression in castrated rats was down regulated and maintained at normal levels in testosterone supplemented animals. DEFB24 and DEFB30 proteins showed potent dose and time dependent antibacterial activity. CONCLUSION: Rat Defb21, Defb24, Defb27, Defb30 and Defb36 are abundantly expressed in the male reproductive tract where they most likely protect against microbial invasion. They are developmentally regulated and androgen is required for full expression in the adult epididymis.


Assuntos
Genitália Masculina/metabolismo , beta-Defensinas/genética , Sequência de Aminoácidos , Animais , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Mapeamento Cromossômico , Clonagem Molecular , Epididimo/metabolismo , Feminino , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genitália Masculina/imunologia , Masculino , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Ratos , Ratos Wistar , Alinhamento de Sequência , Testículo/metabolismo , Distribuição Tecidual , beta-Defensinas/metabolismo , beta-Defensinas/farmacologia
13.
Virus Res ; 114(1-2): 172-6, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16105704

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically associated with Kaposi's sarcoma (KS), primary effusion lymphoma(PEL), multicentric Castleman disease, and other tumors. Progression of KS is dictated by an aberrant production of inflammatory cytokines and increase in KSHV infection of cells. In this study, we analyzed the effect of cigarette smoke concentrate (CSC) on KSHV infection of human foreskin fibroblasts (HFF) using real time quantitative RT-PCR. Our results demonstrated that the CSC-treated cells supported 50% lower infection of KSHV when compared to the untreated cells. Radiolabeled-binding assays indicated that CSC inhibited KSHV infection of cells at a post attachment stage of entry. Taken together, we report for the first time the ability of CSC to specifically inhibit KSHV infection of cells.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 8/patogenicidade , Nicotiana , Fumaça , Células Cultivadas , Dimetil Sulfóxido/farmacologia , Fibroblastos/virologia , Herpesvirus Humano 8/efeitos dos fármacos , Humanos , Masculino , Nicotiana/química
15.
Brain Res Mol Brain Res ; 116(1-2): 163-7, 2003 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12941473

RESUMO

On day 4 after sciatic nerve crush injury, expression and localization of endothelin receptors ET(A) and ET(B) in the lumbar spinal cord were examined. Immunohistochemical staining with antibodies to ET(A) and ET(B) receptors showed cytoplasmic distribution of ET(A) receptors in motor neurons, whereas ET(B) receptors were localized in the perinuclear region. On the injured side of the lumbar spinal cord, when compared to contralateral, results demonstrated an up-regulation of ET(B) and a down-regulation of ET(A) receptors expression at the level of both mRNA and protein. These results suggest that ET(B) receptors may play a role in the regeneration of axotomized motor neurons.


Assuntos
Neurônios Motores/metabolismo , Regeneração Nervosa , Receptores de Endotelina/metabolismo , Medula Espinal/metabolismo , Animais , Western Blotting , Densitometria , Lateralidade Funcional , Imuno-Histoquímica , Região Lombossacral , Masculino , Camundongos , Camundongos Endogâmicos ICR , Compressão Nervosa/métodos , RNA Mensageiro/biossíntese , Receptores de Endotelina/classificação , Receptores de Endotelina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neuropatia Ciática/metabolismo , Medula Espinal/citologia
16.
Neuroreport ; 15(13): 2117-21, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15486493

RESUMO

Expression of vascular endothelial growth factor and its receptors Flt-1 and Flk-1 was studied in lumbar spinal cord after sciatic nerve crush injury. Immunohistochemical staining revealed strikingly different distribution of VEGF, Flt-1, and Flk-1 in lumbar motor neurons. VEGF was observed both in the nuclei and perikarya, while Flk-1 had cytoplasmic and Flt-1 perinuclear localization. Real-time RT-PCR showed a significant increase in the expression of VEGF and Flt-1 on the injured side of the lumbar spinal cord. The increased level of VEGF was also detected by immunoblot. Here we show that lumbar motor neurons increase the expression of VEGF and Flt-1 in response to injury. We propose that VEGF/Flt-1 signaling may be involved in regeneration of the spinal motor neurons.


Assuntos
Proteínas da Matriz Extracelular/biossíntese , Regulação da Expressão Gênica/fisiologia , Neuropatia Ciática/metabolismo , Medula Espinal/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Western Blotting/métodos , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Proteínas da Matriz Extracelular/genética , Lateralidade Funcional/fisiologia , Imuno-Histoquímica/métodos , Masculino , Camundongos , Cadeias Pesadas de Miosina , Compressão Nervosa/métodos , Miosina não Muscular Tipo IIB , RNA Mensageiro/biossíntese , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Neuropatia Ciática/genética , Medula Espinal/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
17.
Sci Transl Med ; 5(187): 187ra69, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23720580

RESUMO

Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.


Assuntos
Inibidores da Angiogênese/farmacologia , Vasos Coronários/efeitos dos fármacos , Coração/efeitos dos fármacos , Indóis/farmacologia , Microvasos/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Pirróis/farmacologia , Animais , Vasos Coronários/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Sunitinibe
18.
J Clin Invest ; 120(2): 472-84, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20071776

RESUMO

PDGFR is an important target for novel anticancer therapeutics because it is overexpressed in a wide variety of malignancies. Recently, however, several anticancer drugs that inhibit PDGFR signaling have been associated with clinical heart failure. Understanding this effect of PDGFR inhibitors has been difficult because the role of PDGFR signaling in the heart remains largely unexplored. As described herein, we have found that PDGFR-beta expression and activation increase dramatically in the hearts of mice exposed to load-induced cardiac stress. In mice in which Pdgfrb was knocked out in the heart in development or in adulthood, exposure to load-induced stress resulted in cardiac dysfunction and heart failure. Mechanistically, we showed that cardiomyocyte PDGFR-beta signaling plays a vital role in stress-induced cardiac angiogenesis. Specifically, we demonstrated that cardiomyocyte PDGFR-beta was an essential upstream regulator of the stress-induced paracrine angiogenic capacity (the angiogenic potential) of cardiomyocytes. These results demonstrate that cardiomyocyte PDGFR-beta is a regulator of the compensatory cardiac response to pressure overload-induced stress. Furthermore, our findings may provide insights into the mechanism of cardiotoxicity due to anticancer PDGFR inhibitors.


Assuntos
Coração/fisiopatologia , Miócitos Cardíacos/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Animais , Peso Corporal , Circulação Coronária , Coração/anatomia & histologia , Coração/efeitos dos fármacos , Coração/fisiologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Fosforilação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/deficiência , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Volume Sistólico , Suporte de Carga
19.
Hematol Oncol Clin North Am ; 23(1): 97-107, viii-ix, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19248973

RESUMO

Small-molecule tyrosine kinase inhibitors (TKIs) have revolutionized the targeted treatment of various cancers, including gastrointestinal stromal tumors (GISTs). Recent evidence suggests the possibility of cardiotoxicity secondary to TKI treatment of GISTs. Preclinical studies indicate that imatinib and sunitinib may be directly toxic to cardiac myocytes. Clinically, cardiotoxicity attributable to imatinib seems to be infrequent and manageable, whereas that attributable to sunitinib is more common and more severe. Further prospective studies with objective cardiac monitoring and long-term follow up are needed to define more accurately the incidence, natural history, and risk factors for developing cardiotoxicity associated with TKIs used in the treatment of patients who have GISTs. In this review, the authors discuss what is known regarding the cardiovascular effects of TKIs used in the treatment of GISTs.


Assuntos
Doenças Cardiovasculares/induzido quimicamente , Tumores do Estroma Gastrointestinal/complicações , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Antraciclinas/efeitos adversos , Tumores do Estroma Gastrointestinal/diagnóstico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Humanos , Inibidores de Proteínas Quinases/uso terapêutico
20.
J Gen Virol ; 87(Pt 5): 1139-1144, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16603514

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman's disease. KSHV infection of cells produces both latent and lytic cycles of infection. In vivo, the virus is found predominantly in the latent state. In vitro, a lytic infection can be induced in KSHV-infected cells by treating with phorbol ester (TPA). However, the exact signalling events that lead to the reactivation of KSHV lytic infection are still elusive. Here, a role is demonstrated for B-Raf/MEK/ERK signalling in TPA-induced reactivation of KSHV latent infection. Inhibiting MEK/ERK signalling by using MEK-specific inhibitors decreased expression of the TPA-induced KSHV lytic-cycle gene ORF8. Transfection of BCBL-1 cells with B-Raf small interfering RNA inhibited TPA-induced KSHV lytic infection significantly. Additionally, overexpression of MEK1 induced a lytic cycle of KSHV infection in BCBL-1 cells. The significance of these findings in understanding the biology of KSHV-associated pathogenesis is discussed.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Linhagem Celular Tumoral , Humanos , Ésteres de Forbol/farmacologia , Transdução de Sinais , Ativação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA