Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Drug Metab Rev ; 54(1): 95-100, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35084271

RESUMO

Gerontological research has accelerated dramatically in the last few decades. However, despite increased public interest, federal funding, an army of researchers, and many notable discoveries and high-impact publications, the goal of achieving even a modest extension of human lifespan seems to be as far away as ever or, at best, remains within the realm of lifestyle and diet optimization efforts. Humanity has already benefited from a lifespan revolution in the first half of the 20th Century, which was brought about by improved sanitation and hygiene, clean water, and our successful war on infectious diseases. Thanks to all these developments, in which gerontologists played no part, our expected lifespan increased by about 40% and our primary causes of death decidedly shifted from extrinsic to intrinsic causality. The next step is not that simple as it implies tackling intrinsic mechanisms of aging, and the lack of working human-specific antiaging solutions likely stems from flawed research strategies.


Assuntos
Geriatria , Envelhecimento , Humanos
2.
J Mol Cell Cardiol ; 80: 101-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25584774

RESUMO

Endothelin-1 (ET-1) plays a major role in regulating myocardial fibrosis in several pathological conditions, such as hypertension and diabetes. Aging is an independent risk factor for myocardial fibrosis. We hypothesized that ET-1 upregulation may be a basis of enhanced collagen synthesis in the senescent fibroblasts resulting in cardiac fibrosis with aging. To examine this hypothesis, we cultured mouse cardiac fibroblasts to passage-30 (P30). ß-Galactosidase activity and several other aging markers were markedly increased in P30 (vs. P3) fibroblasts, indicating that these cells were indeed undergoing senescence. Importantly, ET-1 expression was markedly upregulated in P30 (vs. P3) fibroblasts. Of note, estrogen receptor-α (ER-α), an important negative regulator of ET-1, was downregulated in P30 fibroblasts. We also studied aged (130-weeks old, female) mice hearts, and observed that ET-1 was upregulated and ER-α was downregulated in these hearts (vs. 6-week old mice hearts, female). Similar observations were made in the fibroblasts isolated from aged mice hearts. ET-1 upregulation with aging was also seen in ≈70-year old (vs. ≈30-year old) human heart sections. In concert with ET-1 upregulation, the expression of fibronectin and collagens was found to be markedly increased in P30 cardiac fibroblasts in culture, fibroblasts isolated from the aged mice hearts, and in aged human hearts. Interestingly, inhibition of ET-1 in the senescent P30 fibroblasts by 2 different strategies (the use of siRNA and the use of endothelin converting enzyme inhibitors) markedly suppressed expression of fibrosis signals. Further, treatment with synthetic ET-1 enhanced fibronectin and collagen expression in P3 cardiac fibroblasts. These observations in mice and human hearts suggest that aging-related cardiac fibrosis is, at least partially, dependent on the upregulation of ET-1.


Assuntos
Envelhecimento/genética , Endotelina-1/genética , Regulação da Expressão Gênica , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Senescência Celular/genética , Endotelina-1/metabolismo , Enzimas Conversoras de Endotelina , Ativação Enzimática , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Técnicas de Silenciamento de Genes , Humanos , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/metabolismo , Camundongos , Interferência de RNA , Transdução de Sinais , Regulação para Cima , beta-Galactosidase/metabolismo
3.
J Cardiovasc Pharmacol ; 65(2): 123-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24853489

RESUMO

Growth of atherosclerotic plaque requires neovascularization (angiogenesis). To elucidate the involvement of angiotensin II (Ang II) in angiogenesis, we performed gene microarray and microRNA (miRNA) polymerase chain reaction array analyses on human coronary artery endothelial cells exposed to moderate concentration of Ang II for 2 and 12 hours. At 12, but not 2, hours, cultures treated with Ang II exhibited shifts in transcriptional activity involving 267 genes (>1.5-fold difference; P < 0.05). Resulting transcriptome was most significantly enriched for genes associated with blood vessel development, angiogenesis, and regulation of proliferation. Majority of upregulated genes implicated in angiogenesis shared a commonality of being either regulators (HES1, IL-18, and CXCR4) or targets (ADM, ANPEP, HES1, KIT, NOTCH4, PGF, and SOX18) of STAT3. In line with these findings, STAT3 inhibition attenuated Ang II-dependent stimulation of tube formation in Matrigel assay. Expression analysis of miRNAs transcripts revealed that the pattern of differential expression for miRNAs was largely consistent with proangiogenic response with a prominent theme of upregulation of miRs targeting PTEN (miR-19b-3p, miR-21-5p, 23b-3p, and 24-3p), many of which are directly or indirectly STAT3 dependent. We conclude that STAT3 signaling may be an intrinsic part of Ang II-mediated proangiogenic response in human endothelial cells.


Assuntos
Angiotensina II , Células Endoteliais/metabolismo , Neovascularização Patológica/genética , Placa Aterosclerótica , Angiotensina II/genética , Angiotensina II/metabolismo , Técnicas de Cultura de Células , Proliferação de Células/genética , Vasos Coronários/metabolismo , Vasos Coronários/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Humanos , MicroRNAs/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Receptor Notch4 , Receptores CXCR4/genética , Receptores Notch/genética , Fatores de Transcrição SOXF/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética
4.
Am J Physiol Heart Circ Physiol ; 306(12): H1700-7, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24778168

RESUMO

Myocardial infarct size is determined by the death of nonischemic border zone cardiomyocytes caused by export of injury signals from the infarct zone. The countermeasures to limit infarct size, therefore, should be aimed at nonselective blockade of most, if not all, injury signals from entering nonischemic cells. To test whether inhibition of endocytosis might limit infarct size, HL-1 cardiomyocytes were subjected to anoxia (6 h) and reoxygenation (1 h). Anoxic and reoxygenated cells showed a multifold increase in mitochondrial ROS production accompanied with upregulation of scavenger receptors lectin-like oxidized low-density lipoprotein receptor-1 and CD36 and stimulation of stress signals, including NADPH oxidase subunit p22(phox), SOD2, and beclin-1. Incubation of healthy cardiomyocytes in media from anoxic and reoxygenated cells (conditioned media) resulted in qualitatively similar responses, including increase in the generation of mitochondrial ROS, p22(phox), SOD2, and beclin-1. Anoxia and reoxygenation caused collapse of clathrin-mediated endocytosis and stimulation of macropinocytosis, whereas in cultures exposed to conditioned media, the activity of endocytosis was uniformly higher. Conditioned media also significantly aggravated cytotoxic effects of TNF-α and angiotensin II, and suppression of endocytosis reversed these trends, resulting in an overall increase of metabolic activity. Moreover, inhibition of endocytosis prevented binding of oxidized cellular fragments with greater efficiency than targeted neutralization of the scavenger receptor lectin-like oxidized low-density lipoprotein receptor-1. Many of the observations in HL-1 cardiomyocytes were confirmed in primary cardiomyocyte cultures. Our data suggest that endocytosis is upregulated in border zone cardiomyocytes, and inhibition of endocytosis may be an effective approach to prevent export of injury signals from the infarct zone.


Assuntos
Endocitose/fisiologia , Hipóxia/complicações , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/fisiologia , Oxigênio/farmacologia , Angiotensina II/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Antígenos CD36/metabolismo , Células Cultivadas , Endocitose/efeitos dos fármacos , Camundongos , Modelos Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
5.
Cardiovasc Drugs Ther ; 28(5): 441-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25005756

RESUMO

Lectin-like oxidized low-density lipoprotein (ox-LDL) receptor-1 (LOX-1) is a major receptor for ox-LDL in endothelial cells. Its activation regulates endothelial proliferation, differentiation, migration and apoptosis. Recent in vitro studies show that LOX-1 activation by ox-LDL and angiotensin II (Ang II) induces angiogenesis via activation of NADPH oxidase and subsequent increase in ROS production. In this study, we investigated the effect of LOX-1 gene deletion (LOX-1 knockout or KO mice) on angiogenesis in response to prolonged Ang II infusion in vivo. Our studies showed that Ang II (vs. saline) infusion enhanced capillary formation in subcutaneously injected Matrigel® plugs. Ang II infusion also resulted in marked angiogenesis in the hearts as determined by CD31 immunopositivity. There was an increased expression (RT-PCR and Western blotting) of CD31 and VEGF in the hearts of mice infused with Ang II, indicating pro-angiogenic miliue. More importantly, LOX-1 KO mice reveled markedly limited angiogenesis in the Matrigel® plugs as well as in the hearts despite similar infusion with Ang II (all P < 0.05 vs. wild-type mice). In addition, the hearts of LOX-1 KO mice had attenuated expression of pro-inflammatory and angiogenic signals MCP-1 and IL-1ß following Ang II Infusion. Lastly, the rise in blood pressure in response to Ang II was less in the LOX-1 KO mice (P < 0.05 vs. wild-type mice). Our findings suggest that LOX-1 participates in angiogenesis in hypertension, which may be related to a state of inflammation.


Assuntos
Angiotensina II/farmacologia , Coração/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Depuradores Classe E/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Quimiocina CCL2/biossíntese , Colágeno/fisiologia , Combinação de Medicamentos , Coração/efeitos dos fármacos , Interleucina-1beta/biossíntese , Laminina/fisiologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteoglicanas/fisiologia , Receptores Depuradores Classe E/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese
6.
Can J Physiol Pharmacol ; 92(7): 524-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24959993

RESUMO

As a major receptor for oxidized low density lipoprotein (ox-LDL), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is upregulated in many pathophysiological events, including endothelial cell dysfunction and smooth muscle cell growth, as well as monocyte migration and transformation into foam cells, which are present in atherosclerosis and myocardial ischemia. Excessive production of reactive oxygen species (ROS) increases LOX-1 expression, induces mitochondrial DNA damage, and activates autophagy. Damaged mitochondrial DNA that escapes from autophagy induces an inflammatory response. This paper reviews the potential link between LOX-1, mitochondrial DNA damage, autophagy, and immune response in atherosclerosis.


Assuntos
Aterosclerose/imunologia , Aterosclerose/metabolismo , Autofagia , Dano ao DNA , DNA Mitocondrial/metabolismo , Estresse Oxidativo , Receptores Depuradores Classe E/metabolismo , Aterosclerose/patologia , Humanos
7.
J Mol Cell Cardiol ; 60: 184-90, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23648807

RESUMO

Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) is one of the most important receptors for binding and uptake of ox-LDL in endothelial cells, vascular smooth muscle cells and cardiomyocytes. In this study in cultured mice heart fibroblasts, we describe a decrease in LOX-1 expression as these cells go through successive passages. Further, fibroblast aging is associated with significant changes in morphology and proliferation ability. The same phenomena were observed in primary cardiac fibroblasts isolated from the aged mice (130-week). We also noted that the senescent fibroblasts have increased susceptibility to apoptosis and have a disorganized cytoskeleton. To ascertain the contribution of LOX-1 in the decline in proliferative ability and morphological changes in the aged cells, senescent fibroblasts were transfected with h-LOX-1. Transfection with h-LOX-1 resulted in cytoskeleton reorganization and partial restoration of the expression of related proteins, CDC42 and p70 S6 kinase. Upregulation of LOX-1 also significantly enhanced their proliferation potential and restored the expression of related genes Mdm2 and phos-Akt, and modestly reduced the expression of aging markers 4-HNE and ß-catenin. These findings suggest that LOX-1 contributes, at least in part, to the process of fibroblast senescence and may be viewed as a new aging maker.


Assuntos
Proliferação de Células , Senescência Celular , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Miocárdio/metabolismo , Receptores Depuradores Classe E/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Citoesqueleto/patologia , Fibroblastos/patologia , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocárdio/patologia , Receptores Depuradores Classe E/genética
8.
Biochem Biophys Res Commun ; 440(4): 696-700, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24099770

RESUMO

BACKGROUND: Autophagy and apoptosis are two important regulators of cell survival, and are often observed simultaneously in response to noxious stimuli. Anoxia is a known stimulus for autophagy and apoptosis, and angiotensin (Ang) II is a major mediator of anoxic injury. However, specific responses to anoxia and Ang II in terms of occurrence of autophagy and apoptosis have still not been delineated. METHODS AND RESULTS: We observed that autophagy (measured as LC3 staining, and Beclin-1 and p62 Western blotting) was an early response and apoptosis (measured as TUNEL staining, and Annexin V and Smac/Diablo Western blotting) became dominant as the duration of anoxia was prolonged. Autophagy also occurred quickly in response to low concentrations of Ang II. When exposed to high concentrations of Ang II, a significant number of cells developed apoptosis, while autophagy response decreased. Ang II-mediated apoptosis was blocked by Ang II type 1 receptor (AT1R) blocker losartan as well as by the AT2R blocker PD123319. Ang II-induced autophagy was blocked by losartan, but not by PD123319. CONCLUSION: Exposure to Ang II, a mediator of anoxic injury, initiates a rapid autophagy response, perhaps in an attempt to protect tissues from the impending noxious effects. However, when anoxia (and thereby release of Ang II) is prolonged, the process of apoptosis dominates. These processes will determine the outcome of cardiomyocyte well-being in states of hypoxia.


Assuntos
Angiotensina II/fisiologia , Apoptose , Autofagia , Miócitos Cardíacos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Angiotensina II/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Hipóxia Celular , Linhagem Celular , Proteínas de Choque Térmico/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Proteína Sequestossoma-1
9.
Apoptosis ; 17(8): 908-15, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22527949

RESUMO

Angiotensin II (Ang II) exerts its effects by activating its receptors, primarily type 1 (AT1R) and type 2 (AT2R). While the role of AT1R activation in cardiomyocyte physiology is well known, the role of AT2R in cardiomyocyte apoptosis remains controversial. To define the precise role of AT1R and AT2R in this process, we transfected HL-1 cardiomyocytes with AT1R or AT2R cDNA, and examined markers of apoptosis. We found that AT1R overexpression was associated with upregulation of endogenous AT2R expression, but AT2R overexpression did not affect endogenous AT1R expression. Caspase-3 staining indicated that overexpression of AT1R as well as AT2R resulted in cardiomyocyte apoptosis with appropriate alterations in annexin V, Bax and Bcl2 expression. Overexpression of AT1R and AT2R markedly increased IL-1ß (AT2R>AT1R), iNOS (AT2R>AT1R) and eNOS expression. AT2R-induced cell apoptosis could be blocked by the iNOS selective inhibitor 1,400 W, and did not require exogenous Ang II. These findings suggest that AT2R overexpression induces cardiomyocyte apoptosis, most likely via iNOS upregulation. AT1R-mediated cardiomyocyte apoptosis may be partially mediated by upregulation of endogenous AT2R.


Assuntos
Apoptose , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Angiotensina II/fisiologia , Animais , Anexina A5/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Expressão Gênica , Iminas/farmacologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/fisiologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Proteína X Associada a bcl-2/metabolismo
10.
Biochem Biophys Res Commun ; 426(1): 106-11, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22910414

RESUMO

BACKGROUND: Cell surface heparan sulfate proteoglycans (HSPG) play an important role in atherogenesis. We hypothesized that degradation of HSPG may increase the binding of atherogenic oxidized low density lipoprotein (ox-LDL) to endothelial cells, and result in extensive HSPG degradation as well as autophagy and apoptosis. METHODS: Primary human umbilical vein endothelial cells (HUVECs) were used to study the expression of lectin-like ox-LDL receptor-1 (LOX-1), HSPG, autophagy and apoptosis in response to ox-LDL and heparinase III (Hep III). RESULTS: As expected, ox-LDL treatment resulted in LOX-1 expression, ox-LDL uptake and reactive oxygen species (ROS) generation. Ox-LDL treatment also resulted in a modest degradation of HSPG and increase in autophagy (expression of LC3, beclin-1 and Atg5) and apoptosis (enhanced expression of caspases and Bax, and reduced expression of Bcl-2 and Bcl-xL). The effects of ox-LDL were blocked by pretreatment of cells with LOX-1 antibody or apocynin, an NADPH oxidase inhibitor. Hep III alone caused HSPG degradation and slightly, but significantly, increased ROS generation, and induced autophagy and caspase expression. However, autophagy and apoptosis induced by Hep III were not affected by apocynin or LOX-1 antibody. Importantly, Hep III pretreatment of cells significantly enhanced ox-LDL-induced HSPG degradation, LOX-1 expression, ox-LDL uptake and ROS generation as well as autophagy and apoptosis. CONCLUSION: These data demonstrate that Hep III enhances the pro-atherosclerotic characteristics in HUVECs induced by ox-LDL.


Assuntos
Apoptose , Autofagia , Proteoglicanas de Heparan Sulfato/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Lipoproteínas LDL/metabolismo , Acetofenonas/farmacologia , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipoproteínas LDL/farmacologia , Polissacarídeo-Liases/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores Depuradores Classe E/biossíntese
11.
Toxicol Appl Pharmacol ; 259(3): 346-54, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22306536

RESUMO

Aspirin (acetyl salicylic acid, ASA) is a common drug used for its analgesic and antipyretic effects. Recent studies show that ASA not only blocks cyclooxygenase, but also inhibits NADPH oxidase and resultant reactive oxygen species (ROS) generation, a pathway that underlies pathogenesis of several ailments, including hypertension and tissue remodeling after injury. In these disease states, angiotensin II (Ang II) activates NADPH oxidase via its type 1 receptor (AT1R) and leads to fibroblast growth and collagen synthesis. In this study, we examined if ASA would inhibit NADPH oxidase activation, upregulation of AT1R transcription, and subsequent collagen generation in mouse cardiac fibroblasts challenged with Ang II. Mouse heart fibroblasts were isolated and treated with Ang II with or without ASA. As expected, Ang II induced AT1R expression, and stimulated cardiac fibroblast growth and collagen synthesis. The AT1R blocker losartan attenuated these effects of Ang II. Similarly to losartan, ASA, and its SA moiety suppressed Ang II-mediated AT1R transcription and fibroblast proliferation as well as expression of collagens and MMPs. ASA also suppressed the expression of NADPH oxidase subunits (p22(phox), p47(phox), p67(phox), NOX2 and NOX4) and ROS generation. ASA did not affect total NF-κB p65, but inhibited its phosphorylation and activation. These observations suggest that ASA inhibits Ang II-induced NADPH oxidase expression, NF-κB activation and AT1R transcription in cardiac fibroblasts, and fibroblast proliferation and collagen expression. The critical role of NADPH oxidase activity in stimulation of AT1R transcription became apparent in experiments where ASA also inhibited AT1R transcription in cardiac fibroblasts challenged with H2O2. Since SA had similar effect as ASA on AT1R expression, we suggest that ASA's effect is mediated by its SA moiety.


Assuntos
Aspirina/farmacologia , Colágeno/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética , Angiotensina II/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno/biossíntese , Fibroblastos/metabolismo , Losartan/farmacologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica/efeitos dos fármacos
12.
Curr Atheroscler Rep ; 2012 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-22286193

RESUMO

Atherosclerosis is characterized by accumulation of lipids and inflammatory cells in the arterial wall. Oxidized low-density lipoprotein (ox-LDL) plays important role in the genesis and progression of atheromatous plaque. Various scavenger receptors have been recognized in the past two decades that mediate uptake of ox-LDL leading to formation of foam cells. Inhibition of scavenger receptor A and CD36 has been shown to affect progression of atherosclerosis by decreasing foam cell formation. Lectin-type oxidized LDL receptor 1 (LOX-1) participates at various steps involved in the pathogenesis of atherosclerosis, and in experimental studies its blockade has been shown to affect the progression of atherosclerosis at multiple levels. In this review, we summarize the role of ox-LDL and scavenger receptors in the formation of atheroma with emphasis on effects of LOX-1 blockade.

13.
J Cardiovasc Pharmacol ; 60(2): 187-92, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22561363

RESUMO

Aspirin [acetyl salicylic acid (ASA)] inhibits nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and reactive oxygen species generation, a pathway that underlies formation of new capillaries (angiogenesis). Angiotensin II (Ang II) participates in angiogenesis by activating type 1 receptor (AT1R). We examined if ASA would inhibit AT1R transcription, which requires NADPH oxidase, and thereby new capillary formation. Human umbilical vein endothelial cells were cultured in Matrigel and treated with Ang II with and without ASA. Expression of AT1R and NADPH oxidase was measured by quantitative polymerase chain reaction. Ang II in low concentrations induced AT1R messenger RNA and new capillary formation. ASA and its salicylic acid (SA) moiety both suppressed Ang II-mediated AT1R and vascular endothelial growth factor expression and the subsequent new capillary formation. Of note, the AT1R blocker losartan prevented new capillary formation. ASA and SA also suppressed NADPH oxidase (p22, p47, p67, and gp91 messenger RNA) expression. These observations suggest that ASA can inhibit Ang II-induced capillary formation in part via blocking NADPH oxidase and AT1R transcription. Because SA moiety had similar effect as ASA on AT1R expression, we suggest that the effect of ASA on new capillary formation is mediated by its SA moiety.


Assuntos
Inibidores da Angiogênese/farmacologia , Aspirina/farmacologia , Capilares/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Capilares/metabolismo , Células Cultivadas , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Losartan/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Ácido Salicílico/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 301(2): H506-13, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21602467

RESUMO

Oxidized LDL (ox-LDL) plays a critical role in atherogenesis, including apoptosis. As hypercholesterolemia causes epigenetic changes resulting in long-term phenotypic consequences, we hypothesized that repeated and continuous exposure to ox-LDL may alter the pattern of apoptosis in human umbilical vein endothelial cells (HUVECs). We also analyzed global and promoter-specific methylation of apoptosis-related genes. As expected, ox-LDL evoked a dose-dependent increase in apoptosis in the first passage HUVECs that was completely abrogated by lectin-like ox-LDL receptor (LOX-1)-neutralizing antibody. Ox-LDL-induced apoptosis was associated with upregulation of proapoptotic LOX-1, ANXA5, BAX, and CASP3 and inhibition of antiapoptotic BCL2 and cIAP-1 genes accompanied with reciprocal changes in the methylation of promoter regions of these genes. Subsequent passages of cells displayed attenuated apoptotic response to repeat ox-LDL challenge with blunted gene expression and exaggerated methylation of LOX-1, BAX, ANXA5, and CASP3 genes (all P < 0.05 vs. first exposure to ox-LDL). Treatment of cells with LOX-1 antibody before initial ox-LDL treatment prevented both gene-specific promoter methylation and expression changes and reduction of apoptotic response to repeat ox-LDL challenge. Based on these data, we conclude that exposure of HUVECs to ox-LDL induces epigenetic changes leading to resistance to apoptosis in subsequent generations and that this effect may be related to the LOX-1-mediated increase in DNA methylation.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Aterosclerose/metabolismo , Proliferação de Células , Metilação de DNA , Células Endoteliais/metabolismo , Lipoproteínas LDL/metabolismo , Regiões Promotoras Genéticas , Análise de Variância , Anticorpos Neutralizantes , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Células Endoteliais/patologia , Regulação da Expressão Gênica , Humanos , Fenótipo , Receptores Depuradores Classe E/imunologia , Receptores Depuradores Classe E/metabolismo
15.
Cardiovasc Drugs Ther ; 25(5): 495-500, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21826406

RESUMO

There is much interest in the role of oxidant stress in an ever-increasing list of disease states. However, the precise mediator of oxidant stress and the stressor molecule/s have not been identified. Accordingly, trials of inhibitors of oxidant stress in animal models of disease states have met only limited success. The trials of traditional anti-oxidant vitamins have been largely unsuccessful in the treatment of a wide array of disease states in humans. Recent identification of LOX-1 in vascular endothelial cells and its activation by oxidant species have led to a marked improvement in our understanding of the pathology of several cardiovascular disease states. Here, we review the disease states where therapy targeted at LOX-1 inhibition might be helpful.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/metabolismo , Receptores Depuradores Classe E/antagonistas & inibidores , Receptores Depuradores Classe E/metabolismo , Animais , Humanos , Estresse Oxidativo/efeitos dos fármacos
16.
J Cardiovasc Pharmacol ; 56(6): 635-41, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881612

RESUMO

Hypoxia­reoxygenation (HR) is a primary driver of angiogenesis in both atherogenesis and tumorigenesis. The main target of hypoxia-driven proangiogenic signaling is adherens junctions responsible for contact inhibition of endothelial cells. We analyzed the effects of hypoxia (8­12 hours) followed by a brief period of reoxygenation (2 hours) (HR) on angiogenesis and integrity of adherens junction in cultured human umbilical vein endothelial cells as well as the effects of aspirin on modulation of human umbilical vein endothelial cells' response to HR. Cells exposed to HR displayed considerable enhancement of tube formation (angiogenesis) on matrigel. Immunocytostaining of near-confluent cells revealed that HR caused disruption of adherens junctions and internalization of their components VE-cadherin, p120 catenin, and b-catenin. Additionally, HR resulted in the appearance of binucleated cells, and VE-cadherin in colocalization with b-catenin was found to be positioned between the separating nuclei. Presence of aspirin (acetylsalicylic acid, 1 mM) resulted in preservation of adherens junctions on the cellular membrane and prevented angiogenesis as well as mitosis. HR caused upregulation LOX-1, the p47(phox) subunit of NADPH, while reducing transcription of endothelial nitric oxide synthase. Aspirin had no effect on endothelial nitric oxide synthase and canceled the transcriptional activation of the LOX-1 and p47(phox) subunit of NADPH oxidase. Based on these data, we hypothesize that aspirin preserves the integrity of adherens junctions and thus blunts angiogenic response to HR through downregulation of LOX-1 and the LOX-1-mediated p47(phox) component of NADPH oxidase transcription, thus preventing NADPH oxidase assembly and function.


Assuntos
Inibidores da Angiogênese/farmacologia , Antimitóticos/farmacologia , Aspirina/farmacologia , NADPH Oxidases/fisiologia , Oxigênio/fisiologia , Receptores Depuradores Classe E/fisiologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/enzimologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Biochem Biophys Res Commun ; 379(2): 395-9, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19126408

RESUMO

LOX-1, a cell surface lectin-like receptor, is upregulated by oxidized low-density lipoprotein (ox-LDL) and angiotensin II (Ang II), and plays an important role in host defense. The specific C-type lectin domain on LOX-1 is essential for ox-LDL binding and internalization, generation of oxidant species and eliciting immune response. Here, we show that LOX-1 deletion alters genes that relate to immune response. Microarray (and qPCR) analysis of cardiac tissues showed downregulated expression of several immunoglobulins (Igk-V8, Igk-C, Igh-6, Igj, Ighg, Igh, and Igl-V1) in the LOX-1 knockout (KO) mice [p<0.05 vs. the wild-type (WT) mice]. The expression of these immunoglobulins was upregulated several-fold in the LOX-1 KO mice hearts when these mice were infused with Ang II (p<0.05, vs. WT mice). Importantly, cultured mouse HL-1 cardiomyocytes expressed these immunoglobulins, and pretreatment of cardiomyocytes with a specific anti-LOX-1 antibody enhanced the generation of immunoglobulins upon subsequent exposure to Ang II. These observations mirrored the data obtained from WT and LOX-1 KO mice hearts in the resting state and following Ang II infusion. This study provides first set of data on immunoglobulin expression in cardiac tissues of WT and LOX-1 KO mice and in cultured HL-1 cardiomyocytes, and demonstrates that LOX-1 inactivation leads to upregulation of immunoglobulins in cardiomyocytes upon challenge with Ang II.


Assuntos
Imunoglobulinas/biossíntese , Miócitos Cardíacos/imunologia , Receptores Depuradores Classe E/genética , Angiotensina II/farmacologia , Animais , Regulação para Baixo , Deleção de Genes , Imunidade/genética , Imunoglobulinas/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Depuradores Classe E/metabolismo , Regulação para Cima
18.
Biochem Biophys Res Commun ; 387(3): 543-7, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19615978

RESUMO

Statins, inhibitors of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, have been shown to induce both angiogenic and angiostatic responses. We attempted to resolve this controversy by studying the effects of two different statins, rosuvastatin and simvastatin, in two different assay systems. In the matrigel angiogenesis assay, both statins enhanced tube formation by human umbilical vein endothelial cells (HUVECs, p<0.01 vs. control). In the ex vivo mouse aortic ring sprouting assay, both statins virtually abolished new vessel formation (p<0.01). As a basic difference between the two models of angiogenesis is dispersed state of endothelial cells vs. compact monolayer, we analyzed influence of statins on endothelial junction proteins. RT-PCR analysis and cytoimmunostaining of HUVECs treated with simvastatin revealed increased expression of VE-cadherin (p<0.05). The blockade of VE-cadherin with a specific antibody reversed simvastatin-induced tube formation (p<0.002). These data suggest that statins through VE-cadherin stimulation modulate cell-cell adhesion and diminish the ability of cells to proliferate and migrate. The observations of reduced angiogenesis in the intact vessel may relate to anti-atherosclerotic and anti-cancer effects of statins, and provide a feasible explanation for conflicting data under different experimental conditions.


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Fluorbenzenos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Pirimidinas/farmacologia , Sinvastatina/farmacologia , Sulfonamidas/farmacologia , Animais , Antígenos CD/biossíntese , Aorta/citologia , Aorta/efeitos dos fármacos , Caderinas/antagonistas & inibidores , Caderinas/biossíntese , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Rosuvastatina Cálcica , Veias Umbilicais/citologia , Veias Umbilicais/efeitos dos fármacos
19.
J Neurosci Res ; 87(12): 2748-56, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19437551

RESUMO

Mutations of the survival motor neuron (SMN) gene in spinal muscular atrophy (SMA) lead to anterior horn cell death. The cause is unknown, but motor neurons depend substantially on mitochondrial oxidative phosphorylation (OxPhos) for normal function. Therefore, mitochondrial parameters were analyzed in an SMA cell culture model using small interfering RNA (siRNA) transfection that decreased Smn expression in NSC-34 cells to disease levels. Smn siRNA knock-down resulted in 35% and 66% reduced Smn protein levels 48 and 72 hr posttransfection, respectively. ATP levels were reduced by 14% and 26% at 48 and 72 hr posttransfection, respectively, suggesting decreased ATP production or increased energy demand in neural cells. Smn knock-down resulted in increased mitochondrial membrane potential and increased free radical production. Changes in activity of cytochrome c oxidase (CcO), a key OxPhos component, were observed at 72 hr with a 26% increase in oxygen consumption. This suggests a compensatory activation of the aerobic pathway, resulting in increased mitochondrial membrane potentials, a condition known to lead to the observed increase in free radical production. Further testing suggested that changes in ATP at 24 hr precede observable indices of cell injury at 48 hr. We propose that energy paucity and increased mitochondrial free radical production lead to accumulated cell damage and eventual cell death in Smn-depleted neural cells. Mitochondrial dysfunction may therefore be important in SMA pathology and may represent a new therapeutic target.


Assuntos
Doenças Mitocondriais/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , Fosforilação Oxidativa , Medula Espinal/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Regulação para Baixo/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metabolismo Energético/fisiologia , Radicais Livres/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Mitocôndrias/metabolismo , Doenças Mitocondriais/fisiopatologia , Neurônios Motores/patologia , Atrofia Muscular Espinal/fisiopatologia , Estresse Oxidativo/fisiologia , RNA Interferente Pequeno/genética , Medula Espinal/fisiopatologia , Proteína 1 de Sobrevivência do Neurônio Motor/genética
20.
Mech Ageing Dev ; 166: 55-63, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28765009

RESUMO

Overexpression of telomerase has been shown to significantly increase the lifespan of mice. When mechanistically attributed to repair of critically short telomeres, the lifespan extending action of telomerase cannot be reconciled with the observation that telomerase-null mice do not exhibit shortening of lifespan for at least two generations. We hypothesized that telomerase may interfere with replication of mitochondrial DNA (mtDNA) in a way that reduces formation of deletions - the primary cause of age-dependent cell attrition in non-renewable cells such as myocytes and neurons. Here we show that several tRNA genes may function as alternative origins of replication (ORIs). We also show that telomerase interacts with canonical light strand ORI (ORIL) and tRNAs and modifies their activities. Our results suggest that replication of mitochondrial DNA (mtDNA) proceeds through a variety of mechanisms resulting in a mixture of classic strand-displacement mode, and coupled replication of heavy and light strands. Our results also suggest that effects of telomerase may arise from binding ORIL and thus limiting contribution of the deletion-prone strand displacement mode to mtDNA synthesis. These findings imply that it may be possible to uncouple detrimental and beneficial effects of telomerase, and thereby to improve telomerase-based strategies to extend lifespan.


Assuntos
Replicação do DNA , DNA Mitocondrial/biossíntese , RNA de Transferência/metabolismo , Origem de Replicação , Telomerase/metabolismo , Animais , DNA Mitocondrial/genética , Humanos , Camundongos , RNA de Transferência/genética , Telomerase/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa