Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Cardiovasc Med ; 5: 12, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29564335

RESUMEN

Aging is a complex process that results from a combination of environmental, genetic, and epigenetic factors. A chronic pro-inflammatory status is a pervasive feature of aging. This chronic low-grade inflammation occurring in the absence of overt infection has been defined as "inflammaging" and represents a significant risk factor for morbidity and mortality in the elderly. The low-grade inflammation persists even after reversing pro-inflammatory stimuli such as LDL cholesterol and the renin-angiotensin system (RAS). Recently, several possible sources of chronic low-grade inflammation observed during aging and age-related diseases have been proposed. Cell senescence and dysregulation of innate immunity is one such mechanism by which persistent prolonged inflammation occurs even after the initial stimulus has been removed. Additionally, the coagulation factor that activates inflammatory signaling beyond its role in the coagulation system has been identified. This signal could be a new source of chronic inflammation and cell senescence. Here, we summarized the factors and cellular pathways/processes that are known to regulate low-grade persistent inflammation in aging and age-related disease.

2.
Medicines (Basel) ; 5(2)2018 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-29601487

RESUMEN

The World Health Organization announced that cardiovascular disease is the number one cause of death globally, representing 31% of all global deaths. Coronary artery disease (CAD) affects approximately 5% of the US population aged 40 years and older. With an age-adjusted prevalence of approximately 12%, peripheral artery disease (PAD) affects at least 8 to 12 million Americans. Both CAD and PAD are caused by mainly atherosclerosis, the hardening and narrowing of arteries over the years by lipid deposition in the vascular bed. Despite the significant advances in interventions for revascularization and intensive medical care, patients with CAD or PAD who undergo percutaneous transluminal angioplasty have a persistent high rate of myocardial infarction, amputation, and death. Therefore, new therapeutic strategies are urgently needed for these patients. To overcome this unmet need, therapeutic angiogenesis using angiogenic growth factors has evolved in an attempt to stimulate the growth of new vasculature to compensate for tissue ischemia. After nearly 20 years of investigation, there is growing evidence of successful or unsuccessful gene therapy for ischemic heart and limb disease. This review will discuss basic and clinical data of therapeutic angiogenesis studies employing angiogenic growth factors for PAD patients and will draw conclusions on the basis of our current understanding of the biological processes of new vascularization.

3.
Artículo en Inglés | MEDLINE | ID: mdl-29515523

RESUMEN

Cellular senescence is the complex process of deterioration that drives the aging of an organism, resulting in the progressive loss of organ function and eventually phenotypic aging. Senescent cells undergo irreversible growth arrest, usually by inducing telomere shortening. Alternatively, senescence may also occur prematurely in response to various stress stimuli, such as oxidative stress, DNA damage, or activated oncogenes. Recently, it has been shown that IGF binding protein-5 (IGFBP-5) with the induction of the tumor suppressor p53 is upregulated during cellular senescence. This mechanism mediates interleukin-6/gp130-induced premature senescence in human fibroblasts, irradiation-induced premature senescence in human endothelial cells (ECs), and replicative senescence in human ECs independent of insulin-like growth factor I (IGF-I) and IGF-II. Additionally, a link between IGFBP-5, hyper-coagulation, and inflammation, which occur with age, has been implicated. Thus, IGFBP-5 seems to play decisive roles in controlling cell senescence and cell inflammation. In this review, we describe the accumulating evidence for this role of IGFBP-5 including our new finding.

4.
Sci Rep ; 7(1): 17172, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29215061

RESUMEN

Our previous study demonstrated that coagulation factor Xa (FXa) induced endothelial cell senescence, resulting in inflammation and impaired angiogenesis. This mechanism is dictated through protease-activated receptors, PARs, insulin-like growth factor-binding protein 5 (IGFBP-5), and p53. Activation of PARs contributes to the pathophysiology of several chronic inflammatory diseases, including atherosclerosis. Thus, we speculated that similar mechanism might participate in the progression of atherosclerotic plaques. In the present study, we successfully identified the cells that produced FX/Xa in atherosclerosis using human atherosclerotic plaques obtained from carotid endarterectomy. In situ hybridization for FX revealed that FX was generated in vascular smooth muscle cells (VSMC), inflammatory cells, and endothelial cells. Then, we examined the effects of FXa on the growth of VSMC in vitro. The present study revealed that chronic FXa stimulation significantly induced the senescence of VSMC with concomitant upregulation of IGFBP-5 and p53. Inhibition of FXa signaling with rivaroxaban or knock down of IGFBP-5 significantly reduced FXa-induced VSMC senescence and inflammatory cytokine production. Finally, we confirmed that FXa and IGFBP-5 are co-distributed in atherosclerotic plaques. In conclusion, induction of senescence of VSMC induced by locally produced FX/Xa may contribute to the progression of atherosclerosis.


Asunto(s)
Senescencia Celular , Factor Xa/metabolismo , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Músculo Liso Vascular/patología , Placa Aterosclerótica/patología , Células Cultivadas , Humanos , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/metabolismo , Transducción de Señal
5.
Sci Rep ; 7(1): 130, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28273932

RESUMEN

Obesity and its associated chronic inflammation in adipose tissue initiate insulin resistance, which is related to several pathologies including hypertension and atherosclerosis. Previous reports demonstrated that circulating hepatocyte growth factor (HGF) level was associated with obesity and type 2 diabetes. However, its precise role in obesity and related-pathology is unclear. In this experiment, cardiac-specific over-expression of human HGF in mice (HGF-Tg mice) which showed 4-5 times higher serum HGF levels than wild-type mice were used. While body weight in wild-type mice fed with high fat diet (HFD) for 14 weeks was significantly increased accompanied with insulin resistance, HGF-Tg mice prevented body weight gain and insulin resistance. The accumulation of macrophages and elevated levels of inflammatory mediators in adipose tissue were significantly inhibited in HGF-Tg mice as compared to wild-type mice. The HFD-induced obesity in wild-type mice treated with HGF-neutralizing antibody showed an exacerbated response to the glucose tolerance test. These gain-of-function and loss-of-function studies demonstrated that the elevated HGF level induced by HFD have protective role against obesity and insulin resistance.


Asunto(s)
Dieta Alta en Grasa , Factor de Crecimiento de Hepatocito/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Animales , Glucemia , Peso Corporal , Humanos , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Insulina/sangre , Masculino , Ratones Endogámicos C57BL , Obesidad/complicaciones
6.
Sci Rep ; 6: 35580, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27752126

RESUMEN

Uncontrolled coagulation contributes to the pathophysiology of several chronic inflammatory diseases. In these conditions, senescent cells are often observed and is involved in the generation of inflammation. The coincidence of hyper-coagulation, cell senescence, and inflammation suggests the existence of a common underlying mechanism. Recent evidence indicates that activated coagulation factor X (FXa) plays a role in the processes beyond blood coagulation. This non-hematologic function entails the mediation of inflammation and tissue remodeling. We therefore tested the hypothesis that FXa induces cell senescence resulting in tissue inflammation and impaired tissue regeneration. Human umbilical vein endothelial cells were stimulated with FXa for 14 days. The proliferation of cells treated with FXa was significantly smaller, and the fraction of senescence-associated ß-galactosidase-positive cells was increased as compared to the control group. RT-qPCR array revealed that FXa increased the expression of IGFBP-5, EGR-1, p53, and p16INK4a. Inhibition of FXa by a direct FXa inhibitor, rivaroxaban, or IGFBP-5 by siRNA decreased FXa-induced cell senescence, restoring cell proliferation. Moreover, in an ischemic hind limb mouse model, FXa inhibited neovascularization by endothelial progenitor cell. However, rivaroxaban significantly restored FXa-induced impaired angiogenesis. In summary, FXa induced endothelial cell senescence through IGFBP-5, resulting in impaired angiogenesis.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Factor Xa/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Factor Xa/administración & dosificación , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Regeneración/efectos de los fármacos
7.
Arterioscler Thromb Vasc Biol ; 36(3): 545-52, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26769045

RESUMEN

OBJECTIVE: Peripheral arterial disease is highly prevalent in the elderly and in the subjects with cardiovascular risk factors such as diabetes. Approximately 2% to 4% of those affected with peripheral arterial disease commonly complain of intermittent claudication. Cilostazol, a type III phosphodiesterase inhibitor, is the only Food and Drug Administration-approved drug for the treatment of intermittent claudication. Cilostazol has been shown to be beneficial for the improvement of pain-free walking distance in patients with intermittent claudication in a series of randomized clinical trials. However, the underlying mechanism how cilostazol improved intermittent claudication symptoms is still unclear. APPROACH AND RESULTS: In this study, the effect of cilostazol on ischemic leg was investigated in mouse ischemic hindlimb model. Administration of cilostazol significantly increased the expression of hepatocyte growth factor (HGF), vascular endothelial growth factor, angiopoietin-1, and peroxisome proliferator-activated receptor-γ in vasculature. The capillary density in ischemic leg was also significantly increased in cilostazol treatment group when compared with control and aspirin treatment group. However, an increase in capillary density and the expression of growth factors was almost completely abolished by coadministration of HGF-neutralizing antibody, suggesting that cilostazol enhanced angiogenesis mainly through HGF. In vitro experiment revealed that cilostazol treatment increased HGF production in vascular smooth muscle cells via 2 major pathways: peroxisome proliferator-activated receptor-γ and cAMP pathways. CONCLUSIONS: Our data suggest that the favorable effects of cilostazol on ischemic leg might be through the angiogenesis through the induction of HGF via peroxisome proliferator-activated receptor-γ and cAMP pathways.


Asunto(s)
Inductores de la Angiogénesis/farmacología , AMP Cíclico/metabolismo , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , PPAR gamma/agonistas , Inhibidores de Fosfodiesterasa 3/farmacología , Sistemas de Mensajero Secundario , Tetrazoles/farmacología , Angiopoyetina 1/metabolismo , Animales , Capilares/efectos de los fármacos , Capilares/enzimología , Capilares/fisiopatología , Células Cultivadas , Cilostazol , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Isquemia/enzimología , Isquemia/genética , Isquemia/fisiopatología , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , PPAR gamma/metabolismo , Ratas , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Hypertension ; 67(2): 356-61, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26644236

RESUMEN

We previously reported that overexpression of full-length periostin, Pn-1, resulted in ventricular dilation with enhanced interstitial collagen deposition in a rat model. However, other reports have documented that the short-form splice variants Pn-2 (lacking exon 17) and Pn-4 (lacking exons 17 and 21) promoted cardiac repair by angiogenesis and prevented cardiac rupture after acute myocardial infarction. The apparently differing findings from those reports prompted us to use a neutralizing antibody to selectively inhibit Pn-1 by blockade of exon 17 in a rat acute myocardial infarction model. Administration of Pn neutralizing antibody resulted in a significant decrease in the infarcted and fibrotic areas of the myocardium, which prevented ventricular wall thinning and dilatation. The inhibition of fibrosis by Pn neutralizing antibody was associated with a significant decrease in gene expression of fibrotic markers, including collagen I, collagen III, and transforming growth factor-ß1. Importantly, the number of α-smooth muscle actin-positive myofibroblasts was significantly reduced in the hearts of animals treated with Pn neutralizing antibody, whereas cardiomyocyte proliferation and angiogenesis were comparable in the IgG and neutralizing antibody groups. Moreover, the level of Pn-1 expression was significantly correlated with the severity of myocardial infarction. In addition, Pn-1, but not Pn-2 or Pn-4, inhibited fibroblast and myocyte attachment, which might account for the cell slippage observed during cardiac remodeling. Collectively, these results indicate that therapeutics that specifically inhibit Pn exon-17, via a neutralizing antibody or drug, without suppressing other periostin variants might offer a new class of medication for the treatment of acute myocardial infarction patients.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Ventrículos Cardíacos/fisiopatología , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocardio/patología , Remodelación Ventricular/fisiología , Animales , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Exones , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Reacción en Cadena de la Polimerasa , ARN/genética , Ratas , Ratas Endogámicas Lew
9.
Expert Opin Biol Ther ; 15(3): 381-90, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633211

RESUMEN

INTRODUCTION: Despite the remarkable progress of medicine and endovascular procedures for revascularization, patients with critical limb ischemia (CLI) remain at high risk for amputation and often have a low quality of life due to pain and ulcers in the ischemic leg. Thus, a novel strategy for generating new blood vessels in CLI patients without treatment options is vital. Pre-clinical studies and Phase I clinical trials using VEGF and fibroblast growth factor (FGF) demonstrated promising results; however, more rigorous Phase II and III clinical trials failed to demonstrate benefits for CLI patients. Recently, two multicenter, double-blind, placebo-controlled clinical trials in Japan (Phase III) and the USA (Phase II) showed the benefits of hepatocyte growth factor (HGF) gene therapy for CLI patients. Although the number of patients included in these trials was relatively small, these results imply a distinct beneficial function for HGF over other angiogenic growth factors in a clinical setting. AREAS COVERED: In this review, data from Phase I-III clinical trials of gene therapy for patients with peripheral artery disease (PAD) are examined. In addition, the potential mechanisms behind the success or failure of clinical trials are discussed. EXPERT OPINION: Compared with VEGF and FGF, HGF has a unique molecular effect on inflammation, fibrosis and cell senescence under pathological conditions. These features may explain the clinical benefits of HGF in PAD patients.


Asunto(s)
Terapia Genética/métodos , Enfermedad Arterial Periférica/genética , Enfermedad Arterial Periférica/terapia , Animales , Ensayos Clínicos como Asunto/métodos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/genética , Factor de Crecimiento de Hepatocito/administración & dosificación , Factor de Crecimiento de Hepatocito/genética , Humanos , Isquemia/genética , Isquemia/terapia , Calidad de Vida
10.
Hypertens Res ; 37(6): 500-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24621470

RESUMEN

The paracrine loop involving inflammatory cytokines between adipocytes and macrophages establishes a vicious cycle that aggravates pro-inflammatory changes in adipose tissue. The serum level of hepatocyte growth factor (HGF) is increased in metabolic syndrome, but whether HGF is beneficial or detrimental in inflammatory conditions is unclear. We previously reported that HGF has strong anti-inflammatory effects. We therefore hypothesized that HGF may inhibit chronic inflammation in adipose tissue by inhibiting the vicious cycle between adipocytes and macrophages. We stimulated the macrophage cell line RAW264 with HGF and evaluated pro-inflammatory cytokines. Coculturing differentiated 3T3-L1 adipocytes with RAW264 results in marked upregulation of pro-inflammatory cytokines. We examined whether HGF suppresses the upregulation of pro-inflammatory cytokines in this coculture system. Cardiac-specific HGF transgenic mice (HGF-Tg) were crossed with ApoE KO (knockout) mice, to yield ApoE KO/HGF-Tg mice, which were treated with a high-fat diet (HFD) and received angiotensin (Ang) II. The phenotypes of ApoE KO and ApoE KO/HGF-Tg mice were compared. Treatment with HGF reduced the expression of pro-inflammatory cytokine genes (Tumor necrosis factor-α, monocyte chemoattractant protein-1 and interleukin-6) in RAW264 and the coculture system. The anti-inflammatory effects of HGF were also confirmed in in vivo studies. Macrophage infiltration in epididymal adipose and fatty liver was reduced in ApoE KO/HGF-Tg. Adipocyte diameter was reduced in ApoE KO/HGF-Tg, and the serum adiponectin level was upregulated. These beneficial effects in ApoE KO/HGF-Tg mice under HFD and Ang II infusion were abrogated by an anti-HGF neutralizing antibody. These results suggest that HGF inhibits the vicious cycle of adipocytes and macrophages through the inhibition of macrophage-mediated pro-inflammatory cytokines and upregulation of adiponectin in adipocytes. These favorable effects may suppress chronic inflammation in adipose tissue.


Asunto(s)
Adipocitos/efectos de los fármacos , Antiinflamatorios/farmacología , Citocinas/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Macrófagos/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/fisiopatología , Técnicas In Vitro , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo
11.
Hypertension ; 59(2): 308-16, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22252391

RESUMEN

Angiotensin (Ang) II type 1 receptor blockers have demonstrated beneficial effects beyond blood pressure control in the treatment of chronic kidney disease. There is clinical evidence that telmisartan is more effective than losartan in reducing proteinuria in hypertensive patients with diabetic nephropathy, because it is a partial agonist of peroxisome-proliferator activated receptor-γ (PPARγ), as well as an Ang II type 1 receptor blocker (AMADEO Study [A comparison of telMisartan versus losArtan in hypertensive type 2 DiabEtic patients with Overt nephropathy]). In this study, we examined the role of PPARγ activation in the renal protective actions of telmisartan using Ang II type 1 receptor-deficient mice. Renal injury was induced in Ang II type 1 receptor-deficient mice by producing unilateral ureteral obstruction, which exhibited severe renal interstitial fibrosis and inflammation. In these mice, telmisartan prevented hydronephrosis induced by unilateral ureteral obstruction more strongly than did losartan. Importantly, the prevention of renal atrophy and fibrosis by telmisartan was significantly attenuated by GW9662, a PPARγ antagonist. Interestingly, the downstream effector of PPARγ activation by telmisartan is hepatocyte growth factor (HGF), a well-known antifibrotic factor, because renal HGF expression was significantly increased by telmisartan, and a neutralizing antibody against HGF diminished the renal protective action of telmisartan. These beneficial changes by telmisartan were associated with a decrease in the expression of transforming growth factor-ß1 and other proinflammatory and profibrotic cytokine genes through PPARγ/HGF activation. Our findings provide evidence of organ protective actions of telmisartan through the PPARγ/HGF pathway, independent of Ang II type 1 receptor blockade. Further development of the next generation of Ang II type 1 receptor blockers with added organ protective actions, such as PPARγ activation, might provide new beneficial drugs to treat renal and cardiovascular diseases.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Bencimidazoles/uso terapéutico , Benzoatos/uso terapéutico , Factor de Crecimiento de Hepatocito/fisiología , Hidronefrosis/prevención & control , Hidronefrosis/fisiopatología , PPAR gamma/fisiología , Receptor de Angiotensina Tipo 1/deficiencia , Transducción de Señal/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Anilidas/farmacología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Factor de Crecimiento de Hepatocito/inmunología , Hidronefrosis/etiología , Riñón/efectos de los fármacos , Riñón/fisiopatología , Losartán/farmacología , Losartán/uso terapéutico , Masculino , Ratones , Ratones Noqueados , PPAR gamma/efectos de los fármacos , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/fisiología , Transducción de Señal/efectos de los fármacos , Telmisartán , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/fisiología , Obstrucción Ureteral/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA