Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Clin Cardiol ; 44(3): 386-391, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33595868

RESUMO

BACKGROUND: Current guidelines recommend interventional closure of patent foramen ovale (PFO) in patients with cryptogenic ischemic stroke who are under 60 years of age. HYPOTHESIS: The hypothesis of this study was to compare follow-up results of PFO closure in patients over 60 years of age to those of patients under 60 years of age in order to determine whether the procedure is safe and effective for both age groups. METHODS: We included 293 patients who had a cryptogenic ischemic stroke and a PFO confirmed by transesophageal echocardiography (TEE) and who were scheduled for percutaneous closure of the PFO between 2014 and 2019. The device implantation was completed in all patients using an Amplatzer™, Occlutec™, or Cardia Ultrasept PFO occluder. RESULTS: Follow-up TEE examinations were performed at intervals of 1, 3, and 6 months after implantation. Patients were followed for a median of 3.6 ± 1.2 years. Recurrent ischemic stroke or transient ischemic attack, cardiac death, arrhythmias, and residual shunt were reported equally in both groups. CONCLUSIONS: Interventional closure of PFO can be as safe and effective in patients over 60 years of age as it is in patients under 60 years of age regardless of the device used. In this older patient group, rigorous discussion and a case-by-case decision-making process including cardiologists and neurologists is warranted to ensure optimal procedure selection.


Assuntos
Forame Oval Patente , Dispositivo para Oclusão Septal , Acidente Vascular Cerebral , Idoso , Cateterismo Cardíaco/efeitos adversos , Seguimentos , Forame Oval Patente/diagnóstico , Forame Oval Patente/diagnóstico por imagem , Humanos , Acidente Vascular Cerebral/etiologia , Resultado do Tratamento
3.
Curr Hypertens Rep ; 14(3): 183-92, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22467342

RESUMO

The possibility of carcinogenic side effects of antihypertensive therapies due to their chronic administration has been raised multiple times in the past. Recently, the issue has again drawn attention, this time in relation to angiotensin receptor blockers (ARBs). This, among others, caused both American and European drug regulation authorities to review the underlying evidence concerning the relationship between this class of medications and potential adverse carcinogenic outcome. A plethora of both basic science and preclinical evidence has been generated, and three meta-analyses and one nationwide cohort have focused on this specific question. The current review aims to summarize the contemporary multidisciplinary evidence on whether ARBs may be associated with an increased risk of tumorigenesis.


Assuntos
Antagonistas de Receptores de Angiotensina/efeitos adversos , Anti-Hipertensivos/efeitos adversos , Neoplasias/induzido quimicamente , Transformação Celular Neoplásica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/induzido quimicamente , Fatores de Risco
4.
Cardiovasc Res ; 91(3): 510-8, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21478266

RESUMO

AIMS: Monocytes contribute to arteriogenesis by infiltration to sites of collateral growth and subsequent production and release of growth factors. Transforming growth factor ß1 (TGF-ß1) mediates monocyte motility and stimulates arteriogenesis. TGF-ß1 signalling mechanisms mediating monocyte motility are unknown so far. Moreover, the influence of cardiovascular risk factor diabetes on TGF-ß1-induced monocyte migration remains to be elucidated. METHODS AND RESULTS: Stimulation of primary human monocytes with TGF-ß1 endorsed phosphorylation of v-Akt murine thymoma viral oncogene analogues protein (AKT), p38, and extracellular signal-related kinase 1/2 (ERK1/2), besides the activation of the SMA/MAD homologues protein (SMAD) pathway. Inhibition of the TGF-ßtype 1 receptor, alias activin receptor-like kinase 5 (ALK5), hindered monocyte chemotaxis towards TGF-ß1 and TGF-ß1-activated downstream signalling cascades. Individual genetic knock-downs for receptor-regulated SMAD2 and SMAD3 did not affect monocyte migration to TGF-ß1. Inhibition of phosphoinositide 3 kinase (PI3K) activity, but not AKT, diminished both basal and TGF-ß1-mediated monocyte motility. TGF-ß1-induced monocyte chemotaxis did not rely on ERK1/2, but rather on p38. Remarkably, TGF-ß1 was able to stimulate chemotaxis of diabetic monocytes. CONCLUSION: The current study provides novel insights into the molecular mechanisms of TGF-ß1-induced monocyte migration, requiring ALK5 kinase activity and signalling via PI3K and p38. TGF-ß1-driven monocyte motogenicity is fully functional in diabetic conditions, which is in sharp contrast to the impaired chemotactic responses to certain other arteriogenic cytokines. Therefore, TGF-ß1 may be a promising candidate for endogenously and exogenously stimulating collateral growth in diabetic patients.


Assuntos
Quimiotaxia de Leucócito , Diabetes Mellitus Tipo 2/enzimologia , Monócitos/enzimologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Idoso , Estudos de Casos e Controles , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Diabetes Mellitus Tipo 2/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Países Baixos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptor do Fator de Crescimento Transformador beta Tipo I , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
5.
Arterioscler Thromb Vasc Biol ; 30(11): 2188-95, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20814017

RESUMO

OBJECTIVE: To elucidate the downstream mechanisms of vascular endothelial growth factor receptor 2 (VEGFR2), a key receptor in angiogenesis, which has been associated with atherosclerotic plaque growth and instability. METHODS AND RESULTS: By using a yeast-2-hybrid assay, we identified A Disintegrin And Metalloprotease 10 (ADAM10) as a novel binding partner of VEGFR2. ADAM10 is a metalloprotease with sheddase activity involved in cell migration; however, its exact function in endothelial cells (ECs), angiogenesis, and atherosclerosis is largely unknown. For the first time to our knowledge, we show ADAM10 expression in human atherosclerotic lesions, associated with plaque progression and neovascularization. We demonstrate ADAM10 expression and activity in ECs to be induced by VEGF; also, ADAM10 mediates the ectodomain shedding of VEGFR2. Furthermore, VEGF induces ADAM10-mediated cleavage of vascular endothelium (VE)-cadherin, which could increase vascular permeability and facilitate EC migration. Indeed, VEGF increases vascular permeability in an ADAM10- and ADAM17-dependent way; inhibition of ADAM10 reduces EC migration and chemotaxis. CONCLUSIONS: These data provide the first evidence of ADAM10 expression in atherosclerosis and neovascularization. ADAM10 plays a functional role in VEGF-induced EC function. These data open perspectives for novel therapeutic interventions in vascular diseases.


Assuntos
Proteínas ADAM/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Aterosclerose/fisiopatologia , Células Endoteliais/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Patológica/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteínas ADAM/biossíntese , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/biossíntese , Aterosclerose/metabolismo , Células Cultivadas , Progressão da Doença , Humanos , Proteínas de Membrana/biossíntese , Neovascularização Patológica/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia
6.
Circulation ; 120(2): 150-9, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19564559

RESUMO

BACKGROUND: Monocytes are cellular components of wound repair, arteriogenesis, and atherogenesis. Vascular endothelial growth factor (VEGF)-A and placental growth factor recruit monocytes to sites of arteriogenesis via stimulation of VEGF receptor-1 (VEGFR-1). The chemotactic response of monocytes to VEGF-A is attenuated in individuals with diabetes mellitus (DM). This VEGF resistance correlates with impaired collateral growth. The aim of this study is to elucidate the molecular basis of VEGF resistance and impaired monocyte response in DM. METHODS AND RESULTS: Phosphorylation of Akt, p38, and extracellular signal-regulated kinase 1/2 (ERK1/2) could be stimulated with either placental growth factor-1 or VEGF-A in monocytes from non-DM but not DM individuals. In contrast, formyl-methionyl-leucyl-phenylalanine caused a comparable activation of these molecules in both DM and non-DM monocytes. Baseline phosphorylation of Akt, p38, and ERK1/2 was significantly elevated in monocytes from DM compared with non-DM subjects. Of note, H(2)O(2) activated Akt, p38, and ERK1/2 in non-DM monocytes ex vivo. Protein tyrosine phosphatases had stronger oxidative modifications in monocytes from DM than from non-DM individuals, which reflects functional protein tyrosine phosphatase inhibition, similar to that seen after H(2)O(2) challenge. Overall, protein tyrosine phosphatase and protein tyrosine phosphatase-1B activity were reduced in DM monocytes. DM monocytes revealed higher expression of the receptor for advanced glycation end products. Stimulation with advanced glycation end products ligands resulted in activation of non-DM monocytes and inhibition of VEGFR-1-mediated chemotaxis. The elevated baseline phosphorylation/activation of Akt, p38, and ERK1/2 in DM monocytes likely causes the resistance to further stimulation with specific stimuli such as VEGF-A, revealing a molecular explanation of the DM-related signal transduction defect. CONCLUSIONS: We propose that elevated advanced glycation end products expression and increased oxidative stress in diabetic monocytes lead to activation of VEGFR-1-related signaling pathways and to desensitization of VEGFR-1 responses. These data establish VEGF resistance as a novel molecular concept for DM-related cellular dysfunction.


Assuntos
Diabetes Mellitus/fisiopatologia , Monócitos/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Idoso , Artérias/fisiopatologia , Estudos de Casos e Controles , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neovascularização Patológica/fisiopatologia , Estresse Oxidativo/fisiologia , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Cancer Res ; 68(18): 7342-51, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794121

RESUMO

Several lines of evidence indicate that Flt-1, a fms-like tyrosine kinase receptor, which binds to vascular endothelial growth factor (VEGF)-A, VEGF-B, and PlGF, is a positive regulator of angiogenesis in the context of tumor growth and metastasis. However, the molecular basis of its action is still not clear. Besides endothelial cells, Flt-1 is also expressed by other different cell types, including myeloid hematopoeitic cells (monocytes and macrophages). To examine the functions of Flt-1 expressed by bone marrow-derived myeloid cells in supporting tumor growth and angiogenesis, Flt-1 tyrosine kinase-deficient (Flt-1 TK-/-) bone marrow cells were transplanted into lethally irradiated syngeneic recipients. After hematopoietic reconstitution, we orthotopically implanted syngeneic wild-type glioma cells or glioma cells overexpressing either VEGF(164) or PlGF-2. Loss of Flt-1 signaling in bone marrow-derived myeloid cells led to a significant decrease in tumor volume and vascularization in gliomas. VEGF but not PlGF overexpressed by glioma cells restored the tumor growth rate in Flt-1 TK-/- bone marrow chimera. VEGF and PlGF overexpression by tumor cells induced an accumulation of bone marrow-derived myeloid cells into tumor tissue. This infiltration was decreased in tumors grown in Flt-1 TK-/- bone marrow chimeras. When investigating chemokines and growth factors involved in myeloid cell recruitment, we determined elevated SDF-1/CXCL12 levels in VEGF- and PlGF-overexpressing tumors. Collectively, these results suggest that Flt-1 signaling in myeloid cells is essential to amplify the angiogenic response and to promote glioma growth.


Assuntos
Glioma/enzimologia , Glioma/patologia , Macrófagos/enzimologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/patologia , Permeabilidade Capilar/fisiologia , Processos de Crescimento Celular/fisiologia , Quimiocina CXCL12/biossíntese , Quimera/metabolismo , Glioma/irrigação sanguínea , Glioma/metabolismo , Macrófagos/patologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/patologia , Neovascularização Patológica/enzimologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteínas/genética , Proteínas/metabolismo , Transdução de Sinais , Transfecção , Fator A de Crescimento do Endotélio Vascular/biossíntese
8.
J Clin Invest ; 118(8): 2979-85, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18654662

RESUMO

Novel biomarkers, such as circulating (auto)antibody signatures, may improve early detection and treatment of ruptured atherosclerotic lesions and accompanying cardiovascular events, such as myocardial infarction. Using a phage-display library derived from cDNAs preferentially expressed in ruptured peripheral human atherosclerotic plaques, we performed serological antigen selection to isolate displayed cDNA products specifically interacting with antibodies in sera from patients with proven ruptured peripheral atherosclerotic lesions. Two cDNA products were subsequently evaluated on a validation series of patients with peripheral atherosclerotic lesions, healthy controls, and patients with coronary artery disease at different stages. Our biomarker set was able to discriminate between patients with peripheral ruptured lesions and patients with peripheral stable plaques with 100% specificity and 76% sensitivity. Furthermore, 93% of patients with an acute myocardial infarction (AMI) tested positive for our biomarkers, whereas all patients with stable angina pectoris tested negative. Moreover, 90% of AMI patients who initially tested negative for troponin T, for which a positive result is known to indicate myocardial infarction, tested positive for our biomarkers upon hospital admission. In conclusion, antibody profiling constitutes a promising approach for noninvasive diagnosis of atherosclerotic lesions, because a positive serum response against a set of 2 cDNA products showed a strong association with the presence of ruptured peripheral atherosclerotic lesions and myocardial infarction.


Assuntos
Aterosclerose/diagnóstico , Aterosclerose/patologia , Autoanticorpos , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/patologia , Idoso , Angina Pectoris/sangue , Angina Pectoris/patologia , Antígenos/imunologia , Aterosclerose/sangue , Autoanticorpos/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Estudos de Coortes , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/patologia , Estudos Transversais , DNA Complementar/isolamento & purificação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Biblioteca de Peptídeos , Doenças Vasculares Periféricas/sangue , Doenças Vasculares Periféricas/patologia , Reprodutibilidade dos Testes , Ruptura Espontânea/patologia , Sensibilidade e Especificidade
9.
Arterioscler Thromb Vasc Biol ; 28(2): 322-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18079407

RESUMO

OBJECTIVE: Arteriogenesis, the growth of preexisting arterioles into functional arteries, is dependent on the proper function of monocytes. Likewise, wound healing is monocyte-dependent. The activation of vascular endothelial growth factor receptor-1 (VEGFR-1) in monocytes induces a chemotactic response, triggers the expression of tissue factor, and gene expression of cytokines and chemokines. Little is known about intracellular signaling pathways mediating the biological functions triggered by VEGFR-1 in primary monocytes. METHODS AND RESULTS: Monocytes were isolated from peripheral venous blood of young healthy individuals using indirect magnetic labeling. Stimulation of monocytes with either vascular endothelial growth factor-A (VEGF-A) or placenta growth factor (PlGF-1) triggered VEGFR-1 autophosphorylation and phosphorylation of distinct downstream proteins: phosphatidylinositol-3 kinase (PI-3K), Akt, p38, and extracellular signal-regulated kinase-1/2 (ERK1/2). PI-3K appears to be a central regulator in VEGFR-1 signaling in monocytes as the activation of Akt, p38, and ERK1/2 are PI-3-K-dependent. In addition, Akt activation functions downstream of p38 kinase. VEGFR-1-mediated chemotaxis of monocytes is dependent on the activation of PI-3K, p38 kinase, Akt, and ERK1/2, when assessed in a modified Boyden chamber. CONCLUSIONS: Both PlGF-1 and VEGF-A can activate VEGFR-1-dependent signaling pathways in primary human monocytes, leading to the activation of several intracellular signaling pathways. These pathways are critically involved in primary monocyte chemotaxis.


Assuntos
Monócitos/fisiologia , Proteínas da Gravidez/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Células Cultivadas , Quimiotaxia/fisiologia , Humanos , Fosforilação , Fator de Crescimento Placentário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...