Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
Circ Res ; 107(11): 1326-35, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20930149

RESUMO

RATIONALE: The neuropeptide catestatin is an endogenous nicotinic cholinergic antagonist that acts as a pleiotropic hormone. OBJECTIVE: Catestatin shares several functions with angiogenic factors. We therefore reasoned that catestatin induces growth of new blood vessels. METHODS AND RESULTS: Catestatin induced migration, proliferation, and antiapoptosis in endothelial cells and exerted capillary tube formation in vitro in a Matrigel assay, and such effects were mediated via G protein, mitogen-activated protein kinase, and Akt. Catestatin-induced endothelial cell functions are further mediated by basic fibroblast growth factor, as shown by blockade of effects by a neutralizing fibroblast growth factor antibody. Furthermore, catestatin released basic fibroblast growth factor from endothelial cells and stimulated fibroblast growth factor signaling. In addition to its function on endothelial cells, catestatin also exerted effects on endothelial progenitor cells and vascular smooth muscle cells. In vivo, catestatin induced angiogenesis in the mouse cornea neovascularization assay and increased blood perfusion and number of capillaries in the hindlimb ischemia model. In addition to angiogenesis, catestatin increased density of arterioles/arteries and incorporation of endothelial progenitor cells in the hindlimb ischemia model, indicating induction of arteriogenesis and postnatal vasculogenesis. CONCLUSION: We conclude that catestatin acts as a novel angiogenic cytokine via a basic fibroblast growth factor-dependent mechanism.


Assuntos
Proteínas Angiogênicas/fisiologia , Cromogranina A/fisiologia , Citocinas/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Fisiológica/fisiologia , Neuropeptídeos/fisiologia , Fragmentos de Peptídeos/fisiologia , Animais , Movimento Celular/fisiologia , Células Cultivadas , Endotélio Vascular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Circ Res ; 105(10): 994-1002, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19797703

RESUMO

RATIONALE: The neuropeptide secretoneurin induces angiogenesis and postnatal vasculogenesis and is upregulated by hypoxia in skeletal muscle cells. OBJECTIVE: We sought to investigate the effects of secretoneurin on therapeutic angiogenesis. METHODS AND RESULTS: We generated a secretoneurin gene therapy vector. In the mouse hindlimb ischemia model secretoneurin gene therapy by intramuscular plasmid injection significantly increased secretoneurin content of injected muscles, improved functional parameters, reduced tissue necrosis, and restored blood perfusion. Increased muscular density of capillaries and arterioles/arteries demonstrates the capability of secretoneurin gene therapy to induce therapeutic angiogenesis and arteriogenesis. Furthermore, recruitment of endothelial progenitor cells was enhanced by secretoneurin gene therapy consistent with induction of postnatal vasculogenesis. Additionally, secretoneurin was able to activate nitric oxide synthase in endothelial cells and inhibition of nitric oxide inhibited secretoneurin-induced effects on chemotaxis and capillary tube formation in vitro. In vivo, secretoneurin induced nitric oxide production and inhibition of nitric oxide attenuated secretoneurin-induced effects on blood perfusion, angiogenesis, arteriogenesis, and vasculogenesis. Secretoneurin also induced upregulation of basic fibroblast growth factor and platelet-derived growth factor-B in endothelial cells. CONCLUSIONS: In summary, our data indicate that gene therapy with secretoneurin induces therapeutic angiogenesis, arteriogenesis, and vasculogenesis in the hindlimb ischemia model by a nitric oxide-dependent mechanism.


Assuntos
Indutores da Angiogênese/metabolismo , Citocinas/biossíntese , Terapia Genética , Isquemia/terapia , Neovascularização Fisiológica , Neuropeptídeos/biossíntese , Óxido Nítrico/metabolismo , Secretogranina II/biossíntese , Animais , Citocinas/genética , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Humanos , Isquemia/genética , Isquemia/metabolismo , Camundongos , Neuropeptídeos/genética , Óxido Nítrico Sintase Tipo III/biossíntese , Secretogranina II/genética , Células-Tronco/metabolismo
4.
Ann Neurol ; 65(4): 386-93, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19399887

RESUMO

OBJECTIVE: Randomized, blinded trial of intramuscular gene transfer using plasmid vascular endothelial growth factor (VEGF) to treat diabetic polyneuropathy. METHODS: Diabetic patients with polyneuropathy were randomized to receive a VEGF-to-placebo ratio of 3:1. Three sets of injections were given at eight standardized sites adjacent to the sciatic, peroneal, and tibial nerves of one leg. Primary outcomes were change in symptom score at 6 months and a prespecified overall clinical and electrophysiological improvement score. Secondary outcomes were differences in symptoms, examination scores, visual analog pain scale, nerve conduction, and quantitative sensory testing. RESULTS: Thirty-nine patients received plasmid VEGF and 11 received placebo. Mean symptom score improved in both legs at 6 months, favoring VEGF over placebo (-1.2 +/- 0.5 vs -0.9 +/- 0.5; p < 0.01 after adjustment for change in the untreated leg) and compared with the untreated leg (-0.7 +/- 0.5; p = 0.02). The region of sensory loss and visual analog pain scale improved in the treated group (-1.5 vs -0.5; p = 0.01). Twelve of 39 VEGF versus 2 of 11 placebo patients met criterion for overall improvement. Other measures including nerve conduction potentials did not improve. There were 84 adverse events in VEGF patients, and 22 were serious; there were 51 events in placebo patients, and 2 were serious. INTERPRETATION: Intramuscular plasmid VEGF gene transfer improved diabetic neuropathic symptoms, meeting primary end-point criteria for efficacy but not affecting most secondary measures. Treatment was associated with more serious adverse events that did not reach statistical significance. These results are not conclusive but may justify further clinical study.


Assuntos
Neuropatias Diabéticas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Idoso , Estudos de Coortes , Neuropatias Diabéticas/metabolismo , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Condução Nervosa/genética , Medição da Dor , Nervos Periféricos/fisiopatologia , Índice de Gravidade de Doença , Resultado do Tratamento
5.
J Clin Med ; 8(8)2019 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-31382583

RESUMO

Belatacept is an attractive option for immunosuppression after renal transplantation. Renal allograft function is superior when compared to calcineurin inhibitor (CNI) based therapy in "de novo" treated patients and it has also been proposed that individuals at high cardiovascular (CV) risk may benefit most. In this retrospective cohort study, we assessed the efficacy and safety of treating patients at high cardiovascular risk with Belatacept (n = 34, for 1194 observation months) when compared to a matched control group of 150 individuals under CNI immunosuppression (for 7309 months of observation). The estimated glomerular filtration rate (eGFR) increased for patients taking Belatacept but decreased during CNI-based therapy (+2.60 vs. -0.89 mL/min/1.73 m2/year, p = 0.006). In a multivariate Cox regression model, Belatacept remained the only significant factor associated with the improvement of eGFR (HR 4.35, 95%CI 2.39-7.93). Belatacept treatment was not a significant risk factor for renal allograft rejection or graft loss. In terms of safety, the only significant risk factor for de novo cardiovascular events was a pre-existing cerebrovascular disease, but Belatacept was not associated with a significant risk reduction. Belatacept treatment was not associated with an increased risk of severe infections, cytomegalo virus (CMV) or BK-virus reactivation, malignancy or death in the multivariate Cox regression analysis. Belatacept is an efficient and safe option for patients after renal transplantation at high cardiovascular risk.

6.
FASEB J ; 21(11): 2906-17, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17504977

RESUMO

Expression of angiogenic cytokines like vascular endothelial growth factor is enhanced by hypoxia. We tested the hypothesis that decreased oxygen levels up-regulate the angiogenic factor secretoneurin. In vivo, muscle cells of mouse ischemic hind limbs showed increased secretoneurin expression, and inhibition of secretoneurin by a neutralizing antibody impaired the angiogenic response in this ischemia model. In a mouse soft tissue model of hypoxia, secretoneurin was increased in subcutaneous muscle fibers. In vitro, secretoneurin mRNA and protein were up-regulated in L6 myoblast cells after exposure to low oxygen levels. The hypoxia-dependent regulation of secretoneurin was tissue specific and was not observed in endothelial cells, vascular smooth muscle cells, or AtT20 pituitary tumor cells. The hypoxia-dependent induction of secretoneurin in L6 myoblasts is regulated by hypoxia-inducible factor-1alpha, since inhibition of this factor using si-RNA inhibited up-regulation of secretoneurin. Induction of secretoneurin by hypoxia was dependent on basic fibroblast growth factor in vivo and in vitro, and inhibition of this regulation by heparinase suggests an involvement of low-affinity basic fibroblast growth factor binding sites. In summary, our data show that the angiogenic cytokine secretoneurin is up-regulated by hypoxia in muscle cells by hypoxia-inducible factor-1alpha- and basic fibroblast growth factor-dependent mechanisms.


Assuntos
Hipóxia Celular , Fator 2 de Crescimento de Fibroblastos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mioblastos/metabolismo , Neuropeptídeos/metabolismo , Secretogranina II/metabolismo , Transdução de Sinais , Animais , Western Blotting , Células Cultivadas , Primers do DNA/química , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Extremidades/cirurgia , Imunofluorescência , Isquemia/metabolismo , Isquemia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/irrigação sanguínea , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , NAD/metabolismo , Neovascularização Fisiológica , Neoplasias Hipofisárias/irrigação sanguínea , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Reação em Cadeia da Polimerase , Pró-Proteína Convertases/metabolismo , RNA Interferente Pequeno/farmacologia , Radioimunoensaio , Ratos , Pele/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Eur J Pharmacol ; 598(1-3): 104-11, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-18834877

RESUMO

Several members of the neuropeptide family exert chemotactic actions on blood monocytes consistent with neurogenic inflammation. Furthermore, chromogranin A (CgA) containing Alzheimer plaques are characterized by extensive microglia activation and such activation induces neuronal damage. We therefore hypothesized that the catecholamine release inhibitory peptide catestatin (hCgA(352-372)) would induce directed monocyte migration. We demonstrate that catestatin dose-dependently stimulates chemotaxis of human peripheral blood monocytes, exhibiting its maximal effect at a concentration of 1 nM comparable to the established chemoattractant formylated peptide Met-Leu-Phe (fMLP). The naturally occurring catestatin variants differed in their chemotactic property insofar as that the Pro370Leu variant was even more potent than wild type, whereas the Gly364Ser variant was less effective. Specificity of this effect was shown by inhibition of catestatin-induced chemotaxis by a specific neutralizing antibody. In addition, catestatin mediated effect was blocked by dimethylsphingosine and treatment with endothelial differentiation gene (Edg)-1 and Edg-3 antisense RNA as well as by incubation with pertussis toxin and genistein indicating involvement of tyrosine kinase receptor-, G-protein- and sphingosine-1-phosphate signaling. Catestatin also stimulated Akt- and extracellular signal related kinase (ERK)-phosphorylation and catestatin-induced chemotaxis was blocked by blockers of phosphoinositide-3 (PI-3) kinase and nitric oxide as well as by inhibition of the mitogen-activated protein kinases (MAPK) system indicating involvement of these signal transduction pathways. In summary, our data indicate that catestatin induces monocyte chemotaxis by activation of a variety of signal transduction pathways suggesting a role of this peptide as an inflammatory cytokine.


Assuntos
Cromogranina A/farmacologia , Monócitos/fisiologia , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Movimento Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Cromogranina A/genética , Ativação Enzimática/efeitos dos fármacos , Genisteína/farmacologia , Humanos , Infiltração de Neutrófilos/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Toxina Pertussis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Transfecção
8.
Mol Ther ; 15(1): 69-75, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164777

RESUMO

Toxic neuropathy represents an important clinical problem in the use of the chemotherapeutic substances Taxol and thalidomide. Sensory neuropathy has a high incidence, lacks an effective treatment and is the dose-limiting factor for these drugs. The pathogenic basis of these neuropathies is unknown. We investigated the hypothesis that the experimental toxic neuropathies from Taxol and thalidomide results from destruction of vasa nervorum and can be reversed by the administration of an angiogenic cytokine. In animal models of Taxol- and thalidomide-induced neuropathy, nerve blood flow has been attenuated and the number of vasa nervorum has been reduced. Intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 administered in parallel with Taxol injections completely inhibited deterioration of nerve function and diminution of the peripheral nerve vasculature. Gene therapy in animals with established Taxol- or thalidomide-induced neuropathies resulted in recovery of vascularity and improved nerve electrophysiology. These findings implicate microvascular damage as the basis for toxic neuropathy and suggest that angiogenic growth factors may constitute a novel treatment for this disorder.


Assuntos
Neovascularização Patológica/tratamento farmacológico , Paclitaxel/farmacologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Talidomida/farmacologia , Vasa Nervorum/efeitos dos fármacos , Vasa Nervorum/lesões , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Quimioterapia Combinada , Eletrofisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Humanos , Injeções Intramusculares , Lectinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Circulação Renal
9.
Circulation ; 111(20): 2662-70, 2005 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15897348

RESUMO

BACKGROUND: Toxic neuropathies induced by cisplatin and other chemotherapeutic agents are important clinical problems because of their high incidence, their lack of effective treatment, and the fact that neuropathy represents a dose-limiting factor for these therapies. The pathogenic basis for toxic neuropathies induced by chemotherapeutic agents has not been completely elucidated. METHODS AND RESULTS: We investigated the hypothesis that experimental toxic neuropathy results from an antiangiogenic effect of these drugs, resulting in destruction of the vasa nervorum, and accordingly that the neuropathy could be prevented or reversed by locally administered VEGF gene transfer without augmenting tumor growth. In an animal model of cisplatin-induced neuropathy, nerve blood flow was markedly attenuated, and there was a profound reduction in the number of vasa nervorum associated with marked endothelial cell apoptosis, resulting in a severe peripheral neuropathy with focal axonal degeneration characteristic of ischemic neuropathy. After intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 in animals with an established neuropathy, vascularity and blood flow returned to levels similar to those of control rats, peripheral nerve function was restored, and histological nerve architecture was normalized. Gene therapy administered in parallel with cisplatin chemotherapy completely attenuated endothelial cell apoptosis and inhibited destruction of nerve vasculature, deterioration of nerve function, and axonal degeneration. In a rat tumor model, VEGF gene transfer administered locally did not alter tumor growth or vascularity. CONCLUSIONS: These findings implicate microvascular damage as the basis for toxic neuropathy induced by cisplatin and suggest that local angiogenic gene therapy may constitute a novel prevention or treatment for this disorder without augmenting tumor growth or vascularization.


Assuntos
Cisplatino/efeitos adversos , Doenças do Sistema Nervoso Periférico/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Animais , Apoptose , Endotélio Vascular/citologia , Terapia Genética , Humanos , Masculino , Neoplasias/irrigação sanguínea , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/terapia , Ratos , Ratos Sprague-Dawley , Carga Tumoral/efeitos dos fármacos , Veias Umbilicais/citologia , Vasa Nervorum/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
10.
Circulation ; 109(6): 777-83, 2004 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-14970115

RESUMO

BACKGROUND: Secretoneurin is an abundant neuropeptide of the central, peripheral, and autonomic nervous systems, located in nerve fibers characterized by a close interaction with blood vessels and known to stimulate endothelial cell migration. METHODS AND RESULTS: We hypothesized that secretoneurin might act as an angiogenic cytokine and tested for these effects in vivo using a mouse cornea neovascularization model and in vitro by assessing capillary tube formation in a matrigel assay. In vivo, secretoneurin-induced neovasculature is characterized by a distinct pattern of arterial and venous vessels of large diameter and length. Immunohistochemical staining for CD-31 revealed endothelial lining of the inner surface of these vessels, and recruitment of alpha-smooth muscle actin-positive perivascular cells suggests vessel maturation. In vitro, secretoneurin-induced capillary tube formation was dose dependent and specific, confirming that effects of secretoneurin occur directly on endothelial cells. Secretoneurin also stimulated proliferation and exerted antiapoptotic effects on endothelial cells and activated intracellular phosphatidylinositol 3' kinase/Akt and mitogen-activated protein kinase pathways, as demonstrated by increased phosphorylation of Akt and extracellular signal-regulated kinase. CONCLUSIONS: These data show that secretoneurin represents a novel direct angiogenic cytokine and reiterate the coordinated relationship between nervous and vascular systems.


Assuntos
Indutores da Angiogênese/farmacologia , Neuropeptídeos/farmacologia , Proteínas Serina-Treonina Quinases , Animais , Apoptose/efeitos dos fármacos , Divisão Celular , Células Cultivadas , Córnea/anatomia & histologia , Córnea/irrigação sanguínea , Citocinas/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Secretogranina II , Transdução de Sinais
11.
Circulation ; 110(9): 1121-7, 2004 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-15326074

RESUMO

BACKGROUND: Induction of postnatal vasculogenesis, the mobilization of bone marrow-derived endothelial progenitor cells and incorporation of these cells into sites of blood vessel formation, is a well-known feature of angiogenic cytokines such as vascular endothelial growth factor. We hypothesized that the angiogenic neuropeptide secretoneurin induces this kind of neovascularization. METHODS AND RESULTS: Secretoneurin induced mobilization of endothelial progenitor cells to sites of vasculogenesis in vivo in the cornea neovascularization assay. Progenitor cells were incorporated into vascular structures or were located adjacent to them. Systemic injection of secretoneurin led to increase of circulating stem cells and endothelial progenitor cells. In vitro secretoneurin induced migration, exerted antiapoptotic effects, and increased the number of these cells. Furthermore, secretoneurin stimulated the mitogen-activated protein kinase system, as shown by phosphorylation of extracellular signal-regulated kinase, and activated the protein kinase B/Akt pathway. Activation of mitogen-activated protein kinase was necessary for increase of cell number and migration, whereas Akt seemed to play a role in migration of endothelial progenitor cells. CONCLUSIONS: These data show that the angiogenic neuropeptide secretoneurin stimulates postnatal vasculogenesis by mobilization, migration, and incorporation of endothelial progenitor cells.


Assuntos
Neovascularização da Córnea , Neuropeptídeos/fisiologia , Androstadienos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Transplante de Medula Óssea , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Neovascularização da Córnea/tratamento farmacológico , Flavonoides/farmacologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Óperon Lac , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Neuropeptídeos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Quimera por Radiação , Ratos , Receptor TIE-2/genética , Secretogranina II , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Wortmanina
12.
Curr Pharm Des ; 9(13): 1041-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12678856

RESUMO

Cardiovascular atherosclerotic diseases remain leading causes of morbidity and mortality in the world. Despite the significant progress that has been made in the management of these diseases using medical, surgical and percutaneous therapies over the last three decades, there remains a significant population of patients who are not optimal candidates for surgical or percutaneous revascularization. Substantial research has focused on the administration of angiogenic growth factors, either as recombinant protein or by gene transfer, to promote the development of supplemental collateral blood vessels that will constitute endogenous bypass conduits around occluded native arteries; a strategy termed "therapeutic angiogenesis". While many cytokines have angiogenic activity, the best studied both in animal models and clinical trials are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). This review will discuss gene transfer strategies for therapeutic angiogenesis in critical limb and myocardial ischemia.


Assuntos
Inibidores da Angiogênese/genética , Inibidores da Angiogênese/uso terapêutico , Arteriopatias Oclusivas/fisiopatologia , Arteriopatias Oclusivas/terapia , Citocinas/genética , Citocinas/uso terapêutico , Extremidades/irrigação sanguínea , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Isquemia Miocárdica/prevenção & controle , Neovascularização Fisiológica/genética , Animais , Terapia Genética/estatística & dados numéricos , Humanos , Isquemia/fisiopatologia , Isquemia/prevenção & controle , Isquemia/terapia , Isquemia Miocárdica/fisiopatologia , Coelhos , Ratos
13.
Br J Pharmacol ; 140(4): 614-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12970083

RESUMO

The neuronal system plays a fundamental role in the maturation of primitive embryonic vascular network by providing a paracrine template for blood vessel branching and arterial differentiation. Furthermore, postnatal vascular and neural regeneration cooperate in the healing of damaged tissue. Neurogenesis continues in adulthood although confined to specific brain regions. Following ischaemic insult, neural staminal cells contribute towards the healing process through the stimulation of neurogenesis and vasculogenesis. Evidence indicates that nerves and blood vessels exert a reciprocal control of their own growth by paracrine mechanisms. For instance, guidance factors, including vascular endothelial growth factor A (VEGF-A) and semaphorins, which share the ability of binding neuropilin receptors, play a pivotal role in the tridimensional growth pattern of arterial vessels and nerves. Animal models and clinical studies have demonstrated a role of VEGF-A in the pathogenesis of ischaemic and diabetic neuropathies. Further, supplementation with VEGF-A ameliorates neuronal recovery by exerting protective effects on nerves and stimulating reparative neovascularization. Human tissue kallikrein, a recently discovered angiogenic and arteriogenic factor, accelerates neuronal recovery by stimulating the growth of vasa nervorum. Conversely, the neurotrophin nerve growth factor, known to regulate neuronal survival and differentiation, is now regarded as a stimulator of angiogenesis and arteriogenesis. These results indicate that angiogenesis and neurogenesis are paracrinally regulated by growth factors released by endothelial cells and neurons. Supplementation of these growth factors, alone or in combination, could benefit the treatment of ischaemic diseases and neuropathies.


Assuntos
Neovascularização Fisiológica/fisiologia , Fatores de Crescimento Neural/farmacologia , Neurônios/fisiologia , Comunicação Parácrina/fisiologia , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Crescimento Neural/fisiologia , Neurônios/efeitos dos fármacos
14.
Regul Pept ; 105(1): 35-46, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11853870

RESUMO

Secretoneurin (SN), a newly discovered neuropeptide, may be implicated in inflammatory responses as it was shown to modulate leukocyte, endothelial and mesenchymal cell functions. Neutrophils placed above pulmonary arterial or venous endothelial monolayers migrated through this cellular barrier in response to apical or basal stimulation with SN in a dose-dependent manner. At optimal concentrations of 10(-6) to 10(-8) M, SN was nearly equally effective in stimulating neutrophil transmigration as was tumor necrosis factor-alpha at 10 ng/ml or a chemotactic gradient of formyl-Met-Leu-Phe (10(-8) M). Stimulation of transendothelial migration appears to be specific, since a trypsin digest of SN was ineffective and excess concentrations of anti-SN antibodies completely abolished the effect. Inhibition of cyclooxygenase or nitric oxide synthase did not affect the action of SN. Preincubation of endothelial cells with pertussistoxin (PTx) or choleratoxin (CTx), and the presence of staurosporine significantly inhibited transmigration, suggesting that SN uses a signalling pathway that is coupled to G-proteins and protein kinase C in endothelium. Moreover, SN treatment resulted in transient elevation of cytoplasmatic calcium concentration in endothelial cells. These data support the hypothesis that SN might contribute to neurogenic inflammation in vivo and reveal signalling mechanisms of SN in endothelial cells.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Endotélio Vascular/citologia , Neuropeptídeos/farmacologia , Neutrófilos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Aspirina/farmacologia , Cálcio/metabolismo , Bovinos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , NG-Nitroarginina Metil Éster/farmacologia , Neuropeptídeos/genética , Neuropeptídeos/imunologia , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Inibidores de Proteases/farmacologia , Artéria Pulmonar , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Secretogranina II , Fator de Necrose Tumoral alfa/farmacologia
15.
Medicine (Baltimore) ; 93(26): e143, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25474424

RESUMO

Catastrophic antiphospholipid syndrome (CAPS) is a rare but devastating complication in patients with antiphospholipid syndrome (APS) with a high morbidity and mortality.We describe a case of a 30-year old female patient with immunoglobulin A (IgA) deficiency who underwent splenectomy because of idiopathic thrombocytopenic thrombocytopenia. Subsequently, an APS and finally systemic lupus erythematosus was diagnosed. After an uncomplicated pregnancy that was terminated by cesarean section, the patient developed severe CAPS with cerebral, myocardial, renal, and pulmonary involvement.Because of IgA deficiency, standard therapy consisting of plasmapheresis and intravenous immunoglobulins in addition to steroids was not tolerated. After 8 sessions of immunoadsorption (IAS), massive pulmonary hemorrhage was controlled but relapsed twice whenever IAS was terminated. As other immunosuppressive agents were considered dangerous because of the risk of infections in the face of severe hypogammaglobulinemia, we administered eculizumab, an inhibitor of the terminal complement pathway, which led to a persistent control of her disease. Interestingly, eculizumab therapy was associated with a further decline of complement C3 and C4 serum levels. The patient developed a subsequent flare of her systemic lupus erythematosus, potentially indicating that complement inhibition by eculizumab is not effective in preventing lupus flares.Taken together, we describe a unique case of life-threatening and difficult-to-treat CAPS with a good clinical response after terminal complement complex inhibition with eculizumab. Further controlled trials are necessary to investigate the value of eculizumab in patients with CAPS.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Antifosfolipídica/terapia , Adulto , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/diagnóstico , Doença Catastrófica , Feminino , Humanos , Deficiência de IgA/complicações , Deficiência de IgA/diagnóstico , Deficiência de IgA/terapia , Imunoterapia/métodos , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/terapia
16.
J Am Heart Assoc ; 2(3): e000204, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23735897

RESUMO

BACKGROUND: We compared high-sensitivity cardiac troponin T (hs-cTnT) and standard cTnT for acute myocardial infarction (AMI) diagnosis in everyday clinical practice of an emergency department (ED). METHODS AND RESULTS: cTnT was measured in 2384 consecutive patients (60 ± 21 years, 52% female) on ED admission. Readmissions to the ED (n=720) and mortality (n=101) were followed for an average period of 239 ± 49 days. There were 53 AMIs (delay, 1 to 96 hours; median, 3 hours), 440 chest pain patients, 286 dyspnea patients, 785 acute or chronic cardiac diseases, and 540 neurological diseases, with the remaining having various internal diseases. The diagnostic performances of hs- and standard cTnT were comparable for AMI diagnosis (area under receiver operating characteristics curves [ROC AUC], 0.91 ± 0.02 versus 0.90 ± 0.03; P=0.31). Using the 99th-percentile cutoff, the sensitivities and specificities for AMI in the whole population were 91% and 74% for hs-cTnT and 89% and 80% for standard cTnT. hs-cTnT detected significantly more patients with cardiac diseases (ROC AUC, 0.77 ± 0.01 versus 0.67 ± 0.01; P<0.001). hs-cTnT and standard cTnT were significant predictors of ED readmissions but not of mortality, but both were not independent predictors of ED readmissions or the combined end point of readmission or mortality in binary logistic regression analysis. CONCLUSIONS: In unselected ED patients the diagnostic performances of hs-cTnT and standard cTnT for AMI diagnosis did not differ significantly. hs-cTnT detected significantly more cardiac diseases. hs-cTnT and standard cTnT were not independent predictors of ED readmissions and mortality from all causes.


Assuntos
Testes Diagnósticos de Rotina , Admissão do Paciente , Troponina T/sangue , Emergências , Serviço Hospitalar de Emergência , Feminino , Testes Hematológicos/normas , Humanos , Masculino , Pessoa de Meia-Idade
17.
Cell Physiol Biochem ; 14(1-2): 101-12, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14976411

RESUMO

BACKGROUND/AIMS: Adhesion of intratubular leukocytes to proximal tubules in biopsies of patients with rapidly progressive glomerulonephritis and the appearance of leukocytes in the urine in interstitial nephritis suggest interactions between leukocytes and tubular epithelia in renal disease. The present study was performed to investigate whether incubation of tubular epithelia with cytokines or endotoxin (LPS) does stimulate adhesion and migration of leukocytes through these epithelia in vitro. METHODS: Experiments determined adhesion of PMN to LLC-PK cells cultured on tissue culture plates and transepithelial migration (TEM) through LLC-PK monolayers cultured on microporous membranes. Measurements of transepithelial electrical resistance (TER), cytokine release into apical or basolateral compartments and chemotactic activities of apical and basolateral supernatants were performed. RESULTS: Preincubation of LLC-PK cells with either TNFalpha or LPS resulted in stimulation of PMN adhesion and consequently PMN migration through LLC-PK monolayers in both apical-to-basolateral and basolateral-to-apical direction. TEM was not associated with a reduction of TER. Although largely apical IL-8 secretion by LLC-PK cells was found, apical-to-basolateral migration occurred against a concentration gradient of IL-8 and could not be inhibited by IL-8 antibodies. Chemotactic activities of supernatants were slightly increased by TEM but did not show any significant differences between apical and basolateral compartments independent of the direction of PMN migration. TEM in basolateral-to-apical direction was about twice as efficient as in apical-to-basolateral direction (Transmigration Index = 3,92 +/- 0,55 and 2,28 +/-,21, respectively). Only basolateral-to-apical TEM could be partly inhibited by preincubation of basolateral membrane with IL-8 antibodies. CONCLUSION: Inflammatory mediators stimulate PMN adherence to LLC-PK cells and subsequently TEM. The mechanisms involved in TEM stimulated by cytokines or endotoxin appear to be rather changes in surface receptor properties of LLC-PK cells than chemotactic stimuli. Basolateral-to-apical TEM appears to be the favored direction most likely augmented by IL-8 associated haptotactic PMN stimulation.


Assuntos
Movimento Celular/fisiologia , Túbulos Renais Proximais/metabolismo , Neutrófilos/fisiologia , Animais , Anticorpos/farmacologia , Transporte Biológico/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Citocinas/farmacologia , Relação Dose-Resposta a Droga , Endotoxinas/farmacologia , Humanos , Túbulos Renais Proximais/citologia , Células LLC-PK1 , Leucócitos/metabolismo , Neutrófilos/efeitos dos fármacos , Suínos , Fatores de Tempo
18.
Mol Ther ; 6(5): 576-83, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12409255

RESUMO

This study was designed to test the hypothesis that transcutaneous ultrasound (US) exposure may augment the transfection efficiency and biological outcome associated with nonviral DNA gene transfer. Hindlimb muscles of New Zealand White rabbits were transfected with the reporter plasmid pCMV-beta, with or without US exposure. Optimization studies employed US exposure at various frequencies, mechanical indices, duty cycles, durations of exposure, and exposure time points. Based on these results, we explored the effect of US exposure on nonviral gene transfer of vascular endothelial growth factor (VEGF, phVEGF165) to promote neovascularization of ischemic hindlimbs. Ultrasound at 1 MHz, 100 W/cm(2), 6% duty cycle, and 5 minutes exposure time, applied immediately following DNA injection, was found to be the most effective among the settings tested, increasing beta-galactosidase expression approximately 20 fold. Compared with US exposure alone, or phVEGF165 only, phVEGF165 + US exposure yielded a statistically significant improvement in revascularization, as determined by calf blood pressure ratio, angiographic score, intravascular Doppler blood flow, and capillary/myocyte ratio. These data demonstrate that ultrasound, when applied directly after intramuscular gene transfer, significantly increases transfection efficiency in vivo. The biological significance of this finding was confirmed by augmented limb perfusion in response to US exposure and naked VEGF DNA.


Assuntos
DNA , Técnicas de Transferência de Genes , Terapia Genética/métodos , Músculo Esquelético/metabolismo , Ultrassom , Animais , Capilares/metabolismo , Genes Reporter , Hemodinâmica , Isquemia , Masculino , Células Musculares/metabolismo , Neovascularização Patológica , Plasmídeos/metabolismo , Coelhos , Transfecção , Doenças Vasculares/terapia , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA