Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(17)2022 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-36076955

RESUMO

Spinal strokes may be associated with tremendous spinal cord injury. Erythropoietin (EPO) improves the neurological outcome of animals after spinal cord ischemia (SCI) and its effects on ischemia-induced endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) are considered possible molecular mechanisms. Furthermore, sphingosin-1-phosphate (S1P) is suggested to correlate with SCI. In this study, the effect of recombinant human EPO (rhEPO) and carbamylated EPO (cEPO-Fc) on the outcome of mice after SCI and a prognostic value of S1P were investigated. SCI was induced in 12-month-old male mice by thoracic aortal cross-clamping after administration of rhEPO, cEPO-Fc, or a control. The locomotory behavior of mice was evaluated by the Basso mouse scale and S1P serum levels were measured by liquid chromatography-tandem mass spectrometry. The spinal cord was examined histologically and the expressions of key UPR proteins (ATF6, PERK, and IRE1a, caspase-12) were analyzed utilizing immunohistochemistry and real-time quantitative polymerase chain reaction. RhEPO and cEPO-Fc significantly improved outcomes after SCI. The expression of caspase-12 significantly increased in the control group within the first 24 h of reperfusion. Animals with better locomotory behavior had significantly higher serum levels of S1P. Our data indicate that rhEPO and cEPO-Fc have protective effects on the clinical outcome and neuronal tissue of mice after SCI and that the ER is involved in the molecular mechanisms. Moreover, serum S1P may predict the severity of impairment after SCI.


Assuntos
Eritropoetina , Fármacos Neuroprotetores , Traumatismos da Medula Espinal , Isquemia do Cordão Espinal , Acidente Vascular Cerebral , Animais , Caspase 12 , Epoetina alfa , Eritropoetina/análogos & derivados , Eritropoetina/farmacologia , Humanos , Lactente , Lisofosfolipídeos , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes/farmacologia , Esfingosina/análogos & derivados , Traumatismos da Medula Espinal/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico
2.
Stroke ; 52(12): 3901-3907, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34496616

RESUMO

BACKGROUND AND PURPOSE: The aim of this study was to examine whether sphingosine-1-phosphate (S1P) levels in patients with acute stroke are associated with stroke severity and outcome. METHODS: In a prospective stroke cohort (MARK-STROKE), 374 patients with acute ischemic stroke or transient ischemic attack were enrolled (mean age: 67.9±13.0 years, sex: 64.7% male), and serum-S1P at admission was analyzed with tandem mass spectrometry. In addition to cross-sectional analyses, 79 adverse events (death, stroke, myocardial infarction, rehospitalization) were recorded in 270 patients during follow-up. Regression analyses were adjusted for age, sex, low-density lipoprotein cholesterol, and vascular risk factors. Results were validated in an independent stroke cohort with 219 patients with acute ischemic stroke (CIRCULAS). RESULTS: Low serum-S1P was associated with higher National Institutes of Health Stroke Scale score at admission and with anterior circulation nonlacunar infarcts determined by multivariate regression analyses. During a follow-up of 294±170 days, patients with S1P in the lowest tertile (<1.33 µmol/L) had more adverse events (Kaplan-Meier analysis, P=0.048 for trend). In adjusted Cox regression analysis, the lowest S1P tertile was associated with a worse outcome after stroke (hazard ratio, HR 0.51 [95% confidence interval 0.28-0.92]). Results were confirmed in an independent cohort, ie, low S1P levels were associated with higher National Institutes of Health Stroke Scale, larger infarct volumes and worse outcome after 90 days (ß-coefficient: -0.03, P=0.026; ß-coefficient: -0.099, P=0.009 and odds ratio 0.52 [0.28-0.96], respectively). CONCLUSIONS: Our findings imply a detrimental role of low S1P levels in acute stroke and therefore underpin the therapeutic potential of S1P-mimics.


Assuntos
Biomarcadores/sangue , Isquemia Encefálica/sangue , AVC Isquêmico/sangue , Lisofosfolipídeos/sangue , Esfingosina/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Isquemia Encefálica/complicações , Feminino , Humanos , AVC Isquêmico/etiologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Esfingosina/sangue
3.
J Perinat Med ; 49(7): 932-935, 2021 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-33857362

RESUMO

OBJECTIVES: Sphingosine-1-phosphate (S1P) is a signalling lipid involved in embryonic development, physiological homeostasis, and pathogenic processes in multiple organ systems. Disturbance of S1P homeostasis has been associated with various human diseases in which the immune response and vascular integrity are severely compromised. Up-to-date, no study has analyzed S1P levels in neonates. The objective of this study was to determine S1P serum concentrations in neonates and establish S1P reference ranges. METHODS: S1P levels in the umbilical cord blood of 460 term and preterm neonates were compared to a previously described cohort of healthy adult blood donors. S1P levels were further correlated with demographic characteristics, cellular sources of S1P, and inflammatory markers. RESULTS: The median S1P serum level in neonates was 1.70 µmol/L (IQR 1.41-1.97 µmol/L) and significantly higher than normal values reported in adults. S1P levels correlated positively with the number of red blood cells (p<0.001) and negatively with neutrophil precursors (p=0.028). CONCLUSIONS: Elevated S1P levels in neonates compared to adults possibly result from higher S1P content in its cellular sources due to the essential role of S1P during embryogenesis. Generated S1P ranges may be used as reference ranges for future studies in neonates.


Assuntos
Sangue Fetal/metabolismo , Recém-Nascido/sangue , Lisofosfolipídeos/sangue , Esfingosina/análogos & derivados , Adulto , Biomarcadores/sangue , Feminino , Humanos , Recém-Nascido Prematuro/sangue , Masculino , Espectrometria de Massas , Estudos Prospectivos , Valores de Referência , Esfingosina/sangue
4.
Br J Anaesth ; 125(2): 122-132, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32711724

RESUMO

BACKGROUND: Despite several clinical trials on haemodynamic therapy, the optimal intraoperative haemodynamic management for high-risk patients undergoing major abdominal surgery remains unclear. We tested the hypothesis that personalised haemodynamic management targeting each individual's baseline cardiac index at rest reduces postoperative morbidity. METHODS: In this single-centre trial, 188 high-risk patients undergoing major abdominal surgery were randomised to either routine management or personalised haemodynamic management requiring clinicians to maintain personal baseline cardiac index (determined at rest preoperatively) using an algorithm that guided intraoperative i.v. fluid and/or dobutamine administration. The primary outcome was a composite of major complications (European Perioperative Clinical Outcome definitions) or death within 30 days of surgery. Secondary outcomes included postoperative morbidity (assessed by a postoperative morbidity survey), hospital length of stay, mortality within 90 days of surgery, and neurocognitive function assessed after postoperative Day 3. RESULTS: The primary outcome occurred in 29.8% (28/94) of patients in the personalised management group, compared with 55.3% (52/94) of patients in the routine management group (relative risk: 0.54, 95% confidence interval [CI]: 0.38 to 0.77; absolute risk reduction: -25.5%, 95% CI: -39.2% to -11.9%; P<0.001). One patient assigned to the personalised management group, compared with five assigned to the routine management group, died within 30 days after surgery (P=0.097). There were no clinically relevant differences between the two groups for secondary outcomes. CONCLUSIONS: In high-risk patients undergoing major abdominal surgery, personalised haemodynamic management reduces a composite outcome of major postoperative complications or death within 30 days after surgery compared with routine care. CLINICAL TRIAL REGISTRATION: NCT02834377.


Assuntos
Abdome/cirurgia , Débito Cardíaco/fisiologia , Hidratação/métodos , Hemodinâmica/fisiologia , Cuidados Intraoperatórios/métodos , Complicações Pós-Operatórias/prevenção & controle , Idoso , Feminino , Humanos , Tempo de Internação/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/fisiopatologia , Estudos Prospectivos , Risco
5.
J Vasc Surg ; 68(6S): 201S-207S, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29804740

RESUMO

OBJECTIVE: The main objective of this study was to define a role of sphingosine-1-phosphate receptor 1 (S1PR1) in the arterial injury response of a human artery. The hypotheses were tested that injury induces an expansion of S1PR1-positive cells and that these cells accumulate toward the lumen because they follow the sphingosine-1-phosphate gradient from arterial wall tissue (low) to plasma (high). METHODS: A humanized rat model was used in which denuded human internal mammary artery (IMA) was implanted into the position of the abdominal aorta of immunosuppressed Rowett nude rats. This injury model is characterized by medial as well as intimal hyperplasia, whereby intimal cells are of human origin. At 7, 14, and 28 days after implantation, grafts were harvested and processed for fluorescent immunostaining for S1PR1 and smooth muscle α-actin. Nuclei were stained with 4',6-diamidine-2'-phenylindole dihydrochloride. Using digitally reconstructed, complete cross sections of grafts, intimal and medial areas were measured, whereby the medial area had virtually been divided into an outer (toward adventitia) and inner (toward lumen) layer. The fraction of S1PR1-positive cells was determined in each layer by counting S1PR1-positive and S1PR1-negative cells. RESULTS: The fraction of S1PR1-postive cells in naive IMA is 58.9% ± 6.0% (mean ± standard deviation). At day 28 after implantation, 81.6% ± 4.4% of medial cells were scored S1PR1 positive (P < .01). At day 14, the ratio between S1PR1-positive and S1PR1-negative cells was significantly higher in the lumen-oriented inner layer (9.3 ± 2.1 vs 6.0 ± 1.0; P < .01). Cells appearing in the intima at day 7 and day 14 were almost all S1PR1 positive. At day 28, however, about one-third of intimal cells were scored S1PR1 negative. CONCLUSIONS: From these data, we conclude that denudation of IMA specifically induces the expansion of S1PR1-positive cells. Based on the nonrandom distribution of S1PR1-positive cells, we consider the possibility that much like lymphocytes, S1PR1-positive smooth muscle cells also use S1PR1 to recognize the sphingosine-1-phosphate gradient from tissue (low) to plasma (high) and so migrate out of the media toward the intima of the injured IMA.


Assuntos
Aorta Abdominal/cirurgia , Oclusão de Enxerto Vascular/metabolismo , Artéria Torácica Interna/transplante , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/transplante , Neointima , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Oclusão de Enxerto Vascular/etiologia , Oclusão de Enxerto Vascular/patologia , Humanos , Lisofosfolipídeos/metabolismo , Masculino , Artéria Torácica Interna/metabolismo , Artéria Torácica Interna/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos Nus , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo
6.
Crit Care ; 19: 372, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26498205

RESUMO

INTRODUCTION: Sphingosine-1-phosphate (S1P) is a signaling lipid that regulates pathophysiological processes involved in sepsis progression, including endothelial permeability, cytokine release, and vascular tone. The aim of this study was to investigate whether serum-S1P concentrations are associated with disease severity in patients with sepsis. METHODS: This single-center prospective-observational study includes 100 patients with systemic inflammatory response syndrome (SIRS) plus infection (n = 40), severe sepsis (n = 30), or septic shock (n = 30) and 214 healthy blood donors as controls. Serum-S1P was measured by mass spectrometry. Blood parameters, including C-reactive protein (CRP), procalcitonin (PCT), interleukin-6 (IL-6), lactate, and white blood cells (WBCs), were determined by routine assays. The Sequential Organ Failure Assessment (SOFA) score was generated and used to evaluate disease severity. RESULTS: Serum-S1P concentrations were lower in patients than in controls (P < 0.01), and the greatest difference was between the control and the septic shock groups (P < 0.01). Serum-S1P levels were inversely correlated with disease severity as determined by the SOFA score (P < 0.01) as well as with IL-6, PCT, CRP, creatinine, lactate, and fluid balance. A receiver operating characteristic analysis for the presence or absence of septic shock revealed equally high sensitivity and specificity for S1P compared with the SOFA score. In a multivariate logistic regression model calculated for prediction of septic shock, S1P emerged as the strongest predictor (P < 0.001). CONCLUSIONS: In patients with sepsis, serum-S1P levels are dramatically decreased and are inversely associated with disease severity. Since S1P is a potent regulator of endothelial integrity, low S1P levels may contribute to capillary leakage, impaired tissue perfusion, and organ failure in sepsis.


Assuntos
Lisofosfolipídeos/sangue , Insuficiência de Múltiplos Órgãos/mortalidade , Sepse/mortalidade , Esfingosina/análogos & derivados , Adulto , Feminino , Alemanha , Humanos , Lisofosfolipídeos/deficiência , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/sangue , Estudos Prospectivos , Sepse/sangue , Sepse/terapia , Índice de Gravidade de Doença , Esfingosina/sangue , Esfingosina/deficiência
7.
Arterioscler Thromb Vasc Biol ; 32(4): 955-61, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22308044

RESUMO

OBJECTIVE: The objective of this study was to define a role for sphingosine-1-phosphate receptor 3 (S1PR3) in intimal hyperplasia. METHODS AND RESULTS: A denudation model of the iliac-femoral artery in wild-type and S1PR3-null mice was used to define a role for S1PR3 in the arterial injury response because we found in humans and mice that expression of S1PR3 was higher in these arteries compared with carotid arteries. At 28 days after surgery, wild-type arteries formed significantly larger lesions than S1PR3-null arteries. Bromodeoxyuridine labeling experiments demonstrated that on injury, wild-type arteries exhibited higher medial as well as intimal proliferation than S1PR3-null arteries. Because S1PR3 expression in vitro was low, we expressed S1PR3 in S1PR3-null smooth muscle cells (SMCs) using retroviral-mediated gene transfer to study the effects of S1PR3 on cell functions and signaling. SMCs expressing S1PR3, but not vector-transfected controls, responded to sphingosine-1-phosphate stimulation with activation of Rac, Erk, and Akt. SMCs expressing S1PR3 also migrated more. CONCLUSIONS: In humans and mice, S1PR3 expression was higher in iliac-femoral arteries compared with carotid arteries. S1PR3 promoted neointimal hyperplasia on denudation of iliac-femoral arteries in mice, likely by stimulating cell migration and proliferation through activation of signaling pathways involving Erk, Akt, and Rac.


Assuntos
Proliferação de Células , Artéria Femoral/metabolismo , Artéria Ilíaca/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Túnica Íntima/metabolismo , Lesões do Sistema Vascular/metabolismo , Animais , Artérias Carótidas/metabolismo , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Artéria Femoral/patologia , Humanos , Hiperplasia , Artéria Ilíaca/patologia , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo , Transfecção , Túnica Íntima/patologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia , Proteínas rac de Ligação ao GTP/metabolismo
8.
Hum Mol Genet ; 19(3): 468-79, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19926569

RESUMO

Vascular smooth muscle cells (SMCs) display remarkable phenotypic plasticity in response to environmental cues. The nuclear factor of activated T-cells (NFAT) family of transcription factors plays a critical role in vascular pathology. However, known functional NFAT gene targets in vascular SMCs are currently limited. Publicly available whole-genome expression array data sets were analyzed to identify differentially expressed genes in human, mouse and rat SMCs. Comparison between vehicle and phenotypic modulatory stimuli identified 63 species-conserved, upregulated genes. Integration of the 63 upregulated genes with an in silico NFAT-ome (a species-conserved list of gene promoters containing at least one NFAT binding site) identified 18 putative NFAT-dependent genes. Further intersection of these 18 potential NFAT target genes with a mouse in vivo vascular injury microarray identified four putative NFAT-dependent, injury-responsive genes. In vitro validations substantiated the NFAT-dependent role of Cyclooxygenase 2 (COX2/PTGS2) in SMC phenotypic modulation and uncovered Down Syndrome Candidate Region 1 (DSCR1/RCAN1) as a novel NFAT target gene in SMCs. We show that induction of DSCR1 inhibits calcineurin/NFAT signaling through a negative feedback mechanism; DSCR1 overexpression attenuates NFAT transcriptional activity and COX2 protein expression, whereas knockdown of endogenous DSCR1 enhances NFAT transcriptional activity. Our integrative genomics approach illustrates how the combination of publicly available gene expression arrays, computational databases and empirical research methods can answer specific questions in any cell type for a transcriptional network of interest. Herein, we report DSCR1 as a novel NFAT-dependent, injury-inducible, early gene that may serve to negatively regulate SMC phenotypic switching.


Assuntos
Genômica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição NFATC/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Proteínas de Ligação a DNA , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/genética , Fatores de Transcrição NFATC/genética , Ligação Proteica , Ratos
9.
Arterioscler Thromb Vasc Biol ; 31(7): 1530-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21677296

RESUMO

Conventional views of the tunica adventitia as a poorly organized layer of vessel wall composed of fibroblasts, connective tissue, and perivascular nerves are undergoing revision. Recent studies suggest that the adventitia has properties of a stem/progenitor cell niche in the artery wall that may be poised to respond to arterial injury. It is also a major site of immune surveillance and inflammatory cell trafficking and harbors a dynamic microvasculature, the vasa vasorum, that maintains the medial layer and provides an important gateway for macrophage and leukocyte migration into the intima. In addition, the adventitia is in contact with tissue that surrounds the vessel and may actively participate in exchange of signals and cells between the vessel wall and the tissue in which it resides. This brief review highlights recent advances in our understanding of the adventitia and its resident progenitor cells and discusses progress toward an integrated view of adventitial function in vascular development, repair, and disease.


Assuntos
Tecido Conjuntivo/patologia , Células-Tronco/patologia , Doenças Vasculares/patologia , Animais , Comunicação Celular , Tecido Conjuntivo/imunologia , Tecido Conjuntivo/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Fenótipo , Transdução de Sinais , Nicho de Células-Tronco , Células-Tronco/imunologia , Células-Tronco/metabolismo , Doenças Vasculares/imunologia , Doenças Vasculares/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 31(12): 2827-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21903940

RESUMO

OBJECTIVE: Obesity is characterized by chronic inflammation of adipose tissue, which contributes to insulin resistance and diabetes. Although nitric oxide (NO) signaling has antiinflammatory effects in the vasculature, whether reduced NO contributes to adipose tissue inflammation is unknown. We sought to determine whether (1) obesity induced by high-fat (HF) diet reduces endothelial nitric oxide signaling in adipose tissue, (2) reduced endothelial nitric oxide synthase (eNOS) signaling is sufficient to induce adipose tissue inflammation independent of diet, and (3) increased cGMP signaling can block adipose tissue inflammation induced by HF feeding. METHODS AND RESULTS: Relative to mice fed a low-fat diet, an HF diet markedly reduced phospho-eNOS and phospho-vasodilator-stimulated phosphoprotein (phospho-VASP), markers of vascular NO signaling. Expression of proinflammatory cytokines was increased in adipose tissue of eNOS-/- mice. Conversely, enhancement of signaling downstream of NO by phosphodiesterase-5 inhibition using sildenafil attenuated HF-induced proinflammatory cytokine expression and the recruitment of macrophages into adipose tissue. Finally, we implicate a role for VASP, a downstream mediator of NO-cGMP signaling in mediating eNOS-induced antiinflammatory effects because VASP-/- mice recapitulated the proinflammatory phenotype displayed by eNOS-/- mice. CONCLUSIONS: These results imply a physiological role for endothelial NO to limit obesity-associated inflammation in adipose tissue and hence identify the NO-cGMP-VASP pathway as a potential therapeutic target in the treatment of diabetes.


Assuntos
Tecido Adiposo/fisiopatologia , GMP Cíclico/metabolismo , Gorduras na Dieta/efeitos adversos , Endotélio Vascular/metabolismo , Inflamação/fisiopatologia , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Tecido Adiposo/metabolismo , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Obesidade/induzido quimicamente , Obesidade/metabolismo , Obesidade/fisiopatologia , Inibidores da Fosfodiesterase 5/farmacologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Purinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Citrato de Sildenafila , Sulfonas/farmacologia
11.
Am J Physiol Cell Physiol ; 301(2): C478-89, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21593453

RESUMO

Regulator of G protein signaling (RGS) proteins, and notably members of the RGS-R4 subfamily, control vasocontractility by accelerating the inactivation of Gα-dependent signaling. RGS5 is the most highly and differently expressed RGS-R4 subfamily member in arterial smooth muscle. Expression of RGS5 first appears in pericytes during development of the afferent vascular tree, suggesting that RGS5 is a good candidate for a regulator of arterial contractility and, perhaps, for determining the mass of the smooth muscle coats required to regulate blood flow in the branches of the arterial tree. Consistent with this hypothesis, using cultured vascular smooth muscle cells (VSMCs), we demonstrate RGS5 overexpression inhibits G protein-coupled receptor (GPCR)-mediated hypertrophic responses. The next objective was to determine which physiological agonists directly control RGS5 expression in VSMCs. GPCR agonists failed to directly regulate RGS5 mRNA expression; however, platelet-derived growth factor (PDGF) acutely represses expression. Downregulation of RGS5 results in the induction of migration and the activation of the GPCR-mediated signaling pathways. This stimulation leads to the activation of mitogen-activated protein kinases directly downstream of receptor stimulation, and ultimately VSMC hypertrophy. These results demonstrate that RGS5 expression is a critical mediator of both VSMC contraction and potentially, arterial remodeling.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Angiotensina II/metabolismo , Animais , Becaplermina , Linhagem Celular , Movimento Celular , Regulação da Expressão Gênica , Hipertrofia , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis , Proteínas RGS/deficiência , Proteínas RGS/genética , Interferência de RNA , Ratos , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Vasoconstrição
12.
Arterioscler Thromb Vasc Biol ; 30(4): 758-65, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20093624

RESUMO

OBJECTIVE: Diet-induced obesity (DIO) in mice causes vascular inflammation and insulin resistance that are accompanied by decreased endothelial-derived NO production. We sought to determine whether reduced NO-cGMP signaling contributes to the deleterious effects of DIO on the vasculature and, if so, whether these effects can be blocked by increased vascular NO-cGMP signaling. METHODS AND RESULTS: By using an established endothelial cell culture model of insulin resistance, exposure to palmitate, 100 micromol/L, for 3 hours induced both cellular inflammation (activation of IKK beta-nuclear factor-kappaB) and impaired insulin signaling via the insulin receptor substrate-phosphatidylinositol 3-kinase pathway. Sensitivity to palmitate-induced endothelial inflammation and insulin resistance was increased when NO signaling was reduced using an endothelial NO synthase inhibitor, whereas endothelial responses to palmitate were blocked by pretreatment with either an NO donor or a cGMP analogue. To investigate whether endogenous NO-cGMP signaling protects against vascular responses to nutrient excess in vivo, adult male mice lacking endothelial NO synthase were studied. As predicted, both vascular inflammation (phosphorylated I kappaB alpha and intercellular adhesion molecule levels) and insulin resistance (phosphorylated Akt [pAkt] and phosphorylated eNOS [peNOS] levels) were increased in endothelial NO synthase(-/-) (eNOS(-/-)) mice, reminiscent of the effect of DIO in wild-type controls. Next, we asked whether the vascular response to DIO in wild-type mice can be reversed by a pharmacological increase of cGMP signaling. C57BL6 mice were either fed a high-fat diet or remained on a low-fat diet for 8 weeks. During the final 2 weeks of the study, mice on each diet received either placebo or the phosphodiesterase-5 inhibitor sildenafil, 10 mg/kg per day orally. In high-fat diet-fed mice, vascular inflammation and insulin resistance were completely prevented by sildenafil administration at a dose that had no effect in mice fed the low-fat diet. CONCLUSIONS: Reduced signaling via the NO-cGMP pathway is a mediator of vascular inflammation and insulin resistance during overnutrition induced by high-fat feeding. Therefore, phosphodiesterase-5, soluble guanylyl cyclase, and other molecules in the NO-cGMP pathway (eg, protein kinase G) constitute potential targets for the treatment of vascular dysfunction in the setting of obesity.


Assuntos
Aorta Torácica/metabolismo , Doenças da Aorta/metabolismo , GMP Cíclico/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Resistência à Insulina , Óxido Nítrico/metabolismo , Transdução de Sinais , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiopatologia , Doenças da Aorta/etiologia , Doenças da Aorta/fisiopatologia , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Gorduras na Dieta , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Inibidores Enzimáticos/farmacologia , Humanos , Quinase I-kappa B/metabolismo , Inflamação/etiologia , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Ácido Palmítico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Fosfodiesterase 5 , Inibidores de Fosfodiesterase/farmacologia , Fosforilação , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Purinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Citrato de Sildenafila , Sulfonas/farmacologia
13.
Arterioscler Thromb Vasc Biol ; 29(9): 1356-62, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19592464

RESUMO

OBJECTIVE: Arterial injury induces smooth muscle cell (SMC) proliferation, migration, and intimal accumulation of cells and extracellular matrix. These processes are regulated by the administration of the glycosaminoglycans heparin and heparan sulfate, but little is known about the role of endogenous heparan sulfate proteoglycans in the vessel wall. We investigated the response to carotid injury of syndecan-1-null mice to assess the function of one of a conserved family of transmembrane heparan and chondroitin sulfate proteoglycans. METHODS AND RESULTS: Syndecan-1-null mice developed a large neointimal lesion after injury, whereas wild-type mice made little or none. This was accompanied by a significant increase in both medial and intimal SMC replication. Cultured syndecan-1-null SMCs showed a significant increase in proliferation in response to PDGF-BB, thrombin, FGF2, EGF, and serum. In response to thrombin, PDGF-BB, and serum syndecan-1-null SMCs expressed more PDGF-B chain message than did wild-type SMCs. Downregulation of PDGF-BB or PDGFRbeta inhibited thrombin-, PDGF-BB-, and serum-induced DNA synthesis in syndecan-1-null SMCs. CONCLUSIONS: These results suggest the possibility that syndecan-1 may limit intimal thickening in injured arteries by suppressing SMC activation through inhibition of SMC PDGF-B chain expression and PDGFRbeta activation.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Proliferação de Células , Músculo Liso Vascular/metabolismo , Sindecana-1/metabolismo , Túnica Íntima/metabolismo , Animais , Becaplermina , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Movimento Celular , Células Cultivadas , Replicação do DNA , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Sindecana-1/deficiência , Sindecana-1/genética , Trombina/metabolismo , Fatores de Tempo , Túnica Íntima/patologia
14.
Arterioscler Thromb Vasc Biol ; 29(10): 1644-50, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19608972

RESUMO

OBJECTIVE: This study tests the hypothesis that S1P2R regulates expression of SMC differentiation genes after arterial injury. METHODS AND RESULTS: Carotid ligation injury was performed in wild-type and S1P2R-null mice. At various time points after injury, expression of multiple SMC differentiation genes, myocardin, and S1P receptors (S1P1R, S1P2R, and S1P3R) was measured by quantitative PCR. These experiments demonstrate that at day 7 after injury, S1P2R specifically regulates expression of smooth muscle alpha-actin (SMA) and that this is not mediated by changes in expression of myocardin or any of the S1PRs. In vitro studies using carotid SMCs prepared from wild-type and S1P2R-null mice show that S1P stimulates expression of all SMC-differentiation genes tested, but S1P2R significantly regulates expression of SMA and SM22 alpha only. Chromatin immunoprecipitation assays suggest that S1P-induced recruitment of serum response factor to the SMA promoter and enhancer largely depends on S1P2R. S1P-stimulated SMA expression requires S1P2R-dependent activation of RhoA and mobilization of calcium from intracellular stores. Chelation of calcium does not affect the activation of RhoA by S1P, whereas blockade of Rho by C3 exotoxin partially inhibits the mobilization of calcium by S1P. CONCLUSIONS: The results of this study support the hypothesis that S1P2R regulates expression of SMA after injury. We further conclude that transcriptional regulation of SMA by S1P in vitro requires S1P2R-dependent activation of RhoA and mobilization of calcium from intracellular calcium stores.


Assuntos
Actinas/genética , Lesões das Artérias Carótidas/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Cálcio/metabolismo , Regulação da Expressão Gênica , Lisofosfolipídeos/farmacologia , Masculino , Camundongos , RNA Mensageiro/análise , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo
15.
J Nucl Med ; 59(2): 266-272, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28775206

RESUMO

The CXC-motif chemokine receptor 4 (CXCR4) represents a promising target for molecular imaging of different CXCR4-positive cell types in cardiovascular diseases such as atherosclerosis and arterial wall injury. The aim of this study was to assess the prevalence, pattern, and clinical correlates of arterial wall accumulation of 68Ga-pentixafor, a specific CXCR4 ligand for PET. Methods: The data for 51 patients who underwent 68Ga-pentixafor PET/CT for noncardiovascular indications were retrospectively analyzed. Tracer accumulation in the vessel wall of major arteries was analyzed qualitatively and semiquantitatively by blood-pool-corrected target-to-background ratios. Tracer uptake was compared with calcified plaque burden and cardiovascular risk factors. Results: Focal arterial uptake of 68Ga-pentixafor was seen at 1,411 sites in 51 (100%) of patients. 68Ga-pentixafor uptake was significantly associated with calcified plaque burden (P < 0.0001) and cardiovascular risk factors including age (P < 0.0001), arterial hypertension (P < 0.0001), hypercholesterolemia (P = 0.0005), history of smoking (P = 0.01), and prior cardiovascular events (P = 0.0004). Both the prevalence (P < 0.0001) and the signal intensity (P = 0.009) of 68Ga-pentixafor uptake increased as the number of risk factors increased. Conclusion:68Ga-pentixafor PET/CT is suitable for noninvasive, highly specific PET imaging of CXCR4 expression in the atherosclerotic arterial wall. Arterial wall 68Ga-pentixafor uptake is significantly associated with surrogate markers of atherosclerosis and is linked to the presence of cardiovascular risk factors. 68Ga-pentixafor signal is higher in patients with a high-risk profile and may hold promise for identification of vulnerable plaque.


Assuntos
Calcinose/complicações , Complexos de Coordenação , Regulação da Expressão Gênica , Peptídeos Cíclicos , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/metabolismo , Receptores CXCR4/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Transporte Biológico , Complexos de Coordenação/metabolismo , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Peptídeos Cíclicos/metabolismo , Placa Aterosclerótica/complicações , Estudos Retrospectivos , Fatores de Risco , Adulto Jovem
16.
Biomark Med ; 12(2): 119-127, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29327601

RESUMO

AIM: Sepsis is a serious complication following surgery and identification of patients at risk is of high importance. Syndecan-1 (sSDC1) levels are known to be elevated during sepsis. MATERIALS & METHODS: Fifty-five patients scheduled for major abdominal surgery were prospectively included and sSDC1 concentrations were measured during hospital stay. RESULTS: Patients with postoperative sepsis showed a continued increase of sSDC1 levels and exhibited higher median sSDC1 concentrations at day 1 compared with nonseptic patients 90.3 versus 16.5 ng/ml. A significant association of sSDC1 levels with the incidence of sepsis and death was demonstrated. CONCLUSION: This study identifies sSDC1 as potential biomarker for sepsis and survival after abdominal surgery.


Assuntos
Abdome/cirurgia , Biomarcadores/sangue , Sepse/diagnóstico , Sindecana-1/sangue , Idoso , Área Sob a Curva , Proteína C-Reativa/análise , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Unidades de Terapia Intensiva , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias , Estudos Prospectivos , Curva ROC , Sepse/etiologia , Sepse/mortalidade
17.
Circ Res ; 96(2): 172-9, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15625285

RESUMO

We have shown that the G protein-coupled receptor (GPCR) agonists, thrombin and Factor Xa, stimulate smooth muscle cell (SMC) proliferation through transactivation of the EGF receptor (EGFR) or the FGF receptor (FGFR), both of which are tyrosine kinase receptors. In the present study, we investigated whether platelet-derived growth factor (PDGF), a tyrosine kinase receptor agonist, might transactivate another tyrosine kinase receptor to induce SMC proliferation. Because heparin inhibits PDGF-mediated proliferation in human SMCs, we investigated whether the heparin-binding growth factor basic fibroblast growth factor (bFGF) and one of its receptors, FGFR-1, play a role in the response of human arterial SMCs to PDGF-BB. PDGF-BB induced the release of bFGF and sustained phosphorylation of FGFR-1 (30 minutes to 6 hours). A bFGF-neutralizing antibody inhibited PDGF-BB-mediated phosphorylation of FGFR-1, DNA synthesis, and cell proliferation. In the presence of bFGF antibody, PDGF-BB-induced early activation of ERK (0 to 60 minutes) was not affected, whereas late ERK activation (2 to 4 hours) was reduced. When FGFR-1 expression was suppressed using small interfering RNA (siRNA), ERK activation was reduced at late, but not early, time points after PDGF-BB stimulation. Addition of bFGF antibody to cells treated with siRNA to FGFR-1 had no further effect on ERK activation. Our results provide support for a novel mechanism by which PDGF-BB induces the release of bFGF and activation of FGFR-1 followed by the sustained activation of ERK and proliferation of human SMCs.


Assuntos
Fator 2 de Crescimento de Fibroblastos/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Aorta Abdominal , Becaplermina , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Cromonas/farmacologia , Replicação do DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Flavonoides/farmacologia , Heparina/farmacologia , Humanos , Indóis/farmacologia , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Maleimidas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfolinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-sis , RNA Interferente Pequeno/farmacologia , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Proteínas Recombinantes/farmacologia , Tirfostinas/farmacologia
18.
Trends Cardiovasc Med ; 16(1): 25-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16387627

RESUMO

Platelet-derived growth factor (PDGF) is a major stimulant for smooth muscle cell migration and proliferation, and blockade of PDGF receptors in vivo reduces intimal growth after arterial injury. Signalling by PDGF receptors has been extensively studied and involves multiple signal transduction pathways. These include ras/Extracellular Signal Regulated Kinase (ERK), a pathway critical for controlling cell cycle progression. We have recently discovered that release of fibroblast growth factor 2 and the subsequent activation of fibroblast growth factor receptor 1 are required for maximal induction by PDGF of ERK and of human smooth muscle cell proliferation. This review summarizes our latest findings and discusses the potential implications of this novel signaling mechanism for restenosis.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Becaplermina , Ativação Enzimática/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Humanos , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas c-sis , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Ativação Transcricional/fisiologia
19.
Shock ; 47(6): 666-672, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27922551

RESUMO

Sepsis is an acute life-threatening multiple organ failure caused by a dysregulated host response to infection. Endothelial dysfunction, particularly barrier disruption leading to increased vascular permeability, edema, and insufficient tissue oxygenation, is critical to sepsis pathogenesis. Sphingosine-1-phosphate (S1P) is a signaling lipid that regulates important pathophysiological processes including vascular endothelial cell permeability, inflammation, and coagulation. It is present at high concentrations in blood and lymph and at very low concentrations in tissues due to the activity of the S1P-degrading enzyme S1P-lyase in tissue cells. Recently, four preclinical observational studies determined S1P levels in serum or plasma of sepsis patients, and all found reduced S1P levels associated with the disease. Based on these findings, this review summarizes S1P-regulated processes pertaining to endothelial functions, discusses the possible use of S1P as a marker and possibilities how to manipulate S1P levels and S1P receptor activation to restore endothelial integrity, dampens the inflammatory host response, and improves organ function in sepsis.


Assuntos
Biomarcadores/metabolismo , Lisofosfolipídeos/metabolismo , Sepse/metabolismo , Esfingosina/análogos & derivados , Células Endoteliais/metabolismo , Humanos , Sepse/genética , Transdução de Sinais/fisiologia , Esfingosina/metabolismo
20.
Circ Res ; 94(3): 340-5, 2004 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-14670838

RESUMO

Thrombin and factor Xa (FXa) are agonists for G protein-coupled receptors (GPRCs) and may contribute to vascular lesion formation by stimulating proliferation of vascular smooth muscle cells (SMCs). Mitogenic signaling of GPCRs requires transactivation of receptor tyrosine kinases (RTKs). In rat SMCs, thrombin transactivates the epidermal growth factor receptor (EGFR) via a pathway that involves heparin-binding EGF-like growth factor (HB-EGF) as ligand for EGFR. The purpose of this study was to investigate in human SMCs the role of receptor transactivation in the mitogenic response to thrombin and FXa. Thrombin (10 nmol/L) and FXa (100 nmol/L) cause a 3.3- and 2.6-fold increase in DNA synthesis, respectively. In human SMCs, neither thrombin nor FXa causes EGFR phosphorylation, and blockade of EGFR kinase does not inhibit DNA synthesis. However, DNA synthesis and phosphorylation of fibroblast growth factor receptor-1 (FGFR-1) induced by thrombin or FXa are inhibited by antibodies neutralizing basic fibroblast growth factor (bFGF) or by heparin. Hirudin inhibits thrombin-, but not FXa-induced mitogenesis, indicating that FXa acts independently of thrombin. We further demonstrate by ELISA that upon thrombin and FXa stimulation, bFGF is released and binds to the extracellular matrix. Our data suggest that in human vascular SMCs, both thrombin and FXa rapidly release bFGF into the pericellular matrix. This is followed by transactivation of the FGFR-1 and increased proliferation. Heparin may inhibit the mitogenic effects of thrombin and FXa in human SMCs by preventing bFGF binding to FGFR-1.


Assuntos
DNA/efeitos dos fármacos , Fator Xa/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Trombina/farmacologia , Western Blotting , Células Cultivadas , DNA/biossíntese , Matriz Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor PAR-1/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/genética , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA