Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bosn J Basic Med Sci ; 22(1): 131-139, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34247567

RESUMO

In this study we aimed to develop nomogram models for predicting the overall survival (OS) and cancer-specific survival (CSS) of patients with metaplastic breast cancer (MBC). Data of patients diagnosed with MBC from 1973 to 2015 were collected from the Surveillance, Epidemiology, and End Results (SEER) database. Univariate and multivariate Cox analyses were performed to identify independent prognostic factors for OS and CSS of MBC patients. The obtained prognostic variables were combined to construct nomogram models for predicting OS and CSS in patients with MBC. Model performance was evaluated using concordance index (C-index) and calibration plots. Data from 1125 patients were collected and divided into a training (750) and a validation (375) cohort. The multivariate Cox model identified age, TNM stage, tumor size, and radiotherapy as independent covariates associated with OS and CSS. The nomogram constructed based on these covariates demonstrated excellent accuracy in estimating 3-, and 5-year OS and CSS, with a C-index of 0.769 (95% CI, 0.731-0.808) for OS and 0.761 (95% CI, 0.713-0.809) for CSS in the training cohort. In the validation cohort, the nomogram-predicted C-index was 0.738 (95%CI, 0.676-0.800) for OS and 0.747 (95%CI, 0.667-0.827) for CSS. All calibration curves exhibited good consistency between predicted and actual survival. The nomogram models established in this study may enhance the accuracy of prognosis prediction and therefore may improve individualized assessment of survival risks and enable constructive therapeutic suggestions.


Assuntos
Neoplasias da Mama , Nomogramas , Feminino , Humanos , Prognóstico , Modelos de Riscos Proporcionais , Programa de SEER
2.
Front Oncol ; 11: 704842, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34395277

RESUMO

PURPOSE: To compare survival in different strategies, preoperative systemic treatment versus upfront surgery, in HER2-positive early breast cancer patients in the real world. METHODS: According to the actual upfront treatment, eligible patients from 2012 to 2015 were classified as preoperative systemic treatment or upfront surgery group prospectively. The primary endpoint is disease-free survival; the second endpoint is overall survival. All the outcomes were examined in the propensity score matching model and inverse probability of treatment weighting model. RESULTS: Included in the analysis were 1,067 patients (215 in the preoperative systemic treatment group, 852 in the upfront surgery group). In the propensity score matching model (matching at 1:1 ratio), the disease-free survival of the preoperative systemic treatment group was significantly higher than that of the upfront surgery group (hazard ratio, 0.572, 95%CI, 0.371-0.881, P, 0.012). In the inverse probability of treatment weighting model, there was no significant difference in disease-free survival between the two groups (hazard ratio, 0.946, 95%CI, 0.763-1.172, P, 0.609). For overall survival, there was no significant difference between the two groups. CONCLUSION: The HER2-positive patients who accepted preoperative systemic treatment had better disease-free survival than those who underwent upfront surgery by real-world statistic methods. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov, identifier NCT04249440.

3.
J Am Heart Assoc ; 10(17): e022207, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34459250

RESUMO

Background Interleukin-19 is an immunosuppressive cytokine produced by immune and nonimmune cells, but its role in abdominal aortic aneurysm (AAA) pathogenesis is not known. This study aimed to investigate interleukin-19 expression in, and influences on, the formation and progression of experimental AAAs. Methods and Results Human specimens were obtained at aneurysm repair surgery or from transplant donors. Experimental AAAs were created in 10- to 12-week-old male mice via intra-aortic elastase infusion. Influence and potential mechanisms of interleukin-19 treatment on AAAs were assessed via ultrasonography, histopathology, flow cytometry, and gene expression profiling. Immunohistochemistry revealed augmented interleukin-19 expression in both human and experimental AAAs. In mice, interleukin-19 treatment before AAA initiation via elastase infusion suppressed aneurysm formation and progression, with attenuation of medial elastin degradation, smooth-muscle depletion, leukocyte infiltration, neoangiogenesis, and matrix metalloproteinase 2 and 9 expression. Initiation of interleukin-19 treatment after AAA creation limited further aneurysmal degeneration. In additional experiments, interleukin-19 treatment inhibited murine macrophage recruitment following intraperitoneal thioglycolate injection. In classically or alternatively activated macrophages in vitro, interleukin-19 downregulated mRNA expression of inducible nitric oxide synthase, chemokine C-C motif ligand 2, and metalloproteinases 2 and 9 without apparent effect on cytokine-expressing helper or cytotoxic T-cell differentiation, nor regulatory T cellularity, in the aneurysmal aorta or spleen of interleukin-19-treated mice. Interleukin-19 also suppressed AAAs created via angiotensin II infusion in hyperlipidemic mice. Conclusions Based on human evidence and experimental modeling observations, interleukin-19 may influence the development and progression of AAAs.


Assuntos
Aneurisma da Aorta Abdominal , Interleucinas/uso terapêutico , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/prevenção & controle , Citocinas , Modelos Animais de Doenças , Masculino , Metaloproteinase 2 da Matriz , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática , Proteínas Recombinantes/uso terapêutico
4.
Biosci Rep ; 41(6)2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34042153

RESUMO

BACKGROUND: Long non-coding RNA associated with poor prognosis of hepatocellular carcinoma (AWPPH) is dysregulated in a variety of human cancers. However, the prognostic value of AWPPH in various cancers remains unclear. METHODS: Comprehensive literature search was performed in PubMed, Web of Science, CNKI and Wangfang databases, and eligible studies were obtained according to the inclusion and exclusion criteria. The pooled hazard ratios (HRs) and odds ratios (ORs) were applied to assess the clinical value of AWPPH expression for overall survival (OS) and clinicopathological features. RESULTS: A total of 19 articles including 1699 cancer patients were included in the study. The pooled results demonstrated that evaluated AWPPH expression was positively related to a poorer overall survival of patients with cancers (HR = 1.79, 95%CI: 1.44-2.14, P<0.001). Subgroup analysis revealed that tumor type and sample size affect the predictive value of AWPPH on OS, whereas cut-off value and HR estimation method have no impact on it. In addition, the pooled data also showed that AWPPH was positively linked to advanced TNM stage (OR = 2.50, 95%CI: 1.94-3.22, P<0.001), bigger tumor size (OR = 2.64, 95%CI: 1.47-4.73, P=0.001), macro-vascular invasion (OR = 2.08, 95%CI: 1.04-4.16, P=0.04) and lymph node metastasis (OR = 2.68, 95%CI: 1.82-3.96, P<0.001). Moreover, the results of the trim and fill analysis confirmed the reliability of our finding. CONCLUSIONS: Up-regulation of AWPPH was associated with advanced TNM stage, bigger tumor size, worse lymph node metastasis, macro-vascular invasion and shorter overall survival, suggesting that AWPPH may serve as a biomarker for prognosis and clinicopathological characteristics in human cancers among the Chinese population.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias/genética , RNA Longo não Codificante/genética , Povo Asiático/genética , China/epidemiologia , Intervalo Livre de Doença , Humanos , Neoplasias/etnologia , Neoplasias/mortalidade , Neoplasias/terapia , Valor Preditivo dos Testes , Medição de Risco , Fatores de Risco
5.
Arterioscler Thromb Vasc Biol ; 39(8): 1652-1666, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31294623

RESUMO

OBJECTIVE: We examined the pathogenic significance of VEGF (vascular endothelial growth factor)-A in experimental abdominal aortic aneurysms (AAAs) and the translational value of pharmacological VEGF-A or its receptor inhibition in aneurysm suppression. Approaches and Results: AAAs were created in male C57BL/6J mice via intra-aortic elastase infusion. Soluble VEGFR (VEGF receptor)-2 extracellular ligand-binding domain (delivered in Ad [adenovirus]-VEGFR-2), anti-VEGF-A mAb (monoclonal antibody), and sunitinib were used to sequester VEGF-A, neutralize VEGF-A, and inhibit receptor tyrosine kinase activity, respectively. Influences on AAAs were assessed using ultrasonography and histopathology. In vitro transwell migration and quantitative reverse transcription polymerase chain reaction assays were used to assess myeloid cell chemotaxis and mRNA expression, respectively. Abundant VEGF-A mRNA and VEGF-A-positive cells were present in aneurysmal aortae. Sequestration of VEGF-A by Ad-VEGFR-2 prevented AAA formation, with attenuation of medial elastolysis and smooth muscle depletion, mural angiogenesis and monocyte/macrophage infiltration. Treatment with anti-VEGF-A mAb prevented AAA formation without affecting further progression of established AAAs. Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Additionally, sunitinib treatment reduced circulating monocytes in aneurysmal mice. CONCLUSIONS: VEGF-A and its receptors contribute to experimental AAA formation by suppressing mural angiogenesis, MMP and VEGF-A production, myeloid cell chemotaxis, and circulating monocytes. Pharmacological inhibition of receptor tyrosine kinases by sunitinib or related compounds may provide novel opportunities for clinical aneurysm suppression.


Assuntos
Aneurisma da Aorta Abdominal/etiologia , Elastase Pancreática/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/metabolismo , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Camundongos , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Sunitinibe/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
6.
Curr Drug Targets ; 19(11): 1318-1326, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29359665

RESUMO

BACKGROUND: Abdominal aortic aneurysm (AAA) is a chronic degenerative inflammatory disease. Multi-factors including genetic, environmental and lifestyle factors determine the onsets and progression of AAAs. Currently surgical repair remains the only effective aneurysm treatment, but no pharmacological therapy is available for limiting further enlargement of small AAAs and fetal rupture. OBJECTIVE: This article is to review our current understanding of angiotensin II (Ang II) and its type 1 receptor (AT1) in AAA pathogenesis as well as the translational potential of AT1 receptor blocker (ARB) treatment for treating clinical AAA disease. RESULTS: While many pathways or molecules have been shown to associate with AAA formation and progression, accumulating evidence indicates the most significant importance of peptide hormone Ang II and its receptor AT1 in AAA pathogenesis and suggests the translational value of targeting inhibition of AT1 in treating clinical AAA disease. This review summarized the influences of AT1 deficiency and pharmacological ARB treatment on experimental AAAs. A discussion has also been made on whether and how ARB medication in AAA patients changes the natural course of clinical AAAs, including aneurysm enlargement rate, rupture and AAA-specific mortality. Additionally, we provided information on two registered clinical trials which are to test the efficacy of telmisartan and valsartan in limiting small AAA enlargement. CONCLUSION: Ang II/AT1 pathway plays a critical role in aneurysmal pathogenesis. Targeting AT1 via ARB will help establishing novel pharmacological therapies for limiting continuous enlargement of small AAAs in patients.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Aneurisma da Aorta Abdominal/tratamento farmacológico , Ruptura Aórtica/prevenção & controle , Receptor Tipo 1 de Angiotensina/metabolismo , Angiotensina II/metabolismo , Animais , Aneurisma da Aorta Abdominal/metabolismo , Ruptura Aórtica/metabolismo , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Progressão da Doença , Humanos , Telmisartan/uso terapêutico , Valsartana/uso terapêutico
7.
J Vasc Surg ; 68(5): 1538-1550.e2, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29242064

RESUMO

OBJECTIVE: Mural angiogenesis and macrophage accumulation are two pathologic hallmarks of abdominal aortic aneurysm (AAA) disease. The heterodimeric transcription factor hypoxia-inducible factor 1 (HIF-1) is an essential regulator of angiogenesis and macrophage function. In this study, we investigated HIF-1 expression and activity in clinical and experimental AAA disease. METHODS: Human aortic samples were obtained from 24 AAA patients and six organ donors during open abdominal surgery. Experimental AAAs were created in 10-week-old male C57BL/6J mice by transient intra-aortic infusion of porcine pancreatic elastase (PPE). Expression of HIF-1α and its target gene messenger RNA (mRNA) levels were assessed in aneurysmal and control aortae. The HIF-1α inhibitors 2-methoxyestradiol and digoxin, the prolyl hydroxylase domain-containing protein (PHD) inhibitors cobalt chloride and JNJ-42041935, and the vehicle alone as control were administered daily to mice at varying time points beginning before or after PPE infusion. Influences on experimental AAA formation and progression were assessed by serial transabdominal ultrasound measurements of aortic diameter and histopathologic analysis at sacrifice. RESULTS: The mRNA levels for HIF-1α, vascular endothelial growth factor A, glucose transporter 1, and matrix metalloproteinase 2 were significantly increased in both human and experimental aneurysm tissue. Tissue immunostaining detected more HIF-1α protein in both human and experimental aneurysmal aortae compared with respective control aortae. Treatment with either HIF-1α inhibitor, beginning before or after PPE infusion, prevented enlargement of experimental aneurysms. Both HIF-1α inhibition regimens attenuated medial elastin degradation, smooth muscle cell depletion, and mural angiogenesis and the accumulation of macrophages, T cells, and B cells. Whereas mRNA levels for PHD1 and PHD2 were elevated in experimental aneurysmal aortae, pharmacologic inhibition of PHDs had limited effect on experimental aneurysm progression. CONCLUSIONS: Expression of HIF-1α and its target genes is increased in human and experimental AAAs. Treatment with HIF-1α inhibitors limits experimental AAA progression, with histologic evidence of attenuated mural leukocyte infiltration and angiogenesis. These findings underscore the potential significance of HIF-1α in aneurysm pathogenesis and as a target for pharmacologic suppression of AAA disease.


Assuntos
Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , 2-Metoxiestradiol/farmacologia , Idoso , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/prevenção & controle , Linfócitos T CD4-Positivos/metabolismo , Estudos de Casos e Controles , Quimiotaxia de Leucócito , Digoxina/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neovascularização Patológica , Elastase Pancreática , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Transdução de Sinais , Regulação para Cima
8.
J Vasc Surg ; 67(2): 573-584.e2, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28434702

RESUMO

OBJECTIVE: Angiotensin (Ang) II type 1 receptor (AT1) activation is essential for the development of exogenous Ang II-induced abdominal aortic aneurysms (AAAs) in hyperlipidemic animals. Experimental data derived from this modeling system, however, provide limited insight into the role of endogenous Ang II in aneurysm pathogenesis. Consequently, the potential translational value of AT1 inhibition in clinical AAA disease management remains incompletely understood on the basis of the existing literature. METHODS: AAAs were created in wild-type (WT) and AT1a knockout (KO) mice by intra-aortic infusion of porcine pancreatic elastase (PPE). WT mice were treated with the AT1 receptor antagonist telmisartan, 10 mg/kg/d in chow, or the peroxisome proliferator-activated receptor γ (PPARγ) antagonist GW9662, 3 mg/kg/d through oral gavage, beginning 1 week before or 3 days after PPE infusion. Influences on aneurysm progression as well as mechanistic insights into AT1-mediated pathogenic processes were determined using noninvasive ultrasound imaging, histopathology, aortic gene expression profiling, and flow cytometric analysis. RESULTS: After PPE infusion, aortic enlargement was almost completely abrogated in AT1a KO mice compared with WT mice. As defined by a ≥50% increase in aortic diameter, no PPE-infused, AT1a KO mouse actually developed an AAA. On histologic evaluation, medial smooth muscle cellularity and elastic lamellae were preserved in AT1a KO mice compared with WT mice, with marked attenuation of mural angiogenesis and leukocyte infiltration. In WT mice, telmisartan administration effectively suppressed aneurysm pathogenesis after PPE infusion as well, regardless of whether treatment was initiated before or after aneurysm creation or continued for a limited or extended time. Telmisartan treatment was associated with reduced messenger RNA levels for CCL5 and matrix metalloproteinases 2 and 9 in aneurysmal aortae, with no apparent effect on PPARγ-regulated gene expression. Administration of the PPARγ antagonist GW9662 failed to "rescue" the aneurysm phenotype in telmisartan-treated, PPE-infused WT mice. Neither effector T-cell differentiation nor regulatory T-cell cellularity was affected by telmisartan treatment status. CONCLUSIONS: Telmisartan effectively suppresses the progression of elastase-induced AAAs without apparent effect on PPARγ activation or T-cell differentiation. These findings reinforce the critical importance of endogenous AT1 activation in experimental AAA pathogenesis and reinforce the translational potential of AT1 inhibition in medical aneurysm disease management.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/prevenção & controle , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Elastase Pancreática , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/deficiência , Animais , Aorta Abdominal/diagnóstico por imagem , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Dilatação Patológica , Modelos Animais de Doenças , Regulação da Expressão Gênica , Predisposição Genética para Doença , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptor Tipo 1 de Angiotensina/genética , Transdução de Sinais/efeitos dos fármacos , Telmisartan , Fatores de Tempo , Transcriptoma
9.
Brain Res ; 1650: 172-177, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27608956

RESUMO

Stroke causes brain injury with neuroinflammation which exacerbates the neuronal damage. Recent studies show that anti-inflammatory cytokine interleukin-19 (IL-19) plays a critical part in the inflammatory and ischemic vascular diseases, yet its potential role in ischemic stroke is unknown. Here, we tested the hypothesis that IL-19 exerts protective effects against brain ischemia by modulating inflammation after stroke. Mice were injected intraperitoneally with 10ng/g per day recombinant mouse IL-19 starting pre-stroke, and were subjected to transient middle cerebral artery occlusion. Infarct volume was assessed by triphenyltetrazolium chloride and neurobehavioral outcome by neurological scores. Inflammation was measured using real-time quantitative PCR, immunochemistry, and fluorescence-activated cell sorting. Infarct volume at 72h after stroke was significantly smaller in IL-19 treated group and focal neurological score was significantly better. IL-19 treatment markedly attenuated elevation of the expression of TNF-α and IL-6 mRNA, suppressed increases in the number of microglia, macrophages, CD4+ T cells, CD8+ T cells as well as B cells, and blocked activation of macrophages and neutrophils in the ischemic brain. In peripheral blood, IL-19 injection helped to robustly preserve the reduced immune cells, including macrophages, CD4+ T cells, CD8+ T cells and B cells, compared to control group. IL-19 reduced brain infarction and attenuated neurological deficits following stroke in mice, probably by inhibiting infiltration and activation of immune cells, and by suppressing increases in gene expression of proinflammatory cytokines. This may identify IL-19 as a new therapeutic to limit neuroinflammation after stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Isquemia Encefálica/terapia , Interleucina-10/metabolismo , Interleucina-10/uso terapêutico , Animais , Anti-Inflamatórios/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/terapia , Citocinas/metabolismo , Citometria de Fluxo , Infarto da Artéria Cerebral Média/complicações , Inflamação/metabolismo , Inflamação/terapia , Interleucina-10/imunologia , Interleucina-6/metabolismo , Interleucinas , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Modelos Animais , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/terapia , Fator de Necrose Tumoral alfa/metabolismo
10.
J Vasc Surg ; 64(1): 46-54.e8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27106243

RESUMO

OBJECTIVE: In population-based studies performed on multiple continents during the past two decades, diabetes mellitus has been negatively associated with the prevalence and progression of abdominal aortic aneurysm (AAA) disease. We investigated the possibility that metformin, the primary oral hypoglycemic agent in use worldwide, may influence the progression of AAA disease. METHODS: Preoperative AAA patients with diabetes were identified from an institutional database. After tabulation of individual cardiovascular and demographic risk factors and prescription drug regimens, odds ratios for categorical influences on annual AAA enlargement were calculated through nominal logistical regression. Experimental AAA modeling experiments were subsequently performed in normoglycemic mice to validate the database-derived observations as well as to suggest potential mechanisms of metformin-mediated aneurysm suppression. RESULTS: Fifty-eight patients met criteria for study inclusion. Of 11 distinct classes of medication considered, only metformin use was negatively associated with AAA enlargement. This association remained significant after controlling for gender, age, cigarette smoking status, and obesity. The median enlargement rate in AAA patients not taking oral diabetic medication was 1.5 mm/y; by nominal logistic regression, metformin, hyperlipidemia, and age ≥70 years were associated with below-median enlargement, whereas sulfonylurea therapy, initial aortic diameter ≥40 mm, and statin use were associated with above-median enlargement. In experimental modeling, metformin dramatically suppressed the formation and progression, with medial elastin and smooth muscle preservation and reduced aortic mural macrophage, CD8 T cell, and neovessel density. CONCLUSIONS: Epidemiologic evidence of AAA suppression in diabetes may be attributable to concurrent therapy with the oral hypoglycemic agent metformin.


Assuntos
Aneurisma da Aorta Abdominal/prevenção & controle , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Metformina/administração & dosagem , Administração Oral , Idoso , Animais , Aneurisma da Aorta Abdominal/diagnóstico por imagem , Aneurisma da Aorta Abdominal/epidemiologia , California/epidemiologia , Mineração de Dados , Bases de Dados Factuais , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Modelos Logísticos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Razão de Chances , Fatores de Proteção , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo
11.
Arterioscler Thromb Vasc Biol ; 33(4): 718-26, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23288157

RESUMO

OBJECTIVE: Macrophages are critical contributors to abdominal aortic aneurysm (AAA) disease. We examined the ability of MKEY, a peptide inhibitor of CXCL4-CCL5 interaction, to influence AAA progression in murine models. APPROACH AND RESULTS: AAAs were created in 10-week-old male C57BL/6J mice by transient infrarenal aortic porcine pancreatic elastase infusion. Mice were treated with MKEY via intravenous injection either (1) before porcine pancreatic elastase infusion or (2) after aneurysm initiation. Immunostaining demonstrated CCL5 and CCR5 expression on aneurysmal aortae and mural monocytes/macrophages, respectively. MKEY treatment partially inhibited migration of adaptively transferred leukocytes into aneurysmal aortae in recipient mice. Although all vehicle-pretreated mice developed AAAs, aneurysms formed in only 60% (3/5) and 14% (1/7) of mice pretreated with MKEY at 10 and 20 mg/kg, respectively. MKEY pretreatment reduced aortic diameter enlargement, preserved medial elastin fibers and smooth muscle cells, and attenuated mural macrophage infiltration, angiogenesis, and aortic metalloproteinase 2 and 9 expression after porcine pancreatic elastase infusion. MKEY initiated after porcine pancreatic elastase infusion also stabilized or reduced enlargement of existing AAAs. Finally, MKEY treatment was effective in limiting AAA formation after angiotensin II infusion in apolipoprotein E-deficient mice. CONCLUSIONS: MKEY suppresses AAA formation and progression in 2 complementary experimental models. Peptide inhibition of CXCL4-CCL5 interactions may represent a viable translational strategy to limit progression of human AAA disease.


Assuntos
Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/prevenção & controle , Quimiocina CCL5/antagonistas & inibidores , Oligopeptídeos/farmacologia , Fator Plaquetário 4/antagonistas & inibidores , Angiotensina II , Animais , Aorta Abdominal/imunologia , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Injeções Intravenosas , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Oligopeptídeos/administração & dosagem , Elastase Pancreática , Fator Plaquetário 4/metabolismo , Receptores CCR5/metabolismo , Fatores de Tempo
12.
Ann Clin Lab Sci ; 36(1): 39-46, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16501235

RESUMO

The objective of this study was to investigate whether corticosteroids modulate neuropathic pain by altering cerebral expression of nuclear factor-kappa B (NF-kappaB) and specific cytokines. The effects of topical betamethasone on neuropathic pain and cerebral expression of NF-kappaB and cytokines were studied in a rat model of L5 spinal nerve transaction. Behavioral testing was undertaken on days 1, 3, 7, 14, and 21 post-operation using the von Frey and Hargreaves tests. NF-kappaB activation in the brain was investigated by an electrophoretic mobility shift assay (EMSA), and cerebral expressions of tumor necrosis factor-alpha (TNFalpha), interleukin-1beta (IL-1beta), and interleukin-10 (IL-10) were quantified using enzyme-linked immunosorbent assays (ELISA). Spinal nerve transection induced mechanical allodynia and thermal hyperalgesia, which were significantly ameliorated by topical injection of betamethasone around the site of injury. In addition, betamethasone reduced the activation of NF-kappaB and elevation of TNFalpha and IL-1beta, and induced the expression of IL-10 in the brain, all of which correlated with the changes of pain thresholds in rats. The results suggest that topical betamethasone injection inhibits the development and maintenance of neuropathic pain. Betamethasone may act by regulating the expression of NF-kappaB, TNFalpha, IL-1beta and IL-10 in the brain. This study yields new insight into the mechanisms of corticosteroid action in neuropathic pain and may provide a basis for clinical pain control.


Assuntos
Betametasona/farmacologia , Encéfalo/metabolismo , Glucocorticoides/farmacologia , NF-kappa B/biossíntese , Dor/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Interleucina-1/biossíntese , Interleucina-10/biossíntese , Ligadura , Masculino , Dor/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/fisiopatologia , Ratos , Ratos Sprague-Dawley , Nervos Espinhais , Fator de Necrose Tumoral alfa/biossíntese
13.
Neurosci Lett ; 393(2-3): 255-9, 2006 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-16253423

RESUMO

Glucocorticoids have been used to treat neuropathic pain for many years, but the underlying mechanisms are still unknown. Recent studies indicate that pathological pain states may be mediated by cytokines. We, therefore, examined the effect of betamethasone on neuropathic pain and the relationship between pain behavior and the expression of cytokines in the brain. Rats were given epidural injections of betamethasone (Diprospan) after L5 spinal nerve transection. Mechanical allodynia and thermal hyperalgesia were evaluated on post-operative days 1, 3, 7, 14 and 21 with von Frey and Hargreaves tests. Cerebral expression of NF-kappaB, TNFalpha, IL-1beta and IL-10 was quantified using electrophoretic mobility shift assay (EMSA) or enzyme-linked immunosorbent assay (ELISA). We found that spinal nerve injury caused long-lasting mechanical and thermal hyperalgesia in the hind paw and stimulated the expression of NF-kappaB, TNFalpha, IL-1beta and IL-10 in the brain. A single epidural injection of betamethasone at the time of nerve injury partially inhibited the development of neuropathic hyperalgesia and reduced the subsequent elevated levels of pro-inflammatory cytokines in the brain, while stimulating the expression of the anti-inflammatory cytokine IL-10. These data support our hypothesis that pro-inflammatory cytokines in the brain may mediate the hyperalgesic effects of spinal nerve injury and that the long-acting anti-hyperalgesic effects of epidural glucocorticoid treatment are due to an inhibitory effect on pro-inflammatory cytokine levels and a stimulatory effect on anti-inflammatory cytokine levels in the brain.


Assuntos
Anti-Inflamatórios/uso terapêutico , Betametasona/uso terapêutico , Córtex Cerebral/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Dor/tratamento farmacológico , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Betametasona/administração & dosagem , Betametasona/análogos & derivados , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Modelos Animais de Doenças , Combinação de Medicamentos , Masculino , Dor/etiologia , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/complicações , Fatores de Tempo , Quinase Induzida por NF-kappaB
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...