Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nutr Neurosci ; 25(3): 519-529, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32420816

RESUMO

INTRODUCTION: Parkinson's disease (PD) is a chronic neurological disorder whose pathogenesis involves the loss of dopaminergic neurons and dopamine terminals, formation of Lewy bodies, and microgliosis. Its treatment includes dopamine-based drugs with limited results and adverse effects. Additionally, some neuroleptic drugs used for mental disorders produce side effects referred to as parkinsonism. Dietary interventions with ω-3 polyunsaturated fatty acids (ω-3 PUFA) have attracted attention since they play a key role in most of the processes associated with PD etiology. OBJECTIVE: The purpose of our work was to investigate the effects of an ω-3 PUFA rich algal oil on locomotive alterations induced by haloperidol and D2 receptor protein and gene expression in Wistar rats. METHODOLOGY: Pre- and co-supplementation of algal oil (300 mg of ω-3 FA/kg/day for six weeks) and haloperidol (1.5 mg/kg/day for two weeks) were evaluated. RESULTS: Haloperidol provoked locomotive alterations in the Open Field Test and a 43% diminution in D2 receptor in brain membranes; in pre-supplemented rats a 93% increase in D2 receptor protein expression and a partial maintenance of locomotory performance were observed, while in co-supplemented rats D2 receptor protein expression was maintained as in control rats, although locomotive behavior was found diminished as in haloperidol rats. CONCLUSIONS: These results confirm the beneficial effects of ω-3 PUFA over locomotory alterations and as neuroprotective and neurorestorative compounds and demonstrates a stimulatory action on D2 receptor presence, as a mechanism by which these fatty acids participate in brain health.


Assuntos
Ácidos Graxos Ômega-3 , Transtornos Parkinsonianos , Animais , Dopamina/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Haloperidol , Humanos , Ratos , Ratos Wistar
2.
Basic Res Cardiol ; 110(2): 8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25604439

RESUMO

Chronic activation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway contributes to vascular inflammation and atherosclerosis by inducing expression of genes involved in cell proliferation, differentiation and migration. We aimed to investigate whether enforced expression of negative regulators, the suppressors of cytokine signaling (SOCS1 and SOCS3), inhibits harmful JAK/STAT-mediated responses and affects atherosclerosis in apolipoprotein E knockout mice. Adenovirus-mediated SOCS1 transgene expression impaired the onset and progression of atherosclerosis without impact on lipid profile, whereas SOCS3 was only effective on early atherosclerosis. Mechanistically, SOCS gene delivery, primarily SOCS1, attenuated STAT1 and STAT3 activation and reduced the expression of STAT-dependent genes (chemokine/chemokine receptors, adhesion molecules, pro-inflammatory cytokines and scavenger receptors) in aortic tissue. Furthermore, atherosclerotic plaques exhibit a more stable phenotype characterized by lower lipids, T cells and M1 macrophages and higher M2 macrophages and collagen. Atheroprotection was accompanied by a systemic alteration of T helper- and T regulatory-related genes and a reduced activation state of circulating monocytes. In vascular smooth muscle cells and macrophages, SOCS gene delivery inhibited cytokine-induced STAT activation, pro-inflammatory gene expression, cell migration and proliferation. In conclusion, targeting SOCS proteins, predominantly SOCS1, to suppress pathological mechanisms involved in atheroma plaque progression and destabilization could be an interesting anti-atherosclerotic strategy.


Assuntos
Aterosclerose/patologia , Inflamação/patologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Inflamação/genética , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Supressoras da Sinalização de Citocina/genética , Transdução Genética
3.
Blood ; 121(18): 3768-77, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23479571

RESUMO

The mechanisms by which CC chemokine receptor (CCR)7 ligands are selectively presented on lymphatic endothelium in the presence of inflammatory chemokines are poorly understood. The chemokine-scavenging receptor D6 is expressed on lymphatic endothelial cells (LEC) and contributes to selective presentation of CCR7 ligands by suppressing inflammatory chemokine binding to LEC surfaces. As well as preventing inappropriate inflammatory cell attachment to LECs, D6 is specifically involved in regulating the ability of LEC to discriminate between mature and immature dendritic cells (DCs). D6 overexpression reduces immature DC (iDC) adhesion to LECs, whereas D6 knockdown increases adhesion of iDCs that displace mature DCs. LEC D6 expression is regulated by growth factors, cytokines, and tumor microenvironments. In particular, interleukin-6 and interferon-γ are potent inducers, indicating a preferential role for D6 in inflamed contexts. Expression of the viral interleukin-6 homolog from Kaposi sarcoma-associated herpesvirus is also sufficient to induce significant D6 upregulation both in vitro and in vivo, and Kaposi sarcoma and primary effusion lymphoma cells demonstrate high levels of D6 expression. We therefore propose that D6, which is upregulated in both inflammatory and tumor contexts, is an essential regulator of inflammatory leukocyte interactions with LECs and is required for immature/mature DC discrimination by LECs.


Assuntos
Células Endoteliais/metabolismo , Receptores CCR10/genética , Receptores CCR10/fisiologia , Animais , Células CHO , Comunicação Celular/genética , Comunicação Celular/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Cricetinae , Cricetulus , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/fisiologia , Células Endoteliais/imunologia , Células HEK293 , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores CCR10/análise , Receptores CCR10/metabolismo , Transfecção , Receptor D6 de Quimiocina
4.
Am J Pathol ; 182(5): 1910-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23597852

RESUMO

Atherosclerosis is a chronic inflammatory disease of the arterial wall. NF-κB is a major regulator of inflammation that controls the expression of many genes involved in atherogenesis. Activated NF-κB was detected in human atherosclerotic plaques, and modulation of NF-κB inflammatory activity limits disease progression in mice. Herein, we investigate the anti-inflammatory and atheroprotective effects of a cell-permeable peptide containing the NF-κB nuclear localization sequence (NLS). In vascular smooth muscle cells and macrophages, NLS peptide specifically blocked the importin α-mediated nuclear import of NF-κB and prevented lipopolysaccharide-induced pro-inflammatory gene expression, cell migration, and oxidative stress. In experimental atherosclerosis (apolipoprotein E-knockout mice fed a high-fat diet), i.p., 0.13 µmol/day NLS peptide administration for 5 weeks attenuated NF-κB activation in atherosclerotic plaques. NLS peptide significantly inhibited lesion development at both early (age 10 weeks) and advanced (age 28 weeks) stages of atherosclerosis in mice, without affecting serum lipid levels. Plaques from NLS-treated mice contained fewer macrophages of pro-inflammatory M1 subtype than those from respective untreated controls. By contrast, the relative smooth muscle cell and collagen content was increased, indicating a more stable plaque phenotype. NLS peptide also attenuated pro-inflammatory gene expression and oxidative stress in aortic lesions. Our study demonstrates that targeting NF-κB nuclear translocation hampers inflammation and atherosclerosis development and identifies cell-permeable NLS peptide as a potential anti-atherosclerotic agent.


Assuntos
Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/induzido quimicamente , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Carioferinas/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Sinais de Localização Nuclear/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Placa Aterosclerótica , Transporte Proteico/efeitos dos fármacos
5.
Front Pharmacol ; 15: 1377113, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38666016

RESUMO

The renin-angiotensin system (RAS) is an important cascade of enzymes and peptides that regulates blood pressure, volume, and electrolytes. Within this complex system of reactions, its counter-regulatory axis has attracted attention, which has been associated with the pathophysiology of inflammatory and fibrotic diseases. This review article analyzes the impact of different components of the counter-regulatory axis of the RAS on different pathologies. Of these peptides, Angiotensin-(1-7), angiotensin-(1-9) and alamandine have been evaluated in a wide variety of in vitro and in vivo studies, where not only they counteract the actions of the classical axis, but also exhibit independent anti-inflammatory and fibrotic actions when binding to specific receptors, mainly in heart, kidney, and lung. Other functional peptides are also addressed, which despite no reports associated with inflammation and fibrosis to date were found, they could represent a potential target of study. Furthermore, the association of agonists of the counter-regulatory axis is analyzed, highlighting their contribution to the modulation of the inflammatory response counteracting the development of fibrotic events. This article shows an overview of the importance of the RAS in the resolution of inflammatory and fibrotic diseases, offering an understanding of the individual components as potential treatments.

6.
Brain ; 135(Pt 9): 2826-37, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22961553

RESUMO

Alzheimer's disease is a severely debilitating disease of high and growing proportions. Hypercholesterolaemia is a key risk factor in sporadic Alzheimer's disease that links metabolic disorders (diabetes, obesity and atherosclerosis) with this pathology. Hypercholesterolaemia is associated with increased levels of immunoglobulin G against oxidized lipoproteins. Patients with Alzheimer's disease produce autoantibodies against non-brain antigens and specific receptors for the constant Fc region of immunoglobulin G have been found in vulnerable neuronal subpopulations. Here, we focused on the potential role of Fc receptors as pathological players driving hypercholesterolaemia to Alzheimer's disease. In a well-established model of hypercholesterolaemia, the apolipoprotein E knockout mouse, we report increased brain levels of immunoglobulin G and upregulation of activating Fc receptors, predominantly of type IV, in neurons susceptible to amyloid ß accumulation. In these mice, gene deletion of γ-chain, the common subunit of activating Fc receptors, prevents learning and memory impairments without influencing cholesterolaemia and brain and serum immunoglobulin G levels. These cognition-protective effects were associated with a reduction in synapse loss, tau hyperphosphorylation and intracellular amyloid ß accumulation both in cortical and hippocampal pyramidal neurons. In vitro, activating Fc receptor engagement caused synapse loss, tau hyperphosphorylation and amyloid ß deposition in primary neurons by a mechanism involving mitogen-activated protein kinases and ß-site amyloid precursor protein cleaving enzyme 1. Our results represent the first demonstration that immunoglobulin G Fc receptors contribute to the development of hypercholesterolaemia-associated features of Alzheimer's disease and suggest a new potential target for slowing or preventing Alzheimer's disease in hypercholesterolaemic patients.


Assuntos
Doença de Alzheimer/genética , Transtornos Cognitivos/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de IgG/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Comportamento Animal/fisiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Hipocampo/patologia , Camundongos , Camundongos Knockout , Neurônios/patologia , Receptores de IgG/metabolismo
7.
J Am Soc Nephrol ; 23(9): 1518-27, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22859852

RESUMO

Among patients with diabetes, increased production of immunoglobulins against proteins modified by diabetes is associated with proteinuria and cardiovascular risk, suggesting that immune mechanisms may contribute to the development of diabetes complications, such as nephropathy. We investigated the contribution of IgG Fcγ receptors to diabetic renal injury in hyperglycemic, hypercholesterolemic mice. We used streptozotocin to induce diabetes in apolipoprotein E-deficient mice and in mice deficient in both apolipoprotein E and γ-chain, the common subunit of activating Fcγ receptors. After 15 weeks, the mice lacking Fcγ receptors had significantly less albuminuria and renal hypertrophy, despite similar degrees of hyperglycemia and hypercholesterolemia, immunoglobulin production, and glomerular immune deposits. Moreover, diabetic Fcγ receptor-deficient mice had less mesangial matrix expansion, inflammatory cell infiltration, and collagen and α-smooth muscle actin content in their kidneys. Accordingly, expression of genes involved in leukocyte infiltration, fibrosis, and oxidative stress was significantly reduced in diabetic kidneys and in mesangial cells cultured from Fcγ receptor-deficient mice. In summary, preventing the activation of Fcγ receptors alleviates renal hypertrophy, inflammation, and fibrosis in hypercholesterolemic mice with diabetes, suggesting that modulating Fcγ receptor signaling may be renoprotective in diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/prevenção & controle , Receptores de IgG/deficiência , Estreptozocina/efeitos adversos , Actinas/metabolismo , Albuminúria/epidemiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Colágeno/metabolismo , Nefropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Hipertrofia/epidemiologia , Incidência , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/genética , Transdução de Sinais/fisiologia
8.
Mol Ther Nucleic Acids ; 34: 102041, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37842165

RESUMO

Diabetic kidney disease (DKD) is a common microvascular complication of diabetes, a global health issue. Hyperglycemia, in concert with cytokines, activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway to induce inflammation and oxidative stress contributing to renal damage. There is evidence of microRNA-155 (miR-155) involvement in diabetes complications, but the underlying mechanisms are unclear. In this study, gain- and loss-of-function experiments were conducted to investigate the interplay between miR-155-5p and suppressor of cytokine signaling 1 (SOCS1) in the regulation of the JAK/STAT pathway during renal inflammation and DKD. In experimental models of mesangial injury and diabetes, miR-155-5p expression correlated inversely with SOCS1 and positively with albuminuria and expression levels of cytokines and prooxidant genes. In renal cells, miR-155-5p mimic downregulated SOCS1 and promoted STAT1/3 activation, cytokine expression, and cell proliferation and migration. Conversely, both miR-155-5p antagonism and SOCS1 overexpression protected cells from inflammation and hyperglycemia damage. In vivo, SOCS1 gene delivery decreased miR-155-5p and kidney injury in diabetic mice. Moreover, therapeutic inhibition of miR-155-5p suppressed STAT1/3 activation and alleviated albuminuria, mesangial damage, and renal expression of inflammatory and fibrotic genes. In conclusion, modulation of the miR-155/SOCS1 axis protects kidneys against diabetic damage, thus highlighting its potential as therapeutic target for DKD.

9.
Endocrinol Diabetes Nutr (Engl Ed) ; 69(3): 168-177, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35396115

RESUMO

BACKGROUND: The consumption of artificially sweetened beverages (ASBs) has been linked to metabolic alterations. The effect of reducing the regular consumption of these beverages on the metabolism is currently unknown. OBJECTIVE: To evaluate the effect of reducing consumption of ASBs on the metabolism in overweight young adults. DESIGN: A randomised, single-blind, controlled, 12-week, clinical trial was performed in overweight young adults who regularly consume ASBs. The 45 subjects who participated in the study were randomly divided into two groups: (1) control group (n=21) and (2) intervention group (no intake of ASBs, n=24). Body weight and composition, fasting plasma concentrations of glucose, triglycerides, insulin, cholesterol, low-density lipoproteins and high-density lipoproteins were measured at the beginning and end of the study. and the HOMA-IR was calculated. RESULTS: At the end of 12 weeks, the intervention group showed a significant decrease (as opposed to an increase in the control group) in the percentage of change in body weight (-1.22% vs 1.31%, p<0.004), body fat (-6.28% vs 6.15%, p<0.001) and insulin resistance index (-12.06 vs 38.21%, p<0.00002), as well as in levels of glucose (-4.26% vs 0.51%, p<0.05), triglycerides (-14.74% vs 19.90%, p<0.006), insulin (-8.02% vs 39.23%, p<0.00005), cholesterol (-8.71% vs 0.77%, p<0.01) and LDL (-9.46% vs 9.92%, p<0.004). CONCLUSION: A reduction in habitual consumption of ASBs in overweight young adults decreases biochemical measurements, body weight and composition, suggesting a participation in the metabolic processes.


Assuntos
Sobrepeso , Edulcorantes , Bebidas Adoçadas Artificialmente , Peso Corporal , Fatores de Risco Cardiometabólico , Colesterol , Glucose , Humanos , Insulina , Método Simples-Cego , Edulcorantes/efeitos adversos , Triglicerídeos , Adulto Jovem
10.
J Am Soc Nephrol ; 21(5): 763-72, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20185635

RESUMO

Activation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) is an important mechanism by which hyperglycemia contributes to renal damage, suggesting that modulation of this pathway may prevent renal and vascular complications of diabetes. Here, we investigated the involvement of suppressors of cytokine signaling (SOCS) as intracellular negative regulators of JAK/STAT activation in diabetic nephropathy. In a rat model, inducing diabetes resulted in JAK/STAT activation and increased expression of SOCS1 and SOCS3. In humans, we observed increased expression of glomerular and tubulointerstitial SOCS proteins in biopsies of patients with diabetic nephropathy. In vitro, high concentrations of glucose activated JAK/STAT/SOCS in human mesangial and tubular cells. Overexpression of SOCS reversed the glucose-induced activation of the JAK/STAT pathway, expression of STAT-dependent genes (chemokines, growth factors, and extracellular matrix proteins), and cell proliferation. In vivo, intrarenal delivery of adenovirus expressing SOCS1 and SOCS3 to diabetic rats significantly improved renal function and reduced renal lesions associated with diabetes, such as mesangial expansion, fibrosis, and influx of macrophages. SOCS gene delivery also decreased the activation of STAT1 and STAT3 and the expression of proinflammatory and profibrotic proteins in the diabetic kidney. In summary, these results provide direct evidence for a link between the JAK/STAT/SOCS axis and hyperglycemia-induced cell responses in the kidney. Suppression of the JAK/STAT pathway by increasing intracellular SOCS proteins may have therapeutic potential in diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/metabolismo , Hiperglicemia/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adenoviridae , Idoso , Animais , Estudos de Casos e Controles , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Nefropatias Diabéticas/terapia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Wistar
11.
Front Neurosci ; 15: 579263, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33790729

RESUMO

Hepatic encephalopathy (HE) is one of the most disabling metabolic diseases. It consists of a complication of liver disease through the action of neurotoxins, such as excessive production of ammonia from liver, resulting in impaired brain function. Its prevalence and incidence are not well known, although it has been established that up to 40% of cirrhotic patients may develop HE. Patients with HE episodes display a wide range of neurological disturbances, from subclinical alterations to coma. Recent evidence suggests that the resolution of hepatic encephalopathy does not fully restore cognitive functioning in cirrhotic patients. Therefore, the aim of this review was to evaluate the evidence supporting the presence of lingering cognitive deficits in patients with a history of HE compared to patients without HE history and how liver transplant affects such outcome in these patients. We performed two distinct meta-analysis of continuous outcomes. In both cases the results were pooled using random-effects models. Our results indicate that cirrhotic patients with a history of HE show clear cognitive deficits compared control cirrhotic patients (Std. Mean Difference (in SDs) = -0.72 [CI 95%: -0.94, -0.50]) and that these differences are not fully restored after liver transplant (Std. Mean Difference (in SDs) = -0.72 [CI 95%: -0.94, -0.50]).

12.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-34274304

RESUMO

BACKGROUND: The consumption of artificially sweetened beverages (ASBs) has been linked to metabolic alterations. The effect of reducing the regular consumption of these beverages on the metabolism is currently unknown. OBJECTIVE: To evaluate the effect of reducing consumption of ASBs on the metabolism in overweight young adults. DESIGN: A randomised, single-blind, controlled, 12-week, clinical trial was performed in overweight young adults who regularly consume ASBs. The 45 subjects who participated in the study were randomly divided into two groups: (1) control group (n=21) and (2) intervention group (no intake of ASBs, n=24). Body weight and composition, fasting plasma concentrations of glucose, triglycerides, insulin, cholesterol, low-density lipoproteins and high-density lipoproteins were measured at the beginning and end of the study. and the HOMA-IR was calculated. RESULTS: At the end of 12 weeks, the intervention group showed a significant decrease (as opposed to an increase in the control group) in the percentage of change in body weight (-1.22% vs 1.31%, p<0.004), body fat (-6.28% vs 6.15%, p<0.001) and insulin resistance index (-12.06 vs 38.21%, p<0.00002), as well as in levels of glucose (-4.26% vs 0.51%, p<0.05), triglycerides (-14.74% vs 19.90%, p<0.006), insulin (-8.02% vs 39.23%, p<0.00005), cholesterol (-8.71% vs 0.77%, p<0.01) and LDL (-9.46% vs 9.92%, p<0.004). CONCLUSION: A reduction in habitual consumption of ASBs in overweight young adults decreases biochemical measurements, body weight and composition, suggesting a participation in the metabolic processes.

14.
Arterioscler Thromb Vasc Biol ; 29(4): 525-31, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19164812

RESUMO

OBJECTIVE: Suppressors of cytokine signaling (SOCS) proteins are intracellular regulators of receptor signal transduction, mainly Janus kinase/signal transducers and activators of transcription (JAK/STAT). We investigated the effects of SOCS modulation on the JAK/STAT-dependent responses in vascular cells, and their implication in atherosclerotic plaque development. METHODS AND RESULTS: Immunohistochemistry in human plaques revealed a high expression of SOCS1 and SOCS3 by vascular smooth muscle cells (VSMCs) and macrophages in the inflammatory region of the shoulders, when compared to the fibrous area. SOCS were also increased in aortic lesions from apoE(-/-) mice. In cultured VSMCs, endothelial cells, and monocytes, SOCS1 and SOCS3 were transiently induced by proinflammatory cytokines, proatherogenic lipoproteins, and immune molecules. Furthermore, overexpression of SOCS suppressed STAT activation and reduced inflammatory gene expression and cell growth, whereas SOCS knockdown increased these cell responses. In vivo, antisense oligodeoxynucleotides targeting SOCS3 exacerbated the atherosclerotic process in apoE(-/-) mice by increasing the size, leukocyte content, and chemokine expression in the lesions. CONCLUSIONS: SOCS expressed in atherosclerotic lesions are key regulators of vascular cell responses. Activation of this endogenous antiinflammatory pathway might be of interest in the treatment of atherosclerosis.


Assuntos
Aterosclerose/enzimologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Aorta/enzimologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/patologia , Artérias Carótidas/enzimologia , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/enzimologia , Fosforilação , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Tempo , Transfecção
15.
Arterioscler Thromb Vasc Biol ; 29(12): 2061-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19778942

RESUMO

OBJECTIVE: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily of cytokines. TWEAK binds and activates the Fn14 receptor, and may regulate apoptosis, inflammation, and angiogenesis, in different pathological conditions. We have evaluated the effect of exogenous TWEAK administration as well as the role of endogenous TWEAK on proinflammatory cytokine expression and vascular and renal injury severity in hyperlipidemic ApoE-knockout mice. METHODS AND RESULTS: ApoE(-/-) mice were fed with hyperlipidemic diet for 4 to 10 weeks, then randomized and treated with saline (controls), TWEAK (10 microg/kg/d), anti-TWEAK neutralizing mAb (1000 microg/kg/d), TWEAK plus anti-TWEAK antibody (10 microg TWEAK +1000 microg anti-TWEAK/kg/d), or nonspecific IgG (1000 microg/kg/d) daily for 9 days. In ApoE(-/-) mice, exogenous TWEAK administration in ApoE(-/-) mice induced activation of NF-kappaB, a key transcription factor implicated in the regulation of the inflammatory response, in vascular and renal lesions. Furthermore, TWEAK treatment increased chemokine expression (RANTES and MCP-1), as well as macrophage infiltration in atherosclerotic plaques and renal lesions. These effects were associated with exacerbation of vascular and renal damage. Conversely, treatment of ApoE(-/-) mice with an anti-TWEAK blocking mAb decreased NF-kappaB activation, proinflammatory cytokine expression, macrophage infiltration, and vascular and renal injury severity, indicating a pathological role for endogenous TWEAK. Finally, in murine vascular smooth muscle cells or tubular cells, either ox-LDL or TWEAK treatment increased expression and secretion of both RANTES and MCP-1. Furthermore, ox-LDL and TWEAK synergized for induction of MCP-1 and RANTES expression and secretion. CONCLUSIONS: Our results suggest that TWEAK exacerbates the inflammatory response associated with a high lipid-rich diet. TWEAK may be a novel therapeutic target to prevent vascular and renal damage associated with hyperlipidemia.


Assuntos
Apolipoproteínas E/deficiência , Vasos Sanguíneos/lesões , Hiperlipidemias/patologia , Hiperlipidemias/fisiopatologia , Rim/lesões , Fatores de Necrose Tumoral/fisiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Citocina TWEAK , Citocinas/genética , Dieta Aterogênica , Expressão Gênica/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Lipoproteínas LDL/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Recombinantes/farmacologia , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/imunologia , Fatores de Necrose Tumoral/farmacologia
16.
Phytomedicine ; 63: 153016, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31325683

RESUMO

BACKGROUND: Recently, the study of marine natural products has gained interest due to their relevant biological activities. Specially, seaweeds produce bioactive compounds that could act as modulators of cell signaling pathways involved in a plethora of diseases. Thereby, the description of the molecular mechanisms by which seaweeds elicit its biological functions will certainly pave the way to the pharmacological development of drugs. AIM: This review describes the molecular mechanisms by which seaweeds act and its possible utilization in the design of new drugs. METHODS: This review was conducted according to the PRISMA-P guidelines for systematic reviews. Two independent authors searched into four different databases using combinations of keywords. Two more authors selected the articles following the eligibility criteria. Information extraction was conducted by two separated authors and entered into spreadsheets. Methodological quality and risk of bias were determined applying a 12-question Risk of Bias criteria tool. RESULTS AND DISCUSSION: We found 2360 articles (SCOPUS: 998; PubMed: 678; Wiley: 645 and EBSCO: 39) using the established keywords, of which 113 articles fit the inclusion criteria and were included in the review. This work comprises studies in cell lines, and animal models, any clinical trial was excluded. The articles were published from 2005 up to March 31st 2018. The biggest amount of articles was published in 2017. Furthermore, the seaweeds tested in the studies were collected in 15 countries, mainly in Eastern countries. We found that the main modulated signaling pathways by seaweeds-derivate extracts and compounds were: L-Arginine/NO, TNF-α, MAPKs, PI3K/AKT/GSK, mTOR, NF-κB, extrinsic and intrinsic apoptosis, cell cycle, MMPs and Nrf2. Finally, the articles we analyzed showed moderate risk of bias in almost all the parameters evaluated. However, the studies fail to describe the place and characteristics of sample collection, the sample size, and the blindness of the experimental design. CONCLUSION: In this review we identified and summarized relevant information related to seaweed-isolated compounds and extracts having biological activity; their role in different signal pathways to better understand their potential to further development of cures for cancer, diabetes, and inflammation-related diseases.


Assuntos
Preparações de Plantas/farmacologia , Alga Marinha/química , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Hipoglicemiantes/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Preparações de Plantas/química , Fator de Necrose Tumoral alfa/metabolismo
17.
Circ Res ; 99(11): 1188-96, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17053192

RESUMO

IgG Fc receptors (FcgammaRs) play a role in activating the immune system and in maintaining peripheral tolerance, but their role in atherosclerosis is unknown. We generated double-knockout (DKO) mice by crossing apolipoprotein E-deficient mice (apoE(-/-)) with FcgammaR gamma chain-deficient mice (gamma(-/-)). The size of atherosclerotic lesions along the aorta was approximately 50% lower in DKO compared with apoE(-/-) control mice, without differences in serum lipid levels. The macrophage and T-cell content of lesions in the DKO were reduced by 49+/-6% and 56+/-8%, respectively, compared with the content in apoE(-/-) lesions. Furthermore, the expression of monocyte chemoattractant protein-1 (MCP-1), RANTES (Regulated on Activated Normal T-cell Expressed and Secreted), and intercellular adhesion molecule-1 (ICAM-1) and the activation of nuclear factor-kappaB (NF-kappaB) were significantly reduced in aortic lesions from DKO mice. In vitro, vascular smooth muscle cells (VSMCs) from both gamma(-/-) and DKO mice failed to respond to immune complexes, as shown by impaired chemokine expression and NF-kappaB activation. ApoE(-/-) mice have higher levels of activating FcgammaRI and FcgammaRIIIA, and inhibitory FcgammaRIIB, compared with wild-type mice. The DKO mice express only the inhibitory FcgammaRIIB receptor. We conclude that FcgammaR deficiency limits development and progression of atherosclerosis. In addition to leukocytes, FcgammaR activation in VSMCs contributes to the inflammatory process, in part, by regulating chemokine expression and leukocyte invasion of the vessel wall. These results underscore the critical role of FcgammaRs in atherogenesis and support the use of immunotherapy in the treatment of this disease.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Receptores de IgG/deficiência , Animais , Complexo Antígeno-Anticorpo/metabolismo , Aterosclerose/etiologia , Células Cultivadas , Quimiocinas/metabolismo , Regulação para Baixo , Feminino , Expressão Gênica , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Receptores de IgG/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 26(8): 1864-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16741149

RESUMO

OBJECTIVE: Activation of transcription factor NF-kappaB is an important step in the development of vascular damage, because it controls inducible genes, including many inflammatory mediators. The pharmacological modulation of this process is the main objective in the design of new therapies for atherosclerosis. In this work we analyzed the effects of the natural compound parthenolide (PTN), an NF-kappaB inhibitor. METHODS AND RESULTS: In vascular smooth muscle cells (VSMCs) and monocytes stimulated with lipopolysaccharide (LPS), nontoxic doses of PTN reduced IkappaBalpha degradation, NF-kappaB activation, and MCP-1 expression, without inhibiting AP-1 and MAPK. In apoE mice, treatment with low (2 mg/kg, 20 weeks), medium (4 mg/kg, 10 weeks), and high (10 mg/kg, 10 weeks) dose of PTN reduced the size of aortic lesion, decreased macrophage, and increased VSMC content in the lesions. Treated mice showed reduced serum levels of MCP-1 and attenuated NF-kappaB activity, but not AP-1, in the lesions. Moreover, PTN affects neither apoptotic cell death nor oxidative stress in cultured cells and mice. CONCLUSIONS: NF-kappaB inhibition by PTN retards atherosclerotic lesions in apoE mice, by reducing lesion size and changing plaque composition. This natural compound could represent a novel therapeutic approach to inflammation during vascular damage.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Inflamação/etiologia , Inflamação/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Sesquiterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/etiologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Gorduras na Dieta/administração & dosagem , Humanos , Camundongos
19.
Curr Drug Targets Inflamm Allergy ; 4(3): 341-51, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16101544

RESUMO

The mesangium occupies a central anatomical position in the glomerulus, and also plays an important regulatory role in immune-mediated glomerular diseases, with an active participation in the response to local inflammation. In general, the mesangial cell responses to the pathological stimuli are associated with the main events of glomerular injury: leukocyte infiltration, cell proliferation and fibrosis. Leukocyte migration and infiltration into the glomerulus is responsible for the initiation and amplification of glomerular injury, and is mediated by adhesion molecules and chemokines, which can be locally synthesized by mesangial cells. The increase in mesangial cell number is also due to proliferation of intrinsic mesangial cell population. Regulatory mechanisms of mesangial cell replication include a complex array of factors which control cell proliferation, survival and apoptosis. Mesangial matrix accumulation leading to glomerulosclerosis, is a consequence of an imbalance between matrix production and degradation, and is controlled by growth factors and pro-inflammatory cytokines. The initial phase of immune-mediated glomerular inflammation depends on the interaction of immune complexes with specific Fc receptors in infiltrating leukocytes and resident mesangial cells, the ability of immune complexes to activate complement system, and on local inflammatory processes. Activated mesangial cells then produce many inflammatory mediators leading to amplification of the injury. This review will focus on the biological functions of mesangial cells that contribute to glomerular injury, with special attention to immune-mediated glomerulonephritis. Furthermore, new therapies based on the pathophysiology of the mesangial cell that are being developed in experimental models are also proposed.


Assuntos
Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/patologia , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/patologia , Animais , Proteínas do Sistema Complemento/fisiologia , Fibrose , Mesângio Glomerular/imunologia , Mesângio Glomerular/fisiopatologia , Glomerulonefrite/imunologia , Glomerulonefrite/fisiopatologia , Humanos , Mediadores da Inflamação/fisiologia , Receptores Fc/efeitos dos fármacos
20.
PLoS One ; 8(6): e66754, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23805273

RESUMO

Immunity contributes to arterial inflammation during atherosclerosis. Oxidized low-density lipoproteins induce an autoimmune response characterized by specific antibodies and immune complexes in atherosclerotic patients. We hypothesize that specific Fcγ receptors for IgG constant region participate in atherogenesis by regulating the inflammatory state of lesional macrophages. In vivo we examined the role of activating Fcγ receptors in atherosclerosis progression using bone marrow transplantation from mice deficient in γ-chain (the common signaling subunit of activating Fcγ receptors) to hyperlipidemic mice. Hematopoietic deficiency of Fcγ receptors significantly reduced atherosclerotic lesion size, which was associated with decreased number of macrophages and T lymphocytes, and increased T regulatory cell function. Lesions of Fcγ receptor deficient mice exhibited increased plaque stability, as evidenced by higher collagen and smooth muscle cell content and decreased apoptosis. These effects were independent of changes in serum lipids and antibody response to oxidized low-density lipoproteins. Activating Fcγ receptor deficiency reduced pro-inflammatory gene expression, nuclear factor-κB activity, and M1 macrophages at the lesion site, while increasing anti-inflammatory genes and M2 macrophages. The decreased inflammation in the lesions was mirrored by a reduced number of classical inflammatory monocytes in blood. In vitro, lack of activating Fcγ receptors attenuated foam cell formation, oxidative stress and pro-inflammatory gene expression, and increased M2-associated genes in murine macrophages. Our study demonstrates that activating Fcγ receptors influence the macrophage phenotypic balance in the artery wall of atherosclerotic mice and suggests that modulation of Fcγ receptor-mediated inflammatory responses could effectively suppress atherosclerosis.


Assuntos
Aterosclerose/fisiopatologia , Macrófagos/metabolismo , Receptores de IgG/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/mortalidade , Transplante de Medula Óssea , Colágeno/metabolismo , Modelos Animais de Doenças , Hiperlipidemias/patologia , Lipídeos/sangue , Lipoproteínas LDL/sangue , Lipoproteínas LDL/imunologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Placa Aterosclerótica/patologia , Receptores de IgG/deficiência , Linfócitos T/citologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA