Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 20070, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33208796

RESUMO

Adiposopathy is a pathological adipose tissue (AT) response to overfeeding characterized by reduced AT expandability due to impaired adipogenesis, which favors inflammation, insulin resistance (IR), and abnormal glucose regulation. However, it is unclear whether defective adipogenesis causes metabolic derangement also independently of an increased demand for fat storage. As galectin-3 has been implicated in both adipocyte differentiation and glucose homeostasis, we tested this hypothesis in galectin-3 knockout (Lgal3-/-) mice fed a standard chow. In vitro, Lgal3-/- adipocyte precursors showed impaired terminal differentiation (maturation). Two-month-old Lgal3-/- mice showed impaired AT maturation, with reduced adipocyte size and expression of adipogenic genes, but unchanged fat mass and no sign of adipocyte degeneration/death or ectopic fat accumulation. AT immaturity was associated with AT and whole-body inflammation and IR, glucose intolerance, and hyperglycemia. Five-month-old Lgal3-/- mice exhibited a more mature AT phenotype, with no difference in insulin sensitivity and expression of inflammatory cytokines versus WT animals, though abnormal glucose homeostasis persisted and was associated with reduced ß-cell function. These data show that adipogenesis capacity per se affects AT function, insulin sensitivity, and glucose homeostasis independently of increased fat intake, accumulation and redistribution, thus uncovering a direct link between defective adipogenesis, IR and susceptibility to diabetes.


Assuntos
Adipogenia , Tecido Adiposo/patologia , Galectina 3/fisiologia , Deleção de Genes , Intolerância à Glucose/patologia , Glucose/metabolismo , Resistência à Insulina , Tecido Adiposo/metabolismo , Animais , Feminino , Intolerância à Glucose/etiologia , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Metabolism ; 92: 51-60, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30458177

RESUMO

Obesity-related disease complications reduce life quality and expectancy and increase health-care costs. Some studies have suggested that obesity not always entails metabolic abnormalities and increased risk of cardiometabolic complications. Because of the lack of universally accepted criteria to identify metabolically healthy obesity (MHO), its prevalence varies widely among studies. Moreover, the prognostic value of MHO is hotly debated, mainly because it likely shifts gradually towards metabolically unhealthy obesity (MUO). In this review, we outline the differential factors contributing to the metabolic heterogeneity of obesity by discussing the behavioral, genetic, phenotypical, and biological aspects associated with each of the two metabolic phenotypes (MHO and MUO) of obesity and their clinical implications. Particular emphasis will be laid on the role of adipose tissue biology and function, including genetic determinants of body fat distribution, depot-specific fat metabolism, adipose tissue plasticity and, particularly, adipogenesis. Finally, the emerging role of gut microbiota in obesity and adipose tissue dysfunction as well as the search for novel biomarkers for the obesity-related metabolic traits and associated diseases will be briefly presented. A better understanding of the main determinants of a healthy metabolic status in obesity would allow promotion of this favorable condition by targeting the relevant pathways.


Assuntos
Obesidade Metabolicamente Benigna/metabolismo , Obesidade Metabolicamente Benigna/mortalidade , Obesidade/complicações , Obesidade/metabolismo , Tecido Adiposo/fisiopatologia , Progressão da Doença , Nível de Saúde , Humanos , Microbiota , Obesidade/fisiopatologia , Obesidade/terapia , Obesidade Metabolicamente Benigna/fisiopatologia , Obesidade Metabolicamente Benigna/terapia
3.
J Pathol ; 245(2): 197-208, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29533466

RESUMO

Diabetes is an established risk factor for pancreatic cancer (PaC), together with obesity, a Western diet, and tobacco smoking. The common mechanistic link might be the accumulation of advanced glycation end-products (AGEs), which characterizes all of the above disease conditions and unhealthy habits. Surprisingly, however, the role of AGEs in PaC has not been examined yet, despite the evidence of a tumour-promoting role of receptor for advanced glycation end-products (RAGE), the receptor for AGEs. Here, we tested the hypothesis that AGEs promote PaC through RAGE activation. To this end, we investigated the effects of the AGE Nϵ -carboxymethyllysine (CML) in human pancreatic ductal adenocarcinoma (PDA) cell lines and in a mouse model of Kras-driven PaC interbred with a bioluminescent model of proliferation. Tumour growth was monitored in vivo by bioluminescence imaging and confirmed by histology. CML promoted PDA cell growth and RAGE expression, in a concentration-dependent and time-dependent manner, and activated downstream tumourigenic signalling pathways. These effects were counteracted by RAGE antagonist peptide (RAP). Exogenous AGE administration to PaC-prone mice induced RAGE upregulation in pancreatic intraepithelial neoplasias (PanINs) and markedly accelerated progression to invasive PaC. At 11 weeks of age (6 weeks of CML treatment), PaC was observed in eight of 11 (72.7%) CML-treated versus one of 11 (9.1%) vehicle-treated [control (Ctr)] mice. RAP delayed PanIN development in Ctr mice but failed to prevent PaC promotion in CML-treated mice, probably because of competition with soluble RAGE for binding to AGEs and/or compensatory upregulation of the RAGE homologue CD166/ activated leukocyte cell adhesion molecule, which also favoured tumour spread. These findings indicate that AGEs modulate the development and progression of PaC through receptor-mediated mechanisms, and might be responsible for the additional risk conferred by diabetes and other conditions characterized by increased AGE accumulation. Finally, our data suggest that an AGE reduction strategy, instead of RAGE inhibition, might be suitable for the risk management and prevention of PaC. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proliferação de Células , Lisina/análogos & derivados , Neoplasias Pancreáticas/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma in Situ/tratamento farmacológico , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Complicações do Diabetes/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Genes ras , Humanos , Lisina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Peptídeos/farmacologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Fatores de Risco , Transdução de Sinais , Fatores de Tempo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Metabolism ; 83: 149-158, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29432728

RESUMO

OBJECTIVE: Galectin-3 is constitutively expressed in bone cells and was recently shown to modulate osteogenic transdifferentiation of vascular smooth muscle cells and atherosclerotic calcification. However, the role of galectin-3 in bone physiology is largely undefined. To address this issue, we analyzed (1) the skeletal features of 1-, 3- and 6-month-old galectin-3 null (Lgals3-/-) and wild type (WT) mice and (2) the differentiation and function of osteoblasts and osteoclasts derived from these animals. METHODS: Long bone phenotype, gene expression profile, and remodeling were investigated by micro-computed tomography, real time-PCR, static and dynamic histomorphometry, and assessment of biochemical markers of bone resorption and formation. Bone competence was also evaluated by biomechanical testing at 3 months. In vitro, the effects of galectin-3 deficiency on bone cell differentiation and function were investigated by assessing (a) gene expression of osteoblast markers, alkaline phosphatase activity, mineralization assay, and WNT/ß-catenin signaling (of which galectin-3 is a known regulator) in osteoblasts; and (b) tartrate-resistant acid phosphatase activity and bone resorption activity in osteoclasts. RESULTS: Lgals3-/- mice revealed a wide range of age-dependent alterations including lower bone formation and higher bone resorption, accelerated age-dependent trabecular bone loss (p < 0.01 vs. WT at 3 months) and reduced bone strength (p < 0.01 vs. WT at 3 months). These abnormalities were accompanied by a steady inflammatory state, as revealed by higher bone expression of the pro-inflammatory cytokines interleukin (IL)-1ß and IL-6 (p < 0.001 vs. WT at 3 months), increased content of osteal macrophages (p < 0.01 vs. WT at 3 months), and reduced expression of markers of alternative (M2) macrophage activation. Lgals3-/- osteoblasts and osteoclasts showed impaired terminal differentiation, reduced mineralization capacity (p < 0.01 vs. WT cells) and resorption activity (p < 0.01 vs. WT cells). Mechanistically, impaired differentiation and function of Lgals3-/- osteoblasts was associated with altered WNT/ß-catenin signaling (p < 0.01 vs. WT cells). CONCLUSIONS: These data provide evidence for a contribution of galectin-3 to bone cell maturation and function, bone remodeling, and biomechanical competence, thus identifying galectin-3 as a promising therapeutic target for age-related disorders of bone remodeling.


Assuntos
Remodelação Óssea/genética , Diferenciação Celular/genética , Resistência à Flexão/fisiologia , Galectina 3/fisiologia , Osteoblastos/fisiologia , Osteogênese/genética , Animais , Fenômenos Bioquímicos/genética , Densidade Óssea/genética , Células Cultivadas , Feminino , Galectina 3/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Br J Pharmacol ; 175(1): 53-66, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29053168

RESUMO

BACKGROUND AND PURPOSE: The advanced glycation end products (AGEs) participate in the pathogenesis of diabetic nephropathy (DN) by promoting renal inflammation and injury. L-carnosine acts as a quencher of the AGE precursors reactive carbonyl species (RCS), but is rapidly inactivated by carnosinase. In this study, we evaluated the effect of FL-926-16, a carnosinase-resistant and bioavailable carnosine derivative, on the onset and progression of DN in db/db mice. EXPERIMENTAL APPROACH: Adult male db/db mice and coeval db/m controls were left untreated or treated with FL-926-16 (30 mg·kg-1 body weight) from weeks 6 to 20 (prevention protocol) or from weeks 20 to 34 (regression protocol). KEY RESULTS: In the prevention protocol, FL-926-16 significantly attenuated increases in creatinine (-80%), albuminuria (-77%), proteinuria (-75%), mean glomerular area (-34%), fractional (-40%) and mean (-42%) mesangial area in db/db mice. This protective effect was associated with a reduction in glomerular matrix protein expression and cell apoptosis, circulating and tissue oxidative and carbonyl stress, and renal inflammatory markers, including the NLRP3 inflammasome. In the regression protocol, the progression of DN was completely blocked, although not reversed, by FL-926-16. In cultured mesangial cells, FL-926-16 prevented NLRP3 expression induced by RCS but not by the AGE Nε -carboxymethyllysine. CONCLUSION AND IMPLICATIONS: FL-926-16 is effective at preventing the onset of DN and halting its progression in db/db mice by quenching RCS, thereby reducing the accumulation of their protein adducts and the consequent inflammatory response. In a future perspective, this novel compound may represent a promising AGE-reducing approach for DN therapy.


Assuntos
Carnosina/uso terapêutico , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/prevenção & controle , Dipeptidases/uso terapêutico , Progressão da Doença , Animais , Disponibilidade Biológica , Carnosina/análogos & derivados , Células Cultivadas , Nefropatias Diabéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Distribuição Aleatória
6.
Oxid Med Cell Longev ; 2017: 8962458, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29270247

RESUMO

Molecular mechanisms driving transition from simple steatosis to nonalcoholic steatohepatitis (NASH), a critical step in the progression of nonalcoholic fatty liver disease (NAFLD) to cirrhosis, are poorly defined. This study aimed at investigating the role of the purinergic receptor 2X7 (PR2X7), through the NLRP3 inflammasome, in the development of NASH. To this end, mice knockout for the Pr2x7 gene (Pr2x7-/-) and coeval wild-type (WT) mice were fed a high-fat diet (HFD) or normal-fat diet for 16 weeks. NAFLD grade and stage were lower in Pr2x7-/- than WT mice, and only 1/7 Pr2x7-/- animals showed evidence of NASH, as compared with 4/7 WT mice. Molecular markers of inflammation, oxidative stress, and fibrosis were markedly increased in WT-HFD mice, whereas no or significantly reduced increments were detected in Pr2x7-/- animals, which showed also decreased modulation of genes of lipid metabolism. Deletion of Pr2x7 gene was associated with blunted or abolished activation of NLRP3 inflammasome and expression of its components, which were induced in liver sinusoidal endothelial cells challenged with appropriate stimuli. These data show that Pr2x7 gene deletion protects mice from HFD-induced NASH, possibly through blunted activation of NLRP3 inflammasome, suggesting that PR2X7 and NLRP3 may represent novel therapeutic targets.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , Receptores Purinérgicos P2X7/deficiência , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores Purinérgicos P2X7/metabolismo
7.
Oxid Med Cell Longev ; 2016: 9618092, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26770660

RESUMO

Galectin-3 is an important modulator of several biological functions. It has been implicated in numerous disease conditions, particularly in the long-term complications of diabetes because of its ability to bind the advanced glycation/lipoxidation end products that accumulate in target organs and exert their toxic effects by triggering proinflammatory and prooxidant pathways. Recent evidence suggests that galectin-3 may also participate in the development of obesity and type 2 diabetes. It has been shown that galectin-3 levels are higher in obese and diabetic individuals and parallel deterioration of glucose homeostasis. Two studies in galectin-3 knockout mice fed a high-fat diet (HFD) have shown increased adiposity and adipose tissue and systemic inflammation associated with altered glucose homeostasis, suggesting that galectin-3 negatively modulates the responsiveness of innate and adaptive immunity to overnutrition. However, these studies have also shown that impaired glucose homeostasis occurs in galectin-3 knockout animals independently of obesity. Moreover, another study reported decreased weight and fat mass in HFD-fed galectin-3 knockout mice. In vitro, galectin-3 was found to stimulate differentiation of preadipocytes into mature adipocytes. Altogether, these data indicate that galectin-3 deserves further attention in order to clarify its role as a potential player and therapeutic target in obesity and type 2 diabetes.


Assuntos
Galectina 3/metabolismo , Glucose/metabolismo , Homeostase , Obesidade/metabolismo , Tecido Adiposo/patologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Galectina 3/química , Humanos
8.
Diabetologia ; 58(4): 845-53, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25471794

RESUMO

AIMS/HYPOTHESIS: AGEs are involved in diabetic complications and might be responsible for the phenomenon of 'hyperglycaemic memory'. D-Carnosine-octylester (DCO) has been shown to attenuate AGE formation and vascular and renal injury induced by high-fat diet in Apoe-null mice. This study aimed to verify the protective effect of DCO in atherosclerosis and renal disease induced by experimental diabetes and to discover whether reduction of AGE formation by early vs late DCO treatment provides better macro and microvascular protection. METHODS: Apoe-null mice were rendered diabetic by streptozotocin and were left untreated or were treated with DCO for 20 weeks (DCO-Extended), from week 1 to 11 (DCO-Early) or from week 9 to 19 (DCO-Late). Non-diabetic Apoe-null mice served as controls. Aortic and renal lesions were evaluated by morphometry and protein and gene expression of disease markers were assessed by immunohistochemistry and real-time PCR. RESULTS: DCO-Extended treatment produced a more stable plaque phenotype by markedly attenuating diabetes-induced increases in lesion size, necrotic core area and plaque content of Nε-carboxymethyllysine, levels of apoptotic cells and markers of inflammation and oxidative stress and also reductions in collagen and smooth muscle cells. DCO treatment for 11 weeks afforded partial protection and this was significantly better in DCO-Early mice than in DCO-Late mice. Renal disease was attenuated in DCO-Extended mice and to a lesser extent in those treated for 11 weeks, with no significant difference between DCO-Early mice and DCO-Late mice. CONCLUSIONS/INTERPRETATION: These data show that DCO protects mice from diabetes-induced vascular and renal disease and that protection against atherosclerosis is more effectively achieved by early treatment than by late treatment, thus suggesting that early inhibition of AGE formation attenuates progression of macroangiopathy and favours development of more stable lesions.


Assuntos
Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Carnosina/análogos & derivados , Diabetes Mellitus Experimental/tratamento farmacológico , Angiopatias Diabéticas/prevenção & controle , Nefropatias Diabéticas/prevenção & controle , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/diagnóstico , Doenças da Aorta/genética , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/diagnóstico , Aterosclerose/genética , Biomarcadores/sangue , Carnosina/farmacologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/genética , Angiopatias Diabéticas/sangue , Angiopatias Diabéticas/diagnóstico , Angiopatias Diabéticas/genética , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/diagnóstico , Nefropatias Diabéticas/genética , Progressão da Doença , Feminino , Produtos Finais de Glicação Avançada/sangue , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Knockout , Placa Aterosclerótica , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
Atherosclerosis ; 238(2): 220-30, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25528431

RESUMO

Vascular calcification is an unfavorable event in the natural history of atherosclerosis that predicts cardiovascular morbidity and mortality. However, increasing evidence suggests that different calcification patterns are associated with different or even opposite histopathological and clinical features, reflecting the dual relationship between inflammation and calcification. In fact, initial calcium deposition in response to pro-inflammatory stimuli results in the formation of spotty or granular calcification ("microcalcification"), which induces further inflammation. This vicious cycle favors plaque rupture, unless an adaptive response prevails, with blunting of inflammation and survival of vascular smooth muscle cells (VSMCs). VSMCs promote fibrosis and also undergo osteogenic transdifferentiation, with formation of homogeneous or sheet-like calcification ("macrocalcification"), that stabilizes the plaque by serving as a barrier towards inflammation. Unfortunately, little is known about the molecular mechanisms regulating this adaptive response. The advanced glycation/lipoxidation endproducts (AGEs/ALEs) have been shown to promote vascular calcification and atherosclerosis. Recent evidence suggests that two AGE/ALE receptors, RAGE and galectin-3, modulate in divergent ways, not only inflammation, but also vascular osteogenesis, by favoring "microcalcification" and "macrocalcification", respectively. Galectin-3 seems essential for VSMC transdifferentiation into osteoblast-like cells via direct modulation of the WNT-ß-catenin signaling, thus driving formation of "macrocalcification", whereas RAGE favors deposition of "microcalcification" by promoting and perpetuating inflammation and by counteracting the osteoblastogenic effect of galectin-3. Further studies are required to understand the molecular mechanisms regulating transition from "microcalcification" to "macrocalcification", thus allowing to design therapeutic strategies which favor this adaptive process, in order to limit the adverse effects of established atherosclerotic calcification.


Assuntos
Aterosclerose/metabolismo , Cálcio/metabolismo , Mediadores da Inflamação/metabolismo , Músculo Liso Vascular/metabolismo , Calcificação Vascular/metabolismo , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Transdiferenciação Celular , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Mediadores da Inflamação/imunologia , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/patologia , Osteogênese , Prognóstico , Transdução de Sinais , Calcificação Vascular/imunologia , Calcificação Vascular/patologia
10.
Clin Chem Lab Med ; 52(10): 1413-23, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24940712

RESUMO

Galectin-3 is a versatile molecule which exerts several and sometimes opposite functions in various pathophysiological processes. Recently, galectin-3 has gained attention as a powerful predictor of heart failure and mortality, thus becoming a useful prognostic marker in clinical practice. Moreover, though not specifically investigated in diabetic cohorts, plasma levels of galectin-3 correlated with the prevalence of diabetes and related metabolic conditions, thus suggesting that pharmacological blockade of this lectin might be successful for treating heart failure especially in subjects suffering from these disorders. Indeed, galectin-3 is considered not only as a marker of heart failure, but also as a mediator of the disease, due to its pro-fibrotic action, though evidence comes mainly from studies in galectin-3 deficient mice. However, these studies have provided contrasting results, with either attenuation or acceleration of organ fibrosis and inflammation, depending on the experimental setting and particularly on the levels of advanced glycation endproducts (AGEs)/advanced lipoxidation endproducts (ALEs), of which galectin-3 is a scavenging receptor. In fact, under conditions of increased AGE/ALE levels, galectin-3 ablation was associated with tissue-specific outcomes, reflecting the AGE/ALE-receptor function of this lectin. Conversely, in experimental models of acute inflammation and fibrosis, galectin-3 deficiency resulted in attenuation of tissue injury. There is a need for prospective studies in diabetic patients specifically investigating the relation of galectin-3 levels with complications and for further animal studies in order to establish the effective role of this lectin in organ damage before considering its pharmacological blockade in the clinical setting.


Assuntos
Diabetes Mellitus/metabolismo , Galectina 3/metabolismo , Animais , Biomarcadores/metabolismo , Complicações do Diabetes/metabolismo , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/metabolismo , Humanos
11.
Cardiovasc Res ; 100(3): 472-80, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975852

RESUMO

AIMS: Vascular calcification correlates with inflammation and plaque instability in a dual manner, depending on the spotty/granular (micro) or sheet-like/lamellated (macro) pattern of calcification. Modified lipoproteins trigger both inflammation and calcification via receptors for advanced lipoxidation/glycation endproducts (ALEs/AGEs). This study compared the roles of galectin-3 and receptor for AGEs (RAGE), two ALEs/AGEs-receptors with diverging effects on inflammation and bone metabolism, in the process of vascular calcification. METHODS AND RESULTS: We evaluated galectin-3 and RAGE expression/localization in 62 human carotid plaques and its relation to calcification pattern, plaque phenotype, and markers of inflammation and vascular osteogenesis; and the effect of galectin-3 ablation and/or exposure to an ALE/AGE on vascular smooth muscle cell (VSMC) osteogenic differentiation. While RAGE co-localized with inflammatory cells in unstable regions with microcalcification, galectin-3 was expressed also by VSMCs, especially in macrocalcified areas, where it co-localized with alkaline phosphatase. Expression of galectin-3 and osteogenic markers was higher in macrocalcified plaques, whereas the opposite occurred for RAGE and inflammatory markers. Galectin-3-deficient VSMCs exhibited defective osteogenic differentiation, as shown by altered expression of osteogenic transcription factors and proteins, blunted activation of pro-osteoblastogenic Wnt/ß-catenin signalling and proliferation, enhanced apoptosis, and disorganized mineralization. These abnormalities were associated with RAGE up-regulation, but were only in part prevented by RAGE silencing, and were partially mimicked or exacerbated by treatment with an AGE/ALE. CONCLUSION: These data indicate a novel molecular mechanism by which galectin-3 and RAGE modulate in divergent ways, not only inflammation, but also vascular osteogenesis, by modulating Wnt/ß-catenin signalling, and independently of ALEs/AGEs.


Assuntos
Estenose das Carótidas/metabolismo , Galectina 3/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteogênese , Receptores Imunológicos/metabolismo , Calcificação Vascular/metabolismo , Idoso , Fosfatase Alcalina/metabolismo , Animais , Proteínas Sanguíneas , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Estenose das Carótidas/genética , Estenose das Carótidas/patologia , Diferenciação Celular , Células Cultivadas , Feminino , Galectina 3/deficiência , Galectina 3/genética , Galectinas , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Estresse Oxidativo , Placa Aterosclerótica , Interferência de RNA , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Transdução de Sinais , Transfecção , Calcificação Vascular/genética , Calcificação Vascular/patologia , Via de Sinalização Wnt
12.
J Pathol ; 231(3): 342-53, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23843215

RESUMO

Renal disease associated with type 2 diabetes and the metabolic syndrome is characterized by a distinct inflammatory phenotype. The purinergic 2X7 receptor (P2X7 R) and the nucleotide-binding and oligomerization domain-like receptor containing a pyrin domain 3 (NLRP3) inflammasome have been separately shown to play a role in two models of non-metabolic chronic kidney disease. Moreover, the NLRP3 inflammasome has been implicated in chronic low-grade sterile inflammation characterizing metabolic disorders, though the mechanism(s) involved in inflammasome activation under these conditions are still unknown. We investigated the role of P2X7 R (through activation of the NLRP3 inflammasome) in renal inflammation and injury induced by a high-fat diet, an established model of the metabolic syndrome. On a high-fat diet, mice lacking P2X7 R developed attenuated renal functional and structural alterations as well as reduced inflammation, fibrosis, and oxidative/carbonyl stress, as compared with wild-type animals, in the absence of significant differences in metabolic parameters. This was associated with blunted up-regulation of the NLRP3 inflammasome components NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), pro-caspase 1, pro-interleukin (IL)-1ß, and pro-IL-18, as well as reduced inflammasome activation, as evidenced by decreased formation of mature caspase 1, whereas mature IL-1ß and IL-18 were not detected. Up-regulated expression of NLRP3 and pro-caspase 1, post-translational processing of pro-caspase-1, and release of IL-18 in response to lipopolysaccharide + 2'(3')-O-(4-benzoylbenzoyl)ATP were attenuated by P2X7 R silencing in cultured mouse podocytes. Protein and mRNA expression of P2X7 R, NLRP3, and ASC were also increased in kidneys from subjects with type 2 diabetes and the metabolic syndrome, showing histologically documented renal disease. These data provide evidence of a major role for the purinergic system, at least in part through activation of the NLRP3 inflammasome, in the process driving 'metabolic' renal inflammation and injury and identify P2X7 R and NLRP3 as novel therapeutic targets.


Assuntos
Proteínas de Transporte/metabolismo , Dieta Hiperlipídica , Inflamassomos/metabolismo , Rim/metabolismo , Síndrome Metabólica/metabolismo , Nefrite/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Caspase 1/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Nefropatias Diabéticas/imunologia , Nefropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Fibrose , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Rim/imunologia , Rim/patologia , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nefrite/etiologia , Nefrite/imunologia , Nefrite/patologia , Estresse Oxidativo , Podócitos/imunologia , Podócitos/metabolismo , Carbonilação Proteica , Processamento de Proteína Pós-Traducional , Interferência de RNA , Receptores Purinérgicos P2X7/deficiência , Receptores Purinérgicos P2X7/genética , Transfecção
13.
Br J Pharmacol ; 166(4): 1344-56, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22229552

RESUMO

BACKGROUND AND PURPOSE: Lipoxidation-derived reactive carbonyl species (RCS) such as 4-hydroxy-2-nonenal (HNE) react with proteins to form advanced lipoxidation end products (ALEs), which have been implicated in both atherosclerosis and renal disease. L-carnosine acts as an endogenous HNE scavenger, but it is rapidly inactivated by carnosinase. This study aimed at assessing the effect of the carnosinase-resistant, D-carnosine, on HNE-induced cellular injury and of its bioavailable prodrug D-carnosine octylester on experimental atherosclerosis and renal disease. EXPERIMENTAL APPROACH: Vascular smooth muscle cells (VSMCs) were exposed to HNE or H2O2 plus D-carnosine. ApoE null mice fed a Western, pro-atherogenic diet were treated with D-carnosine octylester for 12 weeks. KEY RESULTS: In vitro, D-carnosine attenuated the effect of HNE, but not of H2O2, on VSMCs. In vivo, D-carnosine octylester-treated mice showed reduced lesion area and a more stable plaque phenotype compared with untreated animals, with reduced foam cell accumulation, inflammation and apoptosis and increased clearance of apoptotic bodies and collagen deposition, resulting in decreased necrotic core formation. Likewise, renal lesions were attenuated in D-carnosine octylester-treated versus untreated mice, with lower inflammation, apoptosis and fibrosis. This was associated with increased urinary levels of HNE-carnosine adducts and reduced protein carbonylation, circulating and tissue ALEs, expression of receptors for these products, and systemic and tissue oxidative stress. CONCLUSIONS AND IMPLICATIONS: These data indicate RCS quenching with a D-carnosine ester was highly effective in attenuating experimental atherosclerosis and renal disease by reducing carbonyl stress and inflammation and that this may represent a promising therapeutic strategy in humans.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Apolipoproteínas E/metabolismo , Aterosclerose/prevenção & controle , Carnosina/análogos & derivados , Sequestradores de Radicais Livres/uso terapêutico , Pró-Fármacos/uso terapêutico , Insuficiência Renal/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Carnosina/química , Carnosina/farmacologia , Carnosina/uso terapêutico , Linhagem Celular , Células Cultivadas , Dieta Aterogênica/efeitos adversos , Feminino , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Rim/efeitos dos fármacos , Rim/imunologia , Rim/metabolismo , Rim/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/imunologia , Células Mesangiais/metabolismo , Células Mesangiais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Estresse Oxidativo/efeitos dos fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Insuficiência Renal/imunologia , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Estereoisomerismo
14.
J Biol Chem ; 286(6): 4471-84, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21135098

RESUMO

The flagellated protozoan Giardia duodenalis is a parasite of the upper part of the small intestine of mammals, including humans, and an interesting biological model. Giardia harbors a single 14-3-3 isoform, a multifunctional protein family, that is modified at the C terminus by polyglycylation, an unusual post-translational modification consisting of the covalent addition of one or multiple glycines on the γ-carboxyl groups of specific glutamic acids. Polyglycylation affects the intracellular localization of g14-3-3, as the shortening of the polyglycine chain is correlated with a partial relocalization of 14-3-3 inside the nuclei during encystation. In this work we demonstrate that the gTTLL3, a member of the tubulin tyrosine ligase-like family, is the enzyme responsible for the 14-3-3 polyglycylation. We also identify two metallopeptidases of the M20 family, here termed gDIP1 (giardial dipeptidase 1) and gDIP2, as enzymes able to shorten the g14-3-3 polyglycine tail both in vivo and in vitro. Finally, we show that the ectopic expression of gDIP2 alters the g14-3-3 localization and strongly hampers the cyst formation. In conclusion, we have identified a polyglycylase and two deglycylases that act in concert to modulate the stage-dependent glycylation status of the multifunctional regulatory g14-3-3 protein in G. duodenalis.


Assuntos
Proteínas 14-3-3/metabolismo , Carboxipeptidases/metabolismo , Giardia/metabolismo , Metaloproteases/metabolismo , Metaloproteases/fisiologia , Peptídeo Sintases/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas 14-3-3/genética , Animais , Carboxipeptidases/genética , Giardia/genética , Glicina/genética , Glicina/metabolismo , Metaloproteases/genética , Peptídeo Sintases/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética
15.
J Hepatol ; 54(5): 975-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21145823

RESUMO

BACKGROUND & AIMS: Excess fatty acid oxidation and generation of reactive carbonyls with formation of advanced lipoxidation endproducts (ALEs) is involved in nonalcoholic steatohepatitis (NASH) by triggering inflammation, hepatocyte injury, and fibrosis. This study aimed at verifying the hypothesis that ablation of the ALE-receptor galectin-3 prevents experimental NASH by reducing receptor-mediated ALE clearance and downstream events. METHODS: Galectin-3-deficient (Lgals3(-/-)) and wild type (Lgals3(+/+)) mice received an atherogenic diet or standard chow for 8 months. Liver tissue was analyzed for morphology, inflammation, cell and matrix turnover, lipid metabolism, ALEs, and ALE-receptors. RESULTS: Steatosis was significantly less pronounced in Lgals3(-/-) than Lgals3(+/+) animals on atherogenic diet. NASH, invariably detected in Lgals3(+/+) mice, was observed, to a lower extent, only in 3/8 Lgals3(-/-) mice, showing less inflammatory, degenerative, and fibrotic phenomena than Lgals3(+/+) mice. This was associated with higher circulating ALE levels and lower tissue ALE accumulation and expression of other ALE-receptors. Up-regulation of hepatic fatty acid synthesis and oxidation, inflammatory cell infiltration, pro-inflammatory cytokines, endoplasmic reticulum stress, hepatocyte apoptosis, myofibroblast transdifferentiation, and impaired Akt phosphorylation were also significantly attenuated in Lgals3(-/-) animals. Galectin-3 silencing in liver endothelial cells resulted in reduced N(ε)-carboxymethyllysine-modified albumin uptake and ALE-receptor expression. CONCLUSIONS: Galectin-3 ablation protects from diet-induced NASH by decreasing hepatic ALE accumulation, with attenuation of inflammation, hepatocyte injury, and fibrosis. It also reduced up-regulation of lipid synthesis and oxidation causing less fat deposition, oxidative stress, and possibly insulin resistance. These data suggest that galectin-3 is a major receptor involved in ALE uptake by the liver.


Assuntos
Fígado Gorduroso , Galectina 3/genética , Galectina 3/metabolismo , Animais , Apoptose/fisiologia , Antígenos CD36/genética , Antígenos CD36/metabolismo , Dieta Aterogênica , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Fibroblastos/patologia , Inativação Gênica , Leucócitos/metabolismo , Leucócitos/patologia , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Estresse Oxidativo/fisiologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA