Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 56(3): 516-530.e9, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36738738

RESUMO

In vitro studies have associated oxidative phosphorylation (OXPHOS) with anti-inflammatory macrophages, whereas pro-inflammatory macrophages rely on glycolysis. However, the metabolic needs of macrophages in tissues (TMFs) to fulfill their homeostatic activities are incompletely understood. Here, we identified OXPHOS as the highest discriminating process among TMFs from different organs in homeostasis by analysis of RNA-seq data in both humans and mice. Impairing OXPHOS in TMFs via Tfam deletion differentially affected TMF populations. Tfam deletion resulted in reduction of alveolar macrophages (AMs) due to impaired lipid-handling capacity, leading to increased cholesterol content and cellular stress, causing cell-cycle arrest in vivo. In obesity, Tfam depletion selectively ablated pro-inflammatory lipid-handling white adipose tissue macrophages (WAT-MFs), thus preventing insulin resistance and hepatosteatosis. Hence, OXPHOS, rather than glycolysis, distinguishes TMF populations and is critical for the maintenance of TMFs with a high lipid-handling activity, including pro-inflammatory WAT-MFs. This could provide a selective therapeutic targeting tool.


Assuntos
Inflamação , Fosforilação Oxidativa , Humanos , Camundongos , Animais , Inflamação/metabolismo , Macrófagos/metabolismo , Homeostase , Lipídeos , Tecido Adiposo/metabolismo
2.
Am J Kidney Dis ; 80(2): 251-263, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34999158

RESUMO

Kidney fibrosis is a hallmark of chronic kidney disease (CKD) and a potential therapeutic target. However, there are conceptual and practical challenges to directly targeting kidney fibrosis. Whether fibrosis is mainly a cause or a consequence of CKD progression has been disputed. It is unclear whether specifically targeting fibrosis is feasible in clinical practice because most drugs that decrease fibrosis in preclinical models target additional and often multiple pathogenic pathways (eg, renin-angiotensin-aldosterone system blockade). Moreover, tools to assess whole-kidney fibrosis in routine clinical practice are lacking. Pirfenidone, a drug used for idiopathic pulmonary fibrosis, is undergoing a phase 2 trial for kidney fibrosis. Other drugs in use or being tested for idiopathic pulmonary fibrosis (eg, nintedanib, PRM-151, epigallocatechin gallate) are also potential candidates to treat kidney fibrosis. Novel therapeutic approaches may include antagomirs (eg, lademirsen) or drugs targeting interleukin 11 or NKD2 (WNT signaling pathway inhibitor). Reversing the dysfunctional tubular cell metabolism that leads to kidney fibrosis offers additional therapeutic opportunities. However, any future drug targeting fibrosis of the kidneys should demonstrate added benefit to a standard of care that combines renin-angiotensin system with mineralocorticoid receptor (eg, finerenone) blockade or with sodium/glucose cotransporter 2 inhibitors.


Assuntos
Fibrose Pulmonar Idiopática , Insuficiência Renal Crônica , Proteínas Adaptadoras de Transdução de Sinal , Antifibróticos , Proteínas de Ligação ao Cálcio , Fibrose , Humanos , Fibrose Pulmonar Idiopática/complicações , Insuficiência Renal Crônica/etiologia , Sistema Renina-Angiotensina
3.
FASEB J ; 35(1): e21213, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33368614

RESUMO

Preclinical studies have demonstrated that activation of the NOTCH pathway plays a key role in the pathogenesis of kidney damage. There is currently no information on the role of the Delta-like homologue 1 (DLK1), a NOTCH inhibitor, in the regulation of renal damage. Here, we investigated the contribution of DLK1 to experimental renal damage and the underlying molecular mechanisms. Using a Dlk1-null mouse model in the experimental renal damage of unilateral ureteral obstruction, we found activation of NOTCH, as shown by increased nuclear translocation of the NOTCH1 intracellular domain, and upregulation of Dlk2/hey-1 expression compared to wild-type (WT) littermates. NOTCH1 over-activation in Dlk1-null injured kidneys was associated with a higher inflammatory response, characterized by infiltration of inflammatory cells, mainly CD4/IL17A + lymphocytes, and activation of the Th17 immune response. Furthermore, pharmacological NOTCH blockade inhibited the transcription factors controlling Th17 differentiation and gene expression of the Th17 effector cytokine IL-17A and other related-inflammatory factors, linked to a diminution of inflammation in the injured kidneys. We propose that the non-canonical NOTCH ligand DLK1 acts as a NOTCH antagonist in renal injury regulating the Th17-mediated inflammatory response.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Deleção de Genes , Imunidade Celular , Nefropatias/imunologia , Rim/imunologia , Células Th17/imunologia , Animais , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Camundongos , Células Th17/patologia , Obstrução Ureteral/genética , Obstrução Ureteral/imunologia , Obstrução Ureteral/patologia
4.
Liver Int ; 42(3): 651-662, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35050550

RESUMO

BACKGROUND AND AIMS: PTTG1 is almost undetectable in adult livers but is highly expressed in hepatocarcinoma. While little is known about its involvement in liver fibrosis, PTTG1 expression is associated with DLK1. We assessed the role of the PTTG1/DLK1 pathway in fibrosis progression and the potential therapeutic effect of PTTG1 silencing in fibrosis. METHODS: Pttg1 and Dlk1 were studied in liver and isolated cell populations of control and fibrotic rats and in human liver biopsies. The fibrotic molecular signature was analysed in Pttg1-/- and Pttg1+/+ fibrotic mice. Finally, Pttg1 silencing was evaluated in rats as a novel antifibrotic therapy. RESULTS: Pttg1 and Dlk1 mRNA selectively increased in fibrotic rats paralleling fibrosis progression. Serum DLK1 concentrations correlated with hepatic collagen content and systemic and portal haemodynamics. Human cirrhotic livers showed greater PTTG1 and DLK1 transcript abundance than non-cirrhotic, and reduced collagen was observed in Pttg1 Pttg1-/- mice. The liver fibrotic molecular signature revealed lower expression of genes related to extracellular matrix remodelling including Mmp8 and 9 and Timp4 and greater eotaxin and Mmp13 than fibrotic Pttg1+/+ mice. Finally, interfering Pttg1 resulted in reduced liver fibrotic area, lower α-Sma and decreased portal pressure than fibrotic animals. Furthermore, Pttg1 silencing decreased the transcription of Dlk1, collagens I and III, Pdgfrß, Tgfrß, Timp1, Timp2 and Mmp2. CONCLUSIONS: Pttg1/Dlk1 are selectively overexpressed in the cirrhotic liver and participate in ECM turnover regulation. Pttg1 disruption decreases Dlk1 transcription and attenuates collagen deposition. PTTG1/DLK1 signalling is a novel pathway for targeting the progression of liver fibrosis.


Assuntos
Proteínas de Ligação ao Cálcio , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana , Neoplasias Hipofisárias , Securina , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Fibrose , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fígado/patologia , Cirrose Hepática/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Oncogenes , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Ratos , Securina/genética , Securina/metabolismo
5.
Kidney Int ; 100(6): 1165-1167, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34802556

RESUMO

The metabolic impairment of kidney tubular cells is a key mechanism underlying the pathophysiology of renal fibrosis. In particular, a drastic reduction in fatty acid oxidation is essentially responsible for the global energy failure occurring in the tubulointerstitial compartment. Piret et al. propose a novel transcriptional regulatory mechanism involving the decrease in the expression of Krüppel-like factor 15 in proximal tubular cells after kidney injury, which results in a major derangement of fatty acid oxidation.


Assuntos
Nefropatias , Túbulos Renais Proximais , Fibrose , Humanos , Rim/patologia , Nefropatias/patologia , Túbulos Renais Proximais/metabolismo , Metabolismo dos Lipídeos
6.
FASEB J ; 34(1): 410-431, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914684

RESUMO

MicroRNAs (miRNAs) regulate gene expression posttranscriptionally and control biological processes (BPs), including fibrogenesis. Kidney fibrosis remains a clinical challenge and miRNAs may represent a valid therapeutic avenue. We show that miR-9-5p protected from renal fibrosis in the mouse model of unilateral ureteral obstruction (UUO). This was reflected in reduced expression of pro-fibrotic markers, decreased number of infiltrating monocytes/macrophages, and diminished tubular epithelial cell injury and transforming growth factor-beta 1 (TGF-ß1)-dependent de-differentiation in human kidney proximal tubular (HKC-8) cells. RNA-sequencing (RNA-Seq) studies in the UUO model revealed that treatment with miR-9-5p prevented the downregulation of genes related to key metabolic pathways, including mitochondrial function, oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), and glycolysis. Studies in human tubular epithelial cells demonstrated that miR-9-5p impeded TGF-ß1-induced bioenergetics derangement. The expression of the FAO-related axis peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)-peroxisome proliferator-activated receptor alpha (PPARα) was reduced by UUO, although preserved by the administration of miR-9-5p. We found that in mice null for the mitochondrial master regulator PGC-1α, miR-9-5p was unable to promote a protective effect in the UUO model. We propose that miR-9-5p elicits a protective response to chronic kidney injury and renal fibrosis by inducing reprogramming of the metabolic derangement and mitochondrial dysfunction affecting tubular epithelial cells.


Assuntos
Reprogramação Celular , Fibrose/prevenção & controle , Regulação da Expressão Gênica , Nefropatias/prevenção & controle , MicroRNAs/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/fisiologia , Obstrução Ureteral/prevenção & controle , Animais , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcriptoma , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
7.
Anal Bioanal Chem ; 412(24): 6391-6405, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32285184

RESUMO

Despite the recent advances in the standardization of untargeted metabolomics workflows, there is still a lack of attention to specific data treatment strategies that require deep knowledge of the biological problem and need to be applied after a well-thought out process to understand the effect of the practice. One of those strategies is data normalization. Data-driven assumptions are critical especially addressing unwanted variation present in the biological model as it can be the case in heterogeneous tissues, cells with different sizes or biofluids with different concentrations. Chronic kidney disease (CKD) is a widespread disorder affecting kidney structure and function. Animal models are being developed to be able to get valuable insights into the etiopathogenesis of the condition and effect of the treatments. Moreover, diagnosis and disease staging still require defining appropriate biomarkers. Untargeted metabolomics has the potential to deal with those challenges. Renal fibrosis is one of the consequences of kidney injury which greatly affects the concentration of metabolites in the same quantity of sample. To overcome this challenge, several data normalization strategies have been applied, following a multilevel normalization method with the overall aim of focussing on the relevant biological information and reducing the influence of disturbing factors. A comprehensive evaluation of the performance of the normalization strategies, both on methods assessing the intragroup variation and on the impact on differential analysis, is provided. Finally, we present evidence of the importance of biological-model-driven guided normalization methods and discuss multiple criteria that need to be taken into consideration to obtain robust and reliable data. Special concern is transmitted on the misleading conclusions that might be the consequence of inappropriate data pre-treatment solutions applied for untargeted methods. Graphical abstract.


Assuntos
Rim/metabolismo , Metabolômica/métodos , Insuficiência Renal Crônica/metabolismo , Animais , Análise Discriminante , Modelos Animais de Doenças , Humanos , Análise dos Mínimos Quadrados , Masculino , Metaboloma , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Eur Heart J ; 40(12): 967-978, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29668883

RESUMO

AIMS: Myocardial fibrosis is associated with profound changes in ventricular architecture and geometry, resulting in diminished cardiac function. There is currently no information on the role of the delta-like homologue 1 (Dlk1) in the regulation of the fibrotic response. Here, we investigated whether Dlk1 is involved in cardiac fibroblast-to-myofibroblast differentiation and regulates myocardial fibrosis and explored the molecular mechanism underpinning its effects in this process. METHODS AND RESULTS: Using Dlk1-knockout mice and adenoviral gene delivery, we demonstrate that overexpression of Dlk1 in cardio-fibroblasts resulted in inhibition of fibroblast proliferation and differentiation into myofibroblasts. This process is mediated by TGF-ß1 signalling, since isolated fibroblasts lacking Dlk1 exhibited a higher activation of the TGF-ß1/Smad-3 pathway at baseline, leading to an earlier acquisition of a myofibroblast phenotype. Likewise, Dlk1-null mice displayed increased TGF-ß1/Smad3 cardiac activity, resulting in infiltration/accumulation of myofibroblasts, induction and deposition of extra-domain A-fibronectin isoform and collagen, and activation of pro-fibrotic markers. Furthermore, these profibrotic events were associated with disrupted myofibril integrity, myocyte hypertrophy, and cardiac dysfunction. Interestingly, Dlk1 expression was down-regulated in ischaemic human and porcine heart tissues. Mechanistically, miR-370 mediated Dlk1's regulation of cardiac fibroblast-myofibroblast differentiation by directly targeting TGFß-R2/Smad-3 signalling, while the Dlk1 canonical target, Notch pathway, does not seem to play a role in this process. CONCLUSION: These findings are the first to demonstrate an inhibitory role of Dlk1 of cardiac fibroblast-to-myofibroblast differentiation by interfering with TGFß/Smad-3 signalling in the myocardium. Given the deleterious effects of continuous activation of this pathway, we propose Dlk1 as a new potential candidate for therapy in cases where aberrant TGFß signalling leads to chronic fibrosis.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Fibroblastos/metabolismo , Fibrose/genética , Miocárdio/patologia , Miofibroblastos/metabolismo , Animais , Diferenciação Celular , Regulação para Baixo , Humanos , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Proteína Smad3/genética , Suínos , Fator de Crescimento Transformador beta1/genética
9.
EMBO Rep ; 16(10): 1358-77, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26315535

RESUMO

Uncontrolled extracellular matrix (ECM) production by fibroblasts in response to injury contributes to fibrotic diseases, including idiopathic pulmonary fibrosis (IPF). Reactive oxygen species (ROS) generation is involved in the pathogenesis of IPF. Transforming growth factor-ß1 (TGF-ß1) stimulates the production of NADPH oxidase 4 (NOX4)-dependent ROS, promoting lung fibrosis (LF). Dysregulation of microRNAs (miRNAs) has been shown to contribute to LF. To identify miRNAs involved in redox regulation relevant for IPF, we performed arrays in human lung fibroblasts exposed to ROS. miR-9-5p was selected as the best candidate and we demonstrate its inhibitory effect on TGF-ß receptor type II (TGFBR2) and NOX4 expression. Increased expression of miR-9-5p abrogates TGF-ß1-dependent myofibroblast phenotypic transformation. In the mouse model of bleomycin-induced LF, miR-9-5p dramatically reduces fibrogenesis and inhibition of miR-9-5p and prevents its anti-fibrotic effect both in vitro and in vivo. In lung specimens from patients with IPF, high levels of miR-9-5p are found. In omentum-derived mesothelial cells (MCs) from patients subjected to peritoneal dialysis (PD), miR-9-5p also inhibits mesothelial to myofibroblast transformation. We propose that TGF-ß1 induces miR-9-5p expression as a self-limiting homeostatic response.


Assuntos
Fibroblastos/citologia , Fibrose/genética , Fibrose Pulmonar Idiopática/genética , MicroRNAs/genética , NADPH Oxidases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fibrose Pulmonar/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Bleomicina , Diferenciação Celular , Fibroblastos/efeitos dos fármacos , Humanos , Camundongos , MicroRNAs/isolamento & purificação , Miofibroblastos/fisiologia , NADPH Oxidase 4 , NADPH Oxidases/genética , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta1/metabolismo
10.
J Am Soc Nephrol ; 26(1): 173-82, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25012164

RESUMO

In patients undergoing peritoneal dialysis (PD), chronic exposure to nonphysiologic PD fluids elicits low-grade peritoneal inflammation, leading to fibrosis and angiogenesis. Phenotype conversion of mesothelial cells into myofibroblasts, the so-called mesothelial-to-mesenchymal transition (MMT), significantly contributes to the peritoneal dysfunction related to PD. A number of factors have been described to induce MMT in vitro and in vivo, of which TGF-ß1 is probably the most important. The vasoconstrictor peptide endothelin-1 (ET-1) is a transcriptional target of TGF-ß1 and mediates excessive scarring and fibrosis in several tissues. This work studied the contribution of ET-1 to the development of peritoneal damage and failure in a mouse model of PD. ET-1 and its receptors were expressed in the peritoneal membrane and upregulated on PD fluid exposure. Administration of an ET receptor antagonist, either bosentan or macitentan, markedly attenuated PD-induced MMT, fibrosis, angiogenesis, and peritoneal functional decline. Adenovirus-mediated overexpression of ET-1 induced MMT in human mesothelial cells in vitro and promoted the early cellular events associated with peritoneal dysfunction in vivo. Notably, TGF-ß1-blocking peptides prevented these actions of ET-1. Furthermore, a positive reciprocal relationship was observed between ET-1 expression and TGF-ß1 expression in human mesothelial cells. These results strongly support a role for an ET-1/TGF-ß1 axis as an inducer of MMT and subsequent peritoneal damage and fibrosis, and they highlight ET-1 as a potential therapeutic target in the treatment of PD-associated dysfunction.


Assuntos
Endotelina-1/fisiologia , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/patologia , Adenoviridae/genética , Animais , Células Cultivadas , Endotelina-1/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Fibrose/metabolismo , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Peritônio/metabolismo , Peritônio/patologia , Fenótipo , Fator de Crescimento Transformador beta1/metabolismo
11.
Biochim Biophys Acta ; 1843(11): 2403-13, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25038307

RESUMO

Endothelial cells in the vascular system are constantly subjected to the frictional force of shear stress due to the pulsatile nature of blood flow. Although several proteins form part of the shear stress mechano-sensing pathway, the identification of mechano-transducing pathways is largely unknown. Given the increasing evidence for a signaling function of mitochondria in endothelial cells, the aim of this study was to investigate their role as mechano-sensor organelles during laminar shear stress (LSS). We demonstrated that LSS activates intracellular signaling pathways that modulate not only mitochondrial dynamics but also mitochondrial function. At early time points of LSS, the fission-related protein Drp1 was recruited from the cytosol to mitochondria and activated mitochondrial fission. LSS-dependent increase in intracellular Ca(2+) concentration was indispensable for mitochondrial fission. As alterations in mitochondrial dynamics have been related to changes in bioenergetics profiles, we studied mitochondrial function after LSS. We found that LSS decreased respiration rate, increased mitochondrial membrane potential and promoted the mitochondrial generation of ROS with the subsequent oxidation and activation of the antioxidant enzyme PRX3. Our data support a novel and active role for mitochondria in endothelial cells as active players, able to transduce the mechanical force of shear stress in the vascular endothelium into a biological response.

12.
Biochim Biophys Acta ; 1833(12): 2586-2595, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23806663

RESUMO

ADP plays critical signaling roles in the vascular endothelium. ADP receptors are targeted by several cardiovascular drugs, yet the intracellular pathways modulated by ADP are incompletely understood. These studies have identified important roles for the phosphatase PTEN in ADP-dependent modulation of the endothelial isoform of nitric oxide synthase (eNOS) as well as of lipid and protein kinase pathways in endothelial cells. We find that ADP-promoted eNOS activation as well as phosphorylation of p38 MAPK are enhanced by siRNA-mediated PTEN knockdown. However, the increase in ADP-dependent eNOS activation promoted by PTEN knockdown is abrogated by siRNA-mediated knockdown of p38 MAPK. These findings indicate that PTEN tonically suppresses both p38 phosphorylation as well as ADP-stimulated eNOS activity. A key enzymatic activity of PTEN is its role as a lipid phosphatase, catalyzing the dephosphorylation of phosphoinositol-3,4,5-trisphosphate (PIP3) to phosphoinositol-4,5-bisphosphate (PIP2). We performed biochemical analyses of cellular phospholipids in endothelial cells to show that siRNA-mediated PTEN knockdown leads to a marked increase in PIP3. Because these complex lipids activate the small GTPase Rac1, we explored the role of PTEN in ADP-modulated Rac1 activation. We used a FRET biosensor for Rac1 to show that ADP-dependent Rac1 activation is blocked by siRNA-mediated PTEN knockdown. We then exploited a FRET biosensor for PIP3 to show that the striking ADP-dependent increase in intracellular PIP3 is entirely blocked by PTEN knockdown. These studies identify a key role for PTEN in the modulation of lipid mediators involved in ADP receptor-regulated endothelial signaling pathways involving eNOS activation in vascular endothelial cells.


Assuntos
Difosfato de Adenosina/farmacologia , Células Endoteliais/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Actinas/metabolismo , Animais , Aorta/citologia , Bovinos , Movimento Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Modelos Biológicos , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 33(3): 621-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23329136

RESUMO

OBJECTIVE: Dyslipidemia contributes to endothelial dysfunction in type 2 diabetes mellitus. Fenofibrate (FF), a ligand of the peroxisome proliferator-activated receptor-α (PPARα), has beneficial effects on microvascular complications. FF may act on the endothelium by regulating vasoactive factors, including endothelin-1 (ET-1). In vitro, FF decreases ET-1 expression in human microvascular endothelial cells. We investigated the molecular mechanisms involved in the effect of FF treatment on plasma levels of ET-1 in type 2 diabetes mellitus patients. METHODS AND RESULTS: FF impaired the capacity of transforming growth factor-ß to induce ET-1 gene expression. PPARα activation by FF increased expression of the transcriptional repressor Krüppel-like factor 11 and its binding to the ET-1 gene promoter. Knockdown of Krüppel-like factor 11 expression potentiated basal and transforming growth factor-ß-stimulated ET-1 expression, suggesting that Krüppel-like factor 11 downregulates ET-1 expression. FF, in a PPARα-independent manner, and insulin enhanced glycogen synthase kinase-3ß phosphorylation thus reducing glycogen synthase kinase-3 activity that contributes to the FF-mediated reduction of ET-1 gene expression. In type 2 diabetes mellitus, improvement of flow-mediated dilatation of the brachial artery by FF was associated with a decrease in plasma ET-1. CONCLUSIONS: FF decreases ET-1 expression by a PPARα-dependent mechanism, via transcriptional induction of the Krüppel-like factor 11 repressor and by PPARα-independent actions via inhibition of glycogen synthase kinase-3 activity.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Endotelina-1/metabolismo , Fenofibrato/uso terapêutico , Hipolipemiantes/uso terapêutico , PPAR alfa/agonistas , Proteínas Reguladoras de Apoptose , Sítios de Ligação , Artéria Braquial/efeitos dos fármacos , Artéria Braquial/fisiopatologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Regulação para Baixo , Dislipidemias/sangue , Dislipidemias/metabolismo , Dislipidemias/fisiopatologia , Células Endoteliais/metabolismo , Endotelina-1/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , PPAR alfa/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Vasodilatação/efeitos dos fármacos
14.
Mol Cell Proteomics ; 11(9): 800-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22647871

RESUMO

Nitroxidative stress in cells occurs mainly through the action of reactive nitrogen and oxygen species (RNOS) on protein thiol groups. Reactive nitrogen and oxygen species-mediated protein modifications are associated with pathophysiological states, but can also convey physiological signals. Identification of Cys residues that are modified by oxidative stimuli still poses technical challenges and these changes have never been statistically analyzed from a proteome-wide perspective. Here we show that GELSILOX, a method that combines a robust proteomics protocol with a new computational approach that analyzes variance at the peptide level, allows a simultaneous analysis of dynamic alterations in the redox state of Cys sites and of protein abundance. GELSILOX permits the characterization of the major endothelial redox targets of hydrogen peroxide in endothelial cells and reveals that hypoxia induces a significant increase in the status of oxidized thiols. GELSILOX also detected thiols that are redox-modified by ischemia-reperfusion in heart mitochondria and demonstrated that these alterations are abolished in ischemia-preconditioned animals.


Assuntos
Estresse Oxidativo , Proteínas/metabolismo , Proteoma/análise , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Marcação por Isótopo , Masculino , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Oxirredução , Proteômica , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Nitrogênio/análise , Espécies Reativas de Oxigênio/análise , Traumatismo por Reperfusão/metabolismo
15.
Free Radic Biol Med ; 222: 85-105, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38838921

RESUMO

Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.

16.
J Clin Med ; 13(9)2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38730996

RESUMO

Background: Rapidly progressive osteoarthritis of the hip (RPOH) is a rare syndrome that involves the femoral head and acetabulum. Methods: We analyzed the incidence of RPOH in 2022. The inclusion criteria included a clinical history of pain for 1-6 months and a decrease in joint space of > 2 mm within one year or a decrease in joint space by 50% in that time accompanied by femoral and/or acetabular bone destruction. Exclusion: There are no other destructive arthropathies and no evolutionary radiological image sequence. Results: There were 15 patients, 16 hips, an incidence around 3.17%, a 1:1 laterality ratio, and 1 bilateral affected. The mean average age is 77.35 years. The male/female ratio is 1:2. The average BMI is 31.2. The time of the onset of the symptoms to the patient's diagnosis is 5 months. The functionally modified Harris scale (MHS) had an average score of 30 points. They had surgery hip arthroplasty with a cementless cup in all cases, a revision cup in one of them, and a double mobility cup in the other, with the stem cemented three times. There were no post-surgical complications. Functionally was achieved at 3 months. The average MHS is 70 at 12 months. The average MHS is 85. Conclusions: RPOH is an idiopathic entity characterized by great clinical involvement and rapid radiological evolution. It is most common in women around 77 years of age. The bone quality requires surgical alternatives to implants, and it has good functional recovery post-surgery.

17.
Biol Chem ; 394(10): 1263-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24002664

RESUMO

Glutathione is considered the main regulator of redox balance in the cellular milieu due to its capacity for detoxifying deleterious molecules. The oxidative stress induced as a result of a variety of stimuli promotes protein oxidation, usually at cysteine residues, leading to changes in their activity. Mild oxidative stress, which may take place in physiological conditions, induces the reversible oxidation of cysteines to sulfenic acid form, while pathological conditions are associated with higher rates of reactive oxygen species production, inducing the irreversible oxidation of cysteines. Among these, neurodegenerative disorders, cardiovascular diseases and diabetes have been proposed to be pathogenetically linked to this state. In diabetes-associated vascular complications, lower levels of glutathione and increased oxidative stress have been reported. S-glutathionylation has been proposed as a posttranslational modification able to protect proteins from over-oxidizing environments. S-glutathionylation has been identified in proteins involved in diabetic models both in vitro and in vivo. In all of them, S-glutathionylation represents a mechanism that regulates the response to diabetic conditions, and has been described to occur in erythrocytes and neutrophils from diabetic patients. However, additional studies are necessary to discern whether this modification represents a biomarker for the early onset of diabetic vascular complications.


Assuntos
Biomarcadores/química , Diabetes Mellitus/fisiopatologia , Glutationa/metabolismo , Animais , Diabetes Mellitus/diagnóstico , Modelos Animais de Doenças , Glutationa/química , Humanos , Espécies Reativas de Oxigênio/metabolismo
18.
Arthritis Rheum ; 64(5): 1653-64, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22492165

RESUMO

OBJECTIVE: Enhanced adhesive signaling, including activation of focal adhesion kinase (FAK), is a hallmark of fibroblasts from lung fibrosis patients, and FAK has therefore been hypothesized to be a key mediator of this disease. This study was undertaken to characterize the contribution of FAK to the development of pulmonary fibrosis both in vivo and in vitro. METHODS: FAK expression and activity were analyzed in lung tissue samples from lung fibrosis patients by immunohistochemistry. Mice orally treated with the FAK inhibitor PF-562,271, or with small interfering RNA (siRNA)-mediated silencing of FAK were exposed to intratracheally instilled bleomycin to induce lung fibrosis, and lungs were harvested for histologic and biochemical analysis. Using endothelin 1 (ET-1) as a stimulus, cell adhesion and contraction, as well as profibrotic gene expression, were studied in fibroblasts isolated from wild-type and FAK-deficient mouse embryos. ET-1-mediated FAK activation and gene expression were studied in primary mouse lung fibroblasts, as well as in wild-type and ß1 integrin-deficient mouse fibroblasts. RESULTS: FAK expression and activity were up-regulated in fibroblast foci and remodeled vessels from lung fibrosis patients. Pharmacologic or siRNA-mediated targeting of FAK resulted in marked abrogation of bleomycin-induced lung fibrosis in mice. Loss of FAK impaired the acquisition of a profibrotic phenotype in response to ET-1. Profibrotic gene expression leading to myofibroblast differentiation required cell adhesion, and was driven by JNK activation through ß1 integrin/FAK signaling. CONCLUSION: These results implicate FAK as a central mediator of fibrogenesis, and highlight this kinase as a potential therapeutic target in fibrotic diseases.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Indóis/farmacologia , Pulmão/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Fibrose Pulmonar/prevenção & controle , Sulfonamidas/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Endotelina-1/farmacologia , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Inativação Gênica , Humanos , Pulmão/enzimologia , Pulmão/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , RNA Interferente Pequeno/genética , Regulação para Cima/efeitos dos fármacos
19.
Front Mol Biosci ; 10: 1161036, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37377862

RESUMO

Background: Chronic kidney disease (CKD) is characterized by the progressive and irreversible deterioration of kidney function and structure with the appearance of renal fibrosis. A significant decrease in mitochondrial metabolism, specifically a reduction in fatty acid oxidation (FAO) in tubular cells, is observed in tubulointerstitial fibrosis, whereas FAO enhancement provides protection. Untargeted metabolomics offers the potential to provide a comprehensive analysis of the renal metabolome in the context of kidney injury. Methodology: Renal tissue from a carnitine palmitoyl transferase 1a (Cpt1a) overexpressing mouse model, which displays enhanced FAO in the renal tubule, subjected to folic acid nephropathy (FAN) was studied through a multiplatform untargeted metabolomics approach based on LC-MS, CE-MS and GC-MS analysis to achieve the highest coverage of the metabolome and lipidome affected by fibrosis. The expression of genes related to the biochemical routes showing significant changes was also evaluated. Results: By combining different tools for signal processing, statistical analysis and feature annotation, we were able to identify variations in 194 metabolites and lipids involved in many metabolic routes: TCA cycle, polyamines, one-carbon metabolism, amino acid metabolism, purine metabolism, FAO, glycerolipids and glycerophospholipids synthesis and degradation, glycosphingolipids interconversion, and sterol metabolism. We found several metabolites strongly altered by FAN, with no reversion induced by Cpt1a overexpression (v.g. citric acid), whereas other metabolites were influenced by CPT1A-induced FAO (v.g. glycine-betaine). Conclusion: It was implemented a successful multiplatform metabolomics approach for renal tissue analysis. Profound metabolic changes accompany CKD-associated fibrosis, some associated with tubular FAO failure. These results highlight the importance of addressing the crosstalk between metabolism and fibrosis when undertaking studies attempting to elucidate the mechanism of CKD progression.

20.
Life Sci Alliance ; 6(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37487638

RESUMO

Tubulointerstitial fibrosis is the common pathological substrate for many etiologies leading to chronic kidney disease. Although perturbations in the circadian rhythm have been associated with renal disease, the role of the molecular clock in the pathogenesis of fibrosis remains incompletely understood. We investigated the relationship between the molecular clock and renal damage in experimental models of injury and fibrosis (unilateral ureteral obstruction, folic acid, and adenine nephrotoxicity), using genetically modified mice with selective deficiencies of the clock components Bmal1, Clock, and Cry We found that the molecular clock pathway was enriched in damaged tubular epithelial cells with marked metabolic alterations. In human tubular epithelial cells, TGFß significantly altered the expression of clock components. Although Clock played a role in the macrophage-mediated inflammatory response, the combined absence of Cry1 and Cry2 was critical for the recruitment of neutrophils, correlating with a worsening of fibrosis and with a major shift in the expression of metabolism-related genes. These results support that renal damage disrupts the kidney peripheral molecular clock, which in turn promotes metabolic derangement linked to inflammatory and fibrotic responses.


Assuntos
Adenina , Rim , Humanos , Animais , Camundongos , Ritmo Circadiano , Células Epiteliais , Macrófagos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA