Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 477
Filtrar
1.
Genes Dev ; 35(21-22): 1395-1397, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725126

RESUMO

Adipose tissue is a complex organ consisting of a mixture of mature adipocytes and stromal vascular cells. It displays a remarkable ability to adapt to environmental and dietary cues by changing its morphology and metabolic capacity. This plasticity is demonstrated by the emergence of interspersed thermogenic beige adipocytes within white depots in response to catecholamines secretion. Coordinated cellular interaction between different cell types within the tissue and a fine-tuned transcriptional program synergistically take place to promote beige remodeling. However, both cell-cell interactions and molecular mechanisms governing beige adipocyte appearance and maintenance are poorly understood. In this and the previous issue of Genes & Development, Shao and colleagues (pp. 1461-1474) and Shan and colleagues (pp. 1333-1338) advance our understanding of these issues and, in doing so, highlight potential therapeutic strategies to combat obesity-associated diseases.


Assuntos
Adipócitos Bege , Termogênese , Adipócitos Bege/metabolismo , Tecido Adiposo , Tecido Adiposo Branco/metabolismo , Termogênese/genética
2.
Genes Dev ; 35(21-22): 1461-1474, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34620682

RESUMO

Energy-storing white adipocytes maintain their identity by suppressing the energy-burning thermogenic gene program of brown and beige adipocytes. Here, we reveal that the protein-protein interaction between the transcriptional coregulator ZFP423 and brown fat determination factor EBF2 is essential for restraining the thermogenic phenotype of white adipose tissue (WAT). Disruption of the ZFP423-EBF2 protein interaction through CRISPR-Cas9 gene editing triggers widespread "browning" of WAT in adult mice. Mechanistically, ZFP423 recruits the NuRD corepressor complex to EBF2-bound thermogenic gene enhancers. Loss of adipocyte Zfp423 induces an EBF2 NuRD-to-BAF coregulator switch and a shift in PPARγ occupancy to thermogenic genes. This shift in PPARγ occupancy increases the antidiabetic efficacy of the PPARγ agonist rosiglitazone in obesity while diminishing the unwanted weight-gaining effect of the drug. These data indicate that ZFP423 controls EBF2 coactivator recruitment and PPARγ occupancy to determine the thermogenic plasticity of adipocytes and highlight the potential of therapeutically targeting transcriptional brakes to induce beige adipocyte biogenesis in obesity.


Assuntos
PPAR gama , Termogênese , Adipócitos Marrons/metabolismo , Adipócitos Brancos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA , Camundongos , PPAR gama/genética , Termogênese/genética , Fatores de Transcrição
3.
Genes Dev ; 32(15-16): 1035-1044, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30006480

RESUMO

The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is known to regulate lipid metabolism in many tissues, including macrophages. Here we report that peritoneal macrophage respiration is enhanced by rosiglitazone, an activating PPARγ ligand, in a PPARγ-dependent manner. Moreover, PPARγ is required for macrophage respiration even in the absence of exogenous ligand. Unexpectedly, the absence of PPARγ dramatically affects the oxidation of glutamine. Both glutamine and PPARγ have been implicated in alternative activation (AA) of macrophages, and PPARγ was required for interleukin 4 (IL4)-dependent gene expression and stimulation of macrophage respiration. Indeed, unstimulated macrophages lacking PPARγ contained elevated levels of the inflammation-associated metabolite itaconate and express a proinflammatory transcriptome that, remarkably, phenocopied that of macrophages depleted of glutamine. Thus, PPARγ functions as a checkpoint, guarding against inflammation, and is permissive for AA by facilitating glutamine metabolism. However, PPARγ expression is itself markedly increased by IL4. This suggests that PPARγ functions at the center of a feed-forward loop that is central to AA of macrophages.


Assuntos
Glutamina/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , PPAR gama/fisiologia , Animais , Respiração Celular , Células Cultivadas , Ácidos Graxos/metabolismo , Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Interleucina-4/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama/genética , Rosiglitazona , Tiazolidinedionas/farmacologia
4.
Clin Endocrinol (Oxf) ; 100(2): 149-163, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37933831

RESUMO

OBJECTIVE: Characteristic features of polycystic ovary syndrome (PCOS) include insulin resistance and an increased risk for type 2 diabetes. To promote improved insulin sensitivity, insulin sensitisers have been used in PCOS. However, direct comparisons across these agents are limited. This study compared the effects of metformin, rosiglitazone and pioglitazone in the management of PCOS to inform the 2023 International Evidence-based PCOS Guideline. DESIGN: Systematic review and meta-analysis of the literature. PATIENTS: Women with PCOS and treatment with insulin sensitisers. MEASUREMENTS: Hormonal and clinical outcomes, as well as side effects. RESULTS: Of 1660 publications identified, 13 randomised controlled trials were included. Metformin was superior in lowering weight (mean difference [MD]: -4.39, 95% confidence interval [CI]: -7.69 to -1.08 kg), body mass index (MD: -0.95, 95% CI: -1.41 to -0.49 kg/m2 ) and testosterone (MD: -0.10, 95% CI: -0.18 to -0.03 nmol/L) versus rosiglitazone, whereas there was no difference when comparing metformin to pioglitazone. Adding rosiglitazone or pioglitazone to metformin did not improve metabolic outcomes. However, rosiglitazone seemed superior to metformin in lowering lipid concentrations. CONCLUSIONS: Metformin should remain the first-line insulin sensitising treatment in adults with PCOS for the prevention and management of weight and metabolic features. The addition of thiazolidinediones appears to offer little benefit.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Metformina , Síndrome do Ovário Policístico , Tiazolidinedionas , Adulto , Humanos , Feminino , Rosiglitazona/uso terapêutico , Hipoglicemiantes/uso terapêutico , Pioglitazona/uso terapêutico , Síndrome do Ovário Policístico/tratamento farmacológico , Insulina/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Metformina/uso terapêutico , Tiazolidinedionas/uso terapêutico
5.
Immunol Cell Biol ; 101(5): 428-443, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36918728

RESUMO

Macrophages exhibit a range of functional pro- and anti-inflammatory states that induce changes in their cellular metabolism. We aimed to elucidate whether these changes affect the molecular properties of their circadian clock focusing on their anti-inflammatory phenotype. Primary cell cultures of bone marrow-derived macrophages (BMDMs; nonpolarized M0 BMDM) from PER2::LUC (fusion protein of PERIOD2 and LUCIFERASE) mice were polarized into the M1 (proinflammatory) or M2 (anti-inflammatory) phenotype, and PER2-driven bioluminescence was recorded in real-time at the cell-population and single-cell levels. Viability, clock gene expression profiles, polarization plasticity and peroxisome proliferator-activated receptor γ (PPARγ) protein levels were analyzed. The effects of pharmacological activation/inhibition of PPARγ (rosiglitazone/GW9662) and inhibition of fatty acid oxidation (FAO) by etomoxir in M2 BMDM cell cultures were examined. The parameters of PER2-driven bioluminescence rhythms differed between M0, M1 and M2 BMDM cultures at cell-population and single-cell levels. Compared with M0, polarization to M2 did not change the period but increased amplitude, mean bioluminescence level and rhythm persistence. Polarization to M1 shortened the period but had no effect on the amplitude of the rhythm. The same period changes were observed after a bidirectional switch between M1- and M2-polarized states in the same culture. Both PPARγ activation/inhibition and FAO inhibition modulated the clock in M2 BMDMs, suggesting metabolic regulation of the M2 clock. Our results indicate that bidirectional changes in the properties of BMDM circadian clocks in response to their actual polarization are mediated via changes in their metabolic state. They provide new information on the interrelationship between the BMDM polarization, their circadian clock and cellular metabolism.


Assuntos
Relógios Circadianos , Camundongos , Animais , PPAR gama/metabolismo , Macrófagos/metabolismo , Rosiglitazona/metabolismo , Anti-Inflamatórios/metabolismo
6.
Exp Eye Res ; 231: 109498, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37169280

RESUMO

Oxidative stress is closely associated with diabetes and can cause free radical accumulation and eventually lead to ocular surface tissue damage. The purpose of this study was to investigate peroxisome proliferator-activated receptor-γ (PPARγ) expression in the lacrimal gland (LG), meibomian gland, and cornea of diabetes-related dry eye mice and whether the PPARγ agonist rosiglitazone can alleviate the oxidative stress of the ocular surface, thereby improving the condition of diabetes-related dry eye. Quantitative RT-PCR (Q-PCR) showed that the PPARγ, catalase, glutathione peroxidase 3, and heme oxygenase-1 (HO-1) mRNA expression levels in the LG of diabetes-related dry eye mice decreased at 8 and 12 weeks. In addition, the increased levels of oxidative stress were confirmed by western blot. Although the mRNA expression levels of antioxidant enzymes in the cornea and meibomian gland decreased at 8 weeks, some of them recovered by 12 weeks. Rosiglitazone alleviated ocular surface damage and increased corneal sensitivity and tear production in diabetes-related dry eye mice. Moreover, the reactive oxygen species accumulation was reduced and the PPARγ, HO-1, and glutathione peroxidase 3 mRNA expression levels were increased in the LG. The PPARγ, HO-1, translocase of the outer membrane 20, and mitochondrial transcription factor A protein levels were also significantly increased. These results demonstrated that rosiglitazone reduced oxidative stress in the LG of diabetes-related dry eye mice, at least in part, by activating PPARγ to up-regulate antioxidant enzyme expression.


Assuntos
Diabetes Mellitus , Síndromes do Olho Seco , Camundongos , Animais , PPAR gama/genética , Rosiglitazona/farmacologia , Antioxidantes/farmacologia , Estresse Oxidativo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Neurochem Res ; 48(9): 2870-2880, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37204549

RESUMO

A growing body of evidence has shown that seizure can trigger inflammatory cascades through increasing the expression of several inflammatory cytokines. It has been proved that peroxisome proliferator-activated receptor-γ agonists have immunomodulatory, anti-inflammatory, and neuroprotective effects beyond the putative hypoglycemic effects. Thus, we investigated the inhibitory effect of rosiglitazone on the development of pentylenetetrazol (PTZ)-induced kindling via affecting the inflammatory pathway. Male C57BL/6 mice were randomly divided into vehicle group (0.1% DMSO), PTZ-group and rosiglitazone-PTZ-group. Kindling was induced by the administration of PTZ (40 mg/kg, i.p) every other day and mice were observed for 20 min after each PTZ injection. Twenty-four hours after the last dose, animals were euthanized and hippocampus was isolated. The level of Malondialdehyde (MDA), Superoxide Dismutase (SOD), and Catalase (CAT) activity were quantified in hippocampus by biochemical methods. The protein levels of IL-1ß, IL-6, IL-10, IFN-γ, TNF-α, caspase-3, iNOS, PPAR-γ, Bcl-2, or Bax factors were measured with western blotting. Also, the quantitative real-time PCR were used to evaluate the mRNA expression of those factors. Pretreatment with rosiglitazone significantly prevented the progression of kindling in comparison with control group. The rosiglitazone significantly decreased the MDA level and increased the CAT, and SOD levels in the rosiglitazone treated mice compared to those in the PTZ group (P < 0.01). Using real-time PCR and Western blotting assay, similar results were obtained. The expression levels of IL-1ß, IL-6, IL-10, IFN-γ, TNF-α, Bax or PPAR-γ were significantly changed in the brain. The results of this study suggest that effect of rosiglitazone may be crucial in its ability to protect against the neuronal damage caused by PTZ induced seizure.


Assuntos
Excitação Neurológica , Pentilenotetrazol , Animais , Masculino , Camundongos , Antioxidantes/farmacologia , Proteína X Associada a bcl-2/metabolismo , Citocinas/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Pentilenotetrazol/toxicidade , PPAR gama/metabolismo , Agonistas PPAR-gama , Piroptose , Rosiglitazona/farmacologia , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Pharmacol Res ; 187: 106638, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36586645

RESUMO

BACKGROUND AND PURPOSE: Selective peroxisome proliferator-activated receptors (PPARs) are widely used to treat metabolic complications; however, the limited effect of PPARα agonists on glucose metabolism and the adverse effects associated with selective PPARγ activators have stimulated the development of novel pan-PPAR agonists to treat metabolic disorders. Here, we synthesized a new prenylated benzopyran (BP-2) and evaluated its PPAR-activating properties, anti-inflammatory effects and impact on metabolic derangements. EXPERIMENTAL APPROACH: BP-2 was used in transactivation assays to evaluate its agonism to PPARα, PPARß/δ and PPARγ. A parallel-plate flow chamber was employed to investigate its effect on TNFα-induced leukocyte-endothelium interactions. Flow cytometry and immunofluorescence were used to determine its effects on the expression of endothelial cell adhesion molecules (CAMs) and chemokines and p38-MAPK/NF-κB activation. PPARs/RXRα interactions were determined using a gene silencing approach. Analysis of its impact on metabolic abnormalities and inflammation was performed in ob/ob mice. KEY RESULTS: BP-2 displayed strong PPARα activity, with moderate and weak activity against PPARß/δ and PPARγ, respectively. In vitro, BP-2 reduced TNFα-induced endothelial ICAM-1, VCAM-1 and fractalkine/CX3CL1 expression, suppressed mononuclear cell arrest via PPARß/δ-RXRα interactions and decreased p38-MAPK/NF-κB activation. In vivo, BP-2 improved the circulating levels of glucose and triglycerides in ob/ob mice, suppressed T-lymphocyte/macrophage infiltration and proinflammatory markers in the liver and white adipose tissue, but increased the expression of the M2-like macrophage marker CD206. CONCLUSION AND IMPLICATIONS: BP-2 emerges as a novel pan-PPAR lead candidate to normalize glycemia/triglyceridemia and minimize inflammation in metabolic disorders, likely preventing the development of further cardiovascular complications.


Assuntos
Doenças Metabólicas , PPAR delta , PPAR beta , Camundongos , Animais , PPAR gama/metabolismo , PPAR alfa/metabolismo , PPAR beta/metabolismo , Fator de Necrose Tumoral alfa , Benzopiranos , NF-kappa B , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico
9.
Pharmacol Res ; 192: 106786, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37146924

RESUMO

Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of disease phenotypes which start with simple steatosis and lipid accumulation in the hepatocytes - a typical histological lesions characteristic. It may progress to non-alcoholic steatohepatitis (NASH) that is characterized by hepatic inflammation and/or fibrosis and subsequent onset of NAFLD-related cirrhosis and hepatocellular carcinoma (HCC). Due to the central role of the liver in metabolism, NAFLD is regarded as a result of and contribution to the metabolic abnormalities seen in the metabolic syndrome. Peroxisome proliferator-activated receptors (PPARs) has three subtypes, which govern the expression of genes responsible for energy metabolism, cellular development, inflammation, and differentiation. The agonists of PPARα, such as fenofibrate and clofibrate, have been used as lipid-lowering drugs in clinical practice. Thiazolidinediones (TZDs) - ligands of PPARγ, such as rosiglitazone and pioglitazone, are also used in the treatment of type 2 diabetes (T2D) with insulin resistance (IR). Increasing evidence suggests that PPARß/δ agonists have potential therapeutic effects in improving insulin sensitivity and lipid metabolism disorders. In addition, PPARs ligands have been considered as potential therapeutic drugs for hypertension, atherosclerosis (AS) or diabetic nephropathy. Their crucial biological roles dictate the significance of PPARs-targeting in medical research and drug discovery. Here, it reviews the biological activities, ligand selectivity and biological functions of the PPARs family, and discusses the relationship between PPARs and the pathogenesis of NAFLD and metabolic syndrome. This will open new possibilities for PPARs application in medicine, and provide a new idea for the treatment of fatty liver and related diseases.


Assuntos
Carcinoma Hepatocelular , Diabetes Mellitus Tipo 2 , Neoplasias Hepáticas , Síndrome Metabólica , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Síndrome Metabólica/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , PPAR alfa/metabolismo , Inflamação/metabolismo , Lipídeos/uso terapêutico
10.
Clin Exp Pharmacol Physiol ; 50(12): 927-935, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37675456

RESUMO

Ferroptosis is a recently discovered non-apoptotic form of cellular death. Acyl-CoA synthetase long-chain family number 4 (ACSL4) is necessary for iron-dependent cellular death, and reactive oxygen species (ROS) produced by ACSL4 are the executioners of ferroptosis. Rosiglitazone improves ferroptosis by inhibiting ACSL4. There is no research indicating whether ACSL4 plays a role in cell death after surgical brain injury (SBI). This study aimed to investigate the role of ACSL4 in SBI via the ferroptosis pathway. Ninety male Sprague-Dawley rats were examined using a model of SBI. Subsequently, the inhibitory effect of rosiglitazone on ACSL4 was assessed via western blot, real-time polymerase chain reaction (PCR), immunofluorescence, fluoro-jade C staining, Perl's staining, ROS assay, and neurological scoring. The results showed that compared with the Sham group, the protein levels of ACSL4 and transferrin were significantly increased after SBI. Administration of rosiglitazone significantly reduced neuronal necrosis, iron deposition, brain water content and ROS in brain tissue and ameliorated neurological deficits at 48 h after SBI, which was concomitant with decreased transferrin expression. These findings demonstrate that SBI-induced upregulation of ACSL4 may be partly mediated by the ferroptosis pathway, which can be reversed by rosiglitazone administration.


Assuntos
Lesões Encefálicas , Neoplasias Encefálicas , Ratos , Masculino , Animais , Rosiglitazona/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ratos Sprague-Dawley , Encéfalo/metabolismo , Lesões Encefálicas/tratamento farmacológico , Ferro , Transferrinas/metabolismo , Ligases/metabolismo
11.
Gynecol Endocrinol ; 39(1): 2254848, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37706434

RESUMO

OBJECTIVE: GuizhiFulingWan (GFW) has been reported to be effective against polycystic ovary syndrome (PCOS) by possessing oxidative stress and inflammation which related to PI3K/AKT/NF-κB, Nrf2/HO-1 pathway. This study aims to probe the effects and mechanisms of GFW combined with rosiglitazone on PCOS via PI3K/AKT/NF-κB and Nrf2/HO-1 pathways. METHODS: A rat PCOS model established by dehydroepiandrosterone (DHEA) injection. The experiment was allocated to control, DHEA, GFW, rosiglitazone, GFW + rosiglitazone groups. Treatment for 30 days, we monitored weight and ovarian weight of rats. Fasting blood glucose (FBG), fasting insulin (FINS), homeostasis model assessment of insulin resistance (HOMA-IR), lipid metabolism indexes, estrous cycle and sex hormone-, inflammation-, oxidative stress-related factors were examined. Hematoxylin&eosin staining assessed ovarian tissue pathological changes. Western blot determined PI3K/AKT/NF-κB, Nrf2/HO-1 pathways-related markers. RESULTS: GFW and rosiglitazone treatment suppressed body weight and ovarian weight in PCOS rats. They also decreased FBG, FINS, HOMA-IR while inhibited total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL) and enhanced high-density lipoprotein (HDL). They ameliorated estrous cycle, ovarian histological changes and follicular development. They restrained testosterone (T), luteinizing hormone (LH) and accelerated estradiol (E2), progesterone (P), follicle stimulating hormone (FSH). They inhibited glutathione peroxidase (GSH-Px), malondialdehyde (MDA), superoxide dismutase (SOD) in serum while increased GSH-Px, SOD and decrease MDA in ovarian tissues. They reduced C-reactive protein, interleukin-18 (IL-18), tumor necrosis factor-α (TNF-α), IL-6, IL-1ß levels. GFW and rosiglitazone co-intervention regulated PI3K/AKT/NF-κB and Nrf2/HO-1 pathways in PCOS rats. CONCLUSION: GFW alleviated ovarian dysfunction in PCOS rats, which may be related to the PI3K/AKT/NF-κB, Nrf2/HO-1 pathways.


Assuntos
NF-kappa B , Síndrome do Ovário Policístico , Feminino , Humanos , Animais , Ratos , Síndrome do Ovário Policístico/tratamento farmacológico , Fator 2 Relacionado a NF-E2 , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Rosiglitazona/farmacologia , Rosiglitazona/uso terapêutico , Desidroepiandrosterona
12.
Anim Biotechnol ; 34(8): 3708-3717, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37149785

RESUMO

Intramuscular fat (IMF) positively influences various aspects of meat quality, while the subcutaneous fat (SF) has negative effect on carcass characteristics and fattening efficiency. Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of adipocyte differentiation, herein, through bioinformatic screen for the potential regulators of adipogenesis from two independent microarray datasets, we identified that PPARγ is a potentially regulator between porcine IMF and SF adipogenesis. Then we treated subcutaneous preadipocytes (SA) and intramuscular preadipocytes (IMA) of pig with RSG (1 µmol/L), and we found that RSG treatment promoted the differentiation of IMA via differentially activating PPARγ transcriptional activity. Besides, RSG treatment promoted apoptosis and lipolysis of SA. Meanwhile, by the treatment of conditioned medium, we excluded the possibility of indirect regulation of RSG from myocyte to adipocyte and proposed that AMPK may mediate the RSG-induced differential activation of PPARγ. Collectively, the RSG treatment promotes IMA adipogenesis, and advances SA lipolysis, this effect may be associated with AMPK-mediated PPARγ differential activation. Our data indicates that targeting PPARγ might be an effective strategy to promote intramuscular fat deposition while reduce subcutaneous fat mass of pig.


Assuntos
Adipogenia , PPAR gama , Suínos , Animais , Adipogenia/fisiologia , Rosiglitazona/farmacologia , PPAR gama/genética , Proteínas Quinases Ativadas por AMP/farmacologia , Adipócitos/fisiologia , Diferenciação Celular
13.
Genes Dev ; 29(1): 7-22, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25504365

RESUMO

Long-term exposure to peroxisome proliferator-activated receptor γ (PPARγ) agonists such as rosiglitazone induces browning of rodent and human adipocytes; however, the transcriptional mechanisms governing this phenotypic switch in adipocytes are largely unknown. Here we show that rosiglitazone-induced browning of human adipocytes activates a comprehensive gene program that leads to increased mitochondrial oxidative capacity. Once induced, this gene program and oxidative capacity are maintained independently of rosiglitazone, suggesting that additional browning factors are activated. Browning triggers reprogramming of PPARγ binding, leading to the formation of PPARγ "superenhancers" that are selective for brown-in-white (brite) adipocytes. These are highly associated with key brite-selective genes. Based on such an association, we identified an evolutionarily conserved metabolic regulator, Kruppel-like factor 11 (KLF11), as a novel browning transcription factor in human adipocytes that is required for rosiglitazone-induced browning, including the increase in mitochondrial oxidative capacity. KLF11 is directly induced by PPARγ and appears to cooperate with PPARγ in a feed-forward manner to activate and maintain the brite-selective gene program.


Assuntos
Adipócitos/metabolismo , Proteínas de Ciclo Celular/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Repressoras/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos Marrons/citologia , Proteínas Reguladoras de Apoptose , Proteínas de Ciclo Celular/genética , Reprogramação Celular , Cromatina/metabolismo , Regulação da Expressão Gênica , Humanos , Hipoglicemiantes/farmacologia , Mitocôndrias/efeitos dos fármacos , Oxirredução , Ligação Proteica , Proteínas Repressoras/genética , Rosiglitazona , Tiazolidinedionas/farmacologia , Ativação Transcricional/efeitos dos fármacos
14.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36835405

RESUMO

Type 2 diabetes (T2D) impairs post-stroke recovery, and the underlying mechanisms are unknown. Insulin resistance (IR), a T2D hallmark that is also closely linked to aging, has been associated with impaired post-stroke recovery. However, whether IR worsens stroke recovery is unknown. We addressed this question in mouse models where early IR, with or without hyperglycemia, was induced by chronic high-fat diet feeding or sucrose supplementation in the drinking water, respectively. Furthermore, we used 10-month-old mice, spontaneously developing IR but not hyperglycemia, where IR was normalized pharmacologically pre-stroke with Rosiglitazone. Stroke was induced by transient middle cerebral artery occlusion and recovery was assessed by sensorimotor tests. Neuronal survival, neuroinflammation and the density of striatal cholinergic interneurons were also assessed by immunohistochemistry/quantitative microscopy. Pre-stroke induction and normalization of IR, respectively, worsened and improved post-stroke neurological recovery. Moreover, our data indicate a potential association of this impaired recovery with exacerbated neuroinflammation and a decreased density of striatal cholinergic interneurons. The global diabetes epidemic and population aging are dramatically increasing the percentage of people in need of post-stroke treatment/care. Our results suggest that future clinical studies should target pre-stroke IR to reduce stroke sequelae in both diabetics and elderly people with prediabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperglicemia , Resistência à Insulina , Acidente Vascular Cerebral , Camundongos , Animais , Doenças Neuroinflamatórias , Infarto da Artéria Cerebral Média
15.
Int J Mol Sci ; 24(7)2023 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-37047198

RESUMO

Traumatic Brain Injury (TBI) is a primary cause of cerebrovascular and neurological disorders worldwide. The current scientific researchers believe that premorbid conditions such as tobacco smoking (TS) can exacerbate post-TBI brain injury and negatively affect recovery. This is related to vascular endothelial dysfunction resulting from the exposure to TS-released reactive oxygen species (ROS), nicotine, and oxidative stress (OS) stimuli impacting the blood-brain barrier (BBB) endothelium. Interestingly, these pathogenic modulators of BBB impairment are similar to those associated with hyperglycemia. Antidiabetic drugs such as metformin (MF) and rosiglitazone (RSG) were shown to prevent/reduce BBB damage promoted by chronic TS exposure. Thus, using in vivo approaches, we evaluated the effectiveness of post-TBI treatment with MF or RSG to reduce the TS-enhancement of BBB damage and brain injury after TBI. For this purpose, we employed an in vivo weight-drop TBI model using male C57BL/6J mice chronically exposed to TS with and without post-traumatic treatment with MF or RSG. Our results revealed that these antidiabetic drugs counteracted TS-promoted downregulation of nuclear factor erythroid 2-related factor 2 (NRF2) expression and concomitantly dampened TS-enhanced OS, inflammation, and loss of BBB integrity following TBI. In conclusion, our findings suggest that MF and RSG could reduce the harmful impact of chronic smoking on post-traumatic brain injuries.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Metformina , Camundongos , Animais , Masculino , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Hipoglicemiantes/metabolismo , Camundongos Endogâmicos C57BL , Barreira Hematoencefálica/metabolismo , Fumar Tabaco , Rosiglitazona/farmacologia , Metformina/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo
16.
Korean J Physiol Pharmacol ; 27(1): 95-103, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36575937

RESUMO

Rosiglitazone is a thiazolidinedione-class antidiabetic drug that reduces blood glucose and glycated hemoglobin levels. We here investigated the interaction of rosiglitazone with Kv3.1 expressed in Chinese hamster ovary cells using the whole-cell patch-clamp technique. Rosiglitazone rapidly and reversibly inhibited Kv3.1 currents in a concentration-dependent manner (IC50 = 29.8 µM) and accelerated the decay of Kv3.1 currents without modifying the activation kinetics. The rosiglitazone-mediated inhibition of Kv3.1 channels increased steeply in a sigmoidal pattern over the voltage range of -20 to +30 mV, whereas it was voltage-independent in the voltage range above +30 mV, where the channels were fully activated. The deactivation of Kv3.1 current, measured along with tail currents, was also slowed by the drug. In addition, the steady-state inactivation curve of Kv3.1 by rosiglitazone shifts to a negative potential without significant change in the slope value. All the results with the use dependence of the rosiglitazone-mediated blockade suggest that rosiglitazone acts on Kv3.1 channels as an open channel blocker.

17.
Eur J Neurosci ; 56(9): 5727-5757, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35128745

RESUMO

Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) commonly co-occur. T2DM increases the risk for AD by approximately twofold. Animal models provide one means of interrogating the relationship of T2DM to AD and investigating brain insulin resistance in the pathophysiology of AD. Animal models show that persistent hyperglycaemia results in chronic low-grade inflammation that may contribute to the development of neuroinflammation and accelerate the pathobiology of AD. Epidemiological studies suggest that patients with T2DM who received treatment with specific anti-diabetic agents have a decreased risk for the occurrence of AD and all-cause dementia. Agents such as metformin ameliorate T2DM and may have other important systemic effects that lower the risk of AD. Glucagon-like peptide 1 (GLP-1) agonists have been associated with a decreased risk for AD in patients with T2DM. Both insulin and non-insulin anti-diabetic treatments have been evaluated for the treatment of AD in clinical trials. In most cases, patients included in the trials have clinical features of AD but do not have T2DM. Many of the trials were conducted prior to the use of diagnostic biomarkers for AD. Trials have had a wide range of durations and population sizes. Many of the agents used to treat T2DM do not cross the blood brain barrier, and the effects are posited to occur via lowering of peripheral hyperglycaemia and reduction of peripheral and central inflammation. Clinical trials of anti-diabetic agents to treat AD are ongoing and will provide insight into the therapeutic utility of these agents.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Hiperglicemia , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Doença de Alzheimer/tratamento farmacológico , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Fatores de Risco , Hiperglicemia/induzido quimicamente , Hiperglicemia/complicações , Hiperglicemia/tratamento farmacológico , Inflamação/complicações
18.
J Virol ; 95(18): e0083521, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34190599

RESUMO

Recruitment of immune cells from the periphery is critical for controlling West Nile virus (WNV) growth in the central nervous system (CNS) and preventing subsequent WNV-induced CNS disease. Neuroinflammatory responses, including the release of proinflammatory cytokines and chemokines by CNS cells, influence the entry and function of peripheral immune cells that infiltrate the CNS. However, these same cytokines and chemokines contribute to tissue damage in other models of CNS injury. Rosiglitazone is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist that inhibits neuroinflammation. We used rosiglitazone in WNV-infected ex vivo brain slice cultures (BSC) to investigate the role of neuroinflammation within the CNS in the absence of peripheral immune cells. Rosiglitazone treatment inhibited WNV-induced expression of proinflammatory chemokines and cytokines, interferon beta (IFN-ß), and IFN-stimulated genes (ISG) and also decreased WNV-induced activation of microglia. These decreased neuroinflammatory responses were associated with activation of astrocytes, robust viral growth, increased activation of caspase 3, and increased neuronal loss. Rosiglitazone had a similar effect on in vivo WNV infection, causing increased viral growth, tissue damage, and disease severity in infected mice, even though the number of infiltrating peripheral immune cells was higher in rosiglitazone-treated, WNV-infected mice than in untreated, infected controls. These results indicate that local neuroinflammatory responses are capable of controlling viral growth within the CNS and limiting neuronal loss and may function to keep the virus in check prior to the infiltration of peripheral immune cells, limiting both virus- and immune-mediated neuronal damage. IMPORTANCE West Nile virus is the most common cause of epidemic encephalitis in the United States and can result in debilitating CNS disease. There are no effective vaccines or treatments for WNV-induced CNS disease in humans. The peripheral immune response is critical for protection against WNV CNS infections. We now demonstrate that intrinsic immune responses also control viral growth and limit neuronal loss. These findings have important implications for developing new therapies for WNV-induced CNS disease.


Assuntos
Encéfalo/imunologia , Morte Celular , Doenças do Sistema Nervoso Central/prevenção & controle , Imunidade Inata/imunologia , Neurônios/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Doenças do Sistema Nervoso Central/etiologia , Doenças do Sistema Nervoso Central/patologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Masculino , Camundongos , Neurônios/patologia , Neurônios/virologia , Febre do Nilo Ocidental/complicações , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/virologia
19.
Toxicol Appl Pharmacol ; 450: 116142, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35777529

RESUMO

Arsenite (As+3) is a group one human carcinogen, which has been associated with many diseases. Previous studies indicated that As+3 could inhibit wound healing and repair. M2a cells are known as tissue remodeling macrophages, which play an important role in wound repair process. Peroxisome proliferator-activated receptor gamma (PPAR-γ), a key regulator of lipid and glucose metabolism, was found to mediate the IL-4-dependent M2a polarization of macrophages. In the present study, As+3 induced dose-dependent inhibition of M2a polarization starting from 0.1 µM in THP-1-derived macrophages stimulated with 20 ng/mL IL-4. Increased lipid accumulation and decreased PPAR-γ expression were also observed in As+3-treated M2a macrophages. Rosiglitazone (RSG), a potent PPAR-γ agonist, alleviated the suppressions of PPAR-γ and M2a polarization induced by 2 µM As+3. Collectively, these results not only demonstrated that As+3 was able to inhibit polarization of M2a cells through PPAR-γ suppression, but also indicated that PPAR-γ could be utilized as a target for the prevention and treatment of As+3-induced immunotoxicity.


Assuntos
Arsenitos , Tiazolidinedionas , Arsenitos/metabolismo , Arsenitos/toxicidade , Regulação para Baixo , Humanos , Interleucina-4/metabolismo , Lipídeos , Macrófagos/metabolismo , PPAR gama/metabolismo
20.
Int Arch Allergy Immunol ; 183(9): 997-1006, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35526523

RESUMO

PURPOSE: Vascular endothelial hyperpermeability and barrier disruption are involved in the initiation and development of sepsis. M1 macrophages promote inflammation in sepsis by releasing pro-inflammatory cytokines and chemokines. This study was designed to investigate the functional relationships between M1 macrophages and human umbilical vein endothelial cells (HUVECs), as well as the underlying molecular mechanisms. METHODS: HUVECs were co-cultured with THP-1-derived M1 macrophages pretreated with or without rosiglitazone (RSG), a peroxisome proliferator-activated receptor (PPAR)-γ agonist. C-X-C chemokine receptor type (CXCR)5 was knocked down by short hairpin RNA lentivirus. Cecal ligation and puncture were used to induce sepsis in a mouse model. Endothelial permeability was evaluated using transendothelial electrical resistance and fluorescein isothiocyanate (FITC)-dextran assays. RESULTS: Chemokine ligand (CXCL)13 was upregulated in M1 macrophages than M0 macrophages, as well as in the culture medium. In HUVECs co-cultured with M1 macrophages, transendothelial electrical resistance decreased, FITC-dextran flux increased, p38 phosphorylation was strengthened, and the expression of tight junction proteins (zonula occludens protein-1, occludin, and claudin-4) decreased. CXCR5 RNA interference or RSG pretreatment partially reversed these effects. A luciferase reporter assay revealed that CXCL13 was a direct target of PPAR-γ. RSG treatment decreased serum levels of creatinine, blood urea nitrogen, CXCL13, tumor necrosis factor-α, and interleukin-6, downregulated CXCL13 in peritoneal macrophages, and enhanced the survival rate of sepsis mice. CONCLUSION: M1 macrophages induced endothelial hyperpermeability and promoted p38 phosphorylation in sepsis by inhibiting PPAR-γ to increase CXCL13 production. PPAR-γ/CXCL13-CXCR5 signaling could be a promising novel therapeutic target for sepsis.


Assuntos
PPAR gama , Sepse , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Quimiocina CXCL13/metabolismo , Quimiocina CXCL13/farmacologia , Células Endoteliais/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , PPAR gama/metabolismo , Permeabilidade , Fosforilação , Receptores CXCR5/metabolismo , Sepse/tratamento farmacológico , Sepse/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA