Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int J Biol Sci ; 20(1): 113-126, 2024.
Article in English | MEDLINE | ID: mdl-38164174

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a global health burden closely linked to insulin resistance, obesity, and type 2 diabetes. The complex pathophysiology of NAFLD involves multiple cellular pathways and molecular factors. Nuclear receptors (NRs) have emerged as crucial regulators of lipid metabolism and inflammation in NAFLD, offering potential therapeutic targets for NAFLD. Targeting PPARs and FXRs has shown promise in ameliorating NAFLD symptoms and halting disease progression. However, further investigation is needed to address side effects and personalize therapy approaches. This review summarizes the current understanding of the involvement of NRs in the pathogenesis of NAFLD and explores their therapeutic potential. We discuss the role of several NRs in modulating lipid homeostasis in the liver, including peroxisome proliferator-activated receptors (PPARs), liver X receptors (LXRs), farnesoid X receptors (FXRs), REV-ERB, hepatocyte nuclear factor 4α (HNF4α), constitutive androstane receptor (CAR) and pregnane X receptor (PXR).The expanding knowledge of NRs in NAFLD offers new avenues for targeted therapies, necessitating exploration of novel treatment strategies and optimization of existing approaches to combat this increasingly prevalent disease.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Liver/metabolism
2.
Clin Nutr ESPEN ; 58: 311-319, 2023 12.
Article in English | MEDLINE | ID: mdl-38057021

ABSTRACT

BACKGROUND: This study investigated the effects of oleoylethanolamide (OEA) supplementation on the expression levels of SIRT1, AMPK, PGC-1α, PPAR-γ, CEBP-α and CEBP-ß genes and serum neuregulin 4 (NRG4) levels in patients with non-alcoholic fatty liver diseases (NAFLD). METHODS: Sixty obese patients with NAFLD were equally allocated into either OEA or placebo group for 12 weeks. The mRNA expression levels of genes were determined using the reverse transcription polymerase chain reaction (RT-PCR) technique. Serum NRG4 level was also assessed using an enzyme-linked immunosorbent assay (ELISA) kit. RESULTS: At the endpoint, mRNA expression levels of SIRT1(p = 0.001), PGC-1α (p = 0.011) and AMPK (p = 0.019) were significantly higher in the OEA group compared to placebo group. However, no significant differences were observed in the expression levels of PPAR-γ, CEBP-α and CEBP-ß between the two groups. Serum NRG4 levels significantly increased in the OEA group compared with the placebo group after controlling for confounders (p = 0.027). In the OEA group, significant relationships were found between percent of changes in the expression levels of the SIRT1, AMPK and PGC-1α as well as serum NRG4 level with percent of changes in some anthropometric measures. Moreover, in the intervention group, percent of changes in high-density lipoprotein cholesterol was positively correlated with percent of changes in the expression levels of the SIRT1 and AMPK. While, percent of changes in triglyceride was inversely correlated with percent of changes in the expression levels of SIRT1. CONCLUSION: OEA could beneficially affect expression levels of some lipid metabolism-related genes and serum NRG4 level. "REGISTERED UNDER IRANIAN REGISTRY OF CLINICAL TRIALS IDENTIFIER NO: IRCT20090609002017N32".


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Lipid Metabolism/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 1/therapeutic use , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Iran , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/therapeutic use , Neuregulins/metabolism , Neuregulins/therapeutic use , RNA, Messenger/metabolism , RNA, Messenger/therapeutic use , Dietary Supplements
3.
Clin Res Hepatol Gastroenterol ; 47(10): 102231, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865226

ABSTRACT

INTRODUCTION: Linalool is a monoterpene that occurs naturally in various aromatic plants and is identified in our previous study as a potential candidate for protection against high-fat diet (HFD)-induced metabolic dysfunction-associated steatotic liver disease (MASLD). However, little is known about its direct effects on hepatic lipid metabolism and oxidative stress. Therefore, this study aims to investigate the therapeutic effect of linalool against MASLD and the underlying mechanism. METHODS: To establish a rat model of MASLD, male Wistar rats were fed HFD for 16 weeks and orally administered linalool (100 mg/kg body weight) for 45 days starting from week 14. RESULTS: Linalool significantly reduced HFD-induced liver lipid accumulation and restored altered adipokine levels. Mechanistically, linalool downregulated the mRNA expression of sterol regulatory element binding protein 1 and its lipogenesis target genes fatty acid synthase and acetyl-CoA carboxylase, and upregulated the mRNA expression of genes involved in fatty acid oxidation (peroxisome proliferator-activated receptor (PPAR)-alpha [PPAR-α], lipoprotein lipase and protein kinase B [Akt]) as well as the upstream mediators sirtuin 1 (Sirt1) and AMP-activated protein kinase (AMPK) in the liver of MASLD rats. In addition, linalool also curbed oxidative stress by increasing antioxidant enzymes and activating nuclear erythroid-2-related factor 2 (Nrf-2) and its downstream target genes involved in antioxidant properties. CONCLUSION: Therefore, this study concludes that linalool attenuates lipid accumulation in the liver by inhibiting de novo lipogenesis, promoting fatty acid oxidation, and attenuating oxidative stress by regulating Sirt1/Akt/PPRA-α/AMPK and Nrf-2/ HO-1 signaling pathways.


Subject(s)
Fatty Liver , Non-alcoholic Fatty Liver Disease , Rats , Animals , Sirtuin 1/metabolism , Sirtuin 1/pharmacology , Sirtuin 1/therapeutic use , Proto-Oncogene Proteins c-akt , AMP-Activated Protein Kinases/metabolism , Antioxidants/therapeutic use , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Rats, Wistar , Liver/metabolism , Lipid Metabolism , Signal Transduction , Oxidative Stress , Fatty Acids , Lipids , RNA, Messenger/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy
4.
Clin Gastroenterol Hepatol ; 21(8): 2076-2087, 2023 07.
Article in English | MEDLINE | ID: mdl-36809835

ABSTRACT

Primary biliary cholangitis (PBC) is an archetypal autoimmune disease. Chronic lymphocytic cholangitis is associated with interface hepatitis, ductopenia, cholestasis, and progressive biliary fibrosis. People living with PBC are frequently symptomatic, experiencing a quality-of-life burden dominated by fatigue, itch, abdominal pain, and sicca complex. Although the female predominance, specific serum autoantibodies, immune-mediated cellular injury, as well as genetic (HLA and non-HLA) risk factors, identify PBC as autoimmune, to date treatment has focused on cholestatic consequences. Biliary epithelial homeostasis is abnormal and contributes to disease. The impact of cholangiocyte senescence, apoptosis, and impaired bicarbonate secretion enhances chronic inflammation and bile acid retention. First-line therapy is a non-specific anti-cholestatic agent, ursodeoxycholic acid. For those with residual cholestasis biochemically, obeticholic acid is introduced, and this semisynthetic farnesoid X receptor agonist adds choleretic, anti-fibrotic, and anti-inflammatory activity. Future PBC licensed therapy will likely include peroxisome proliferator activated receptor (PPAR) pathway agonists, including specific PPAR-delta agonism (seladelpar), as well as elafibrinor and saroglitazar (both with broader PPAR agonism). These agents dovetail the clinical and trial experience for off-label bezafibrate and fenofibrate use. Symptom management is essential, and encouragingly, PPAR agonists reduce itch; IBAT inhibition (eg, linerixibat) also appears promising for pruritus. For those where liver fibrosis is the target, NOX inhibition is being evaluated. Earlier stage therapies in development include therapy to impact immunoregulation in patients, as well other approaches to treating pruritus (eg, antagonists of MrgprX4). Collectively the PBC therapeutic landscape is exciting. Therapy goals are increasingly proactive and individualized and aspire to rapidly achieve normal serum tests and quality of life with prevention of end-stage liver disease.


Subject(s)
Cholangitis , Cholestasis , Liver Cirrhosis, Biliary , Humans , Female , Male , Liver Cirrhosis, Biliary/drug therapy , Liver Cirrhosis, Biliary/complications , Quality of Life , Peroxisome Proliferator-Activated Receptors/therapeutic use , Ursodeoxycholic Acid/therapeutic use , Cholangitis/diagnosis , Pruritus/etiology
5.
Fundam Clin Pharmacol ; 37(3): 629-638, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36649007

ABSTRACT

The current study was designed to evaluate the therapeutic potential of beta-glucan (BG), which is a key bioactive compound predominantly present in mushrooms and cereals against high-fat-diet (HFD)-induced obesity, and to understand its mechanism of action. Obesity was induced in rats by supplementing the diet with HFD and BG (40 mg/kg body weight) for a period of 6 weeks. At the end of the experimental period, the body weight, as well as hyperglycemic, dyslipidemia, and obesogenic marker expressions, was assessed in the control group and in the experimental obese rats. Administration of BG to obese rats significantly reduced body weight gain and attenuated hyperglycemia, which was confirmed by the decreased blood glucose and insulin resistance. At the same time, BG mitigated dyslipidemia by altering expressions of peroxisome proliferator-activated receptor-gamma (PPAR-γ), sterol regulatory element-binding protein 1 (SREBP-1c), fatty acid synthase (FAS), 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, and fatty acid-binding protein-4 (Fab-4) in HFD-induced obese rats. In conclusion, this study revealed that BG is a potential candidate to ameliorate HFD-induced obesity by modulating obesogenic marker expressions, especially by regulating the master regulator PPAR-γ.


Subject(s)
Dyslipidemias , Lipid Metabolism , Rats , Animals , Mice , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/therapeutic use , Obesity/drug therapy , Body Weight , Diet, High-Fat/adverse effects , Dyslipidemias/drug therapy , Liver/metabolism , Mice, Inbred C57BL
6.
Nutr Res ; 110: 74-86, 2023 02.
Article in English | MEDLINE | ID: mdl-36689814

ABSTRACT

Obesity is a chronic metabolic disease that involves excessive accumulation of fat in white adipose tissue (WAT). Apart from storing excess fats, WAT also serves as an important endocrine organ secreting adipocytokines such as adiponectin and leptin. Adiponectin and leptin bind to their transmembrane receptors adiponectin receptor 1 (AdipoR1)/adiponectin receptor 2 (AdipoR2) and Ob-R, respectively, and mediate their effect on metabolism by regulating multiple downstream targets. Dietary fat is considered the main culprit behind obesity development. Numerous preclinical studies have highlighted role of essential polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, in prevention of obesity. Despite emerging data, there still is no clear understanding of the mechanism of action of n-3 PUFAs and n-6 PUFAs on adipose tissue function in two functionally and anatomically different depots of WAT: visceral and subcutaneous. We designed this study using a high fat diet (HFD) fed rodent model of obesity to test our hypothesis that n-3 and n-6 PUFAs possibly differentially modulate adipokine secretion and downstream metabolic pathways such as peroxisome proliferator-activated receptor-γ (PPAR-γ), protein kinase B (AKT)-forkhead box O1 (FOXO1), and Janus kinase-signal transducer and activator of transcription in obesity. The results of the current study showed that n-3 PUFAs upregulate the expression of AdipoR1/R2 and ameliorate the effects of HFD by modulating adipogenesis via PPAR-γ and by improving glucose tolerance and lipid metabolism via AKT-FOXO1 axis in fish oil fed rats. However, n-6 PUFAs did not show any remarkable change compared with HFD fed animals. Our study highlights that n-3 PUFAs modulate expression of various targets in adiponectin and leptin signaling cascade, bringing about an overall reduction in obesity and improvement in adipose tissue function in HFD induced obesity.


Subject(s)
Diet, High-Fat , Fatty Acids, Omega-3 , Rats , Animals , Diet, High-Fat/adverse effects , Adiponectin , Leptin/metabolism , Rats, Wistar , Proto-Oncogene Proteins c-akt/metabolism , Fatty Acids, Omega-6/pharmacology , Receptors, Adiponectin/metabolism , Receptors, Adiponectin/therapeutic use , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Obesity/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Adipokines/metabolism , Fatty Acids, Unsaturated/metabolism , Signal Transduction
7.
J Adv Res ; 46: 173-188, 2023 04.
Article in English | MEDLINE | ID: mdl-35700921

ABSTRACT

INTRODUCTION: Luteolin is a plant-derived flavonoid that exhibits a broad range of pharmacological activities. Studies on luteolin have mainly focused on its use for hyperlipidaemia prevention, whereas the capacity of the flavonoid to hinder hyperglycaemia development remains underexplored. OBJECTIVES: To probe the anti-hyperglycemic mechanism of 6,8-guanidyl luteolin quinone-chromium coordination (GLQ.Cr), and to assess its regulatory effect on intestinal microbiota in type 2 diabetes mellitus (T2DM) mice. METHODS: High-sucrose/high-fat diet-induced and intraperitoneal injection of streptozotocin was used to develop a T2DM model. Glycometabolism related indicators, histopathology, and gut microbiota composition in caecum samples were evaluated, and RNA sequencing (RNA-seq) of liver samples was conducted. Faecal microbiota transplantation (FMT) was further used to verify the anti-hyperglycemic activity of intestinal microbiota. RESULTS: The administration of GLQ.Cr alleviated hyperglycaemia symptoms by improving liver and pancreatic functions and modulating gut microbe communities (Lactobacillus, Alistipes, Parabacteroides, Lachnoclostridium, and Desulfovibrio). RNA-seq analysis showed that GLQ.Cr mainly affected the peroxisome proliferative activated receptor (PPAR) signalling pathway in order to regulate abnormal glucose metabolism. FMT significantly modulated the abundance of Lactobacillus, Alloprevotella, Alistipes, Bacteroides, Ruminiclostridium, Brevundimonas and Pseudomonas in the caecum to balance blood glucose levels and counteract T2DM mice inflammation. CONCLUSION: GLQ.Cr improved the abnormal glucose metabolism in T2DM mice by regulating the PPAR signalling pathway and modulating intestinal microbial composition. FMT can improve the intestinal microecology of the recipient and in turn ameliorate the symptoms of T2DM-induced hyperglycaemia.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Hyperglycemia , Mice , Animals , Gastrointestinal Microbiome/physiology , Diabetes Mellitus, Type 2/drug therapy , Luteolin/pharmacology , Luteolin/therapeutic use , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Quinones/pharmacology , Quinones/therapeutic use , Glucose/pharmacology , Glucose/therapeutic use , Hyperglycemia/drug therapy
8.
Sci Transl Med ; 14(675): eabq0021, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36516270

ABSTRACT

Despite the success of cancer immunotherapies such as programmed cell death-1 (PD-1) and PD-1 ligand 1 (PD-L1) inhibitors, patients often develop resistance. New combination therapies with PD-1/PD-L1 inhibitors are needed to overcome this issue. Bezafibrate, a ligand of peroxisome proliferator-activated receptor-γ coactivator 1α/peroxisome proliferator-activated receptor complexes, has shown a synergistic antitumor effect with PD-1 blockade in mice that is mediated by activation of mitochondria in T cells. We have therefore now performed a phase 1 trial (UMIN000017854) of bezafibrate with nivolumab in previously treated patients with advanced non-small cell lung cancer. The primary end point was the percentage of patients who experience dose-limiting toxicity, and this combination regimen was found to be well tolerated. Preplanned comprehensive analysis of plasma metabolites and gene expression in peripheral cytotoxic T cells indicated that bezafibrate promoted T cell function through up-regulation of mitochondrial metabolism including fatty acid oxidation and may thereby have prolonged the duration of response. This combination strategy targeting T cell metabolism thus has the potential to maintain antitumor activity of immune checkpoint inhibitors and warrants further validation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor , Bezafibrate/therapeutic use , Peroxisome Proliferator-Activated Receptors/therapeutic use , Ligands , B7-H1 Antigen
9.
Iran J Allergy Asthma Immunol ; 21(4): 407-417, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36243929

ABSTRACT

Hashimoto's thyroiditis (HT) results from chemoattraction of inflammatory cells toward the thyroid gland by inducing the production of interferon-gamma (IFNγ)-induced protein 10 (IP10) by T helper (Th) 1 cells. Vitamin D may suppress the IFNγ-IP10 axis, but this new function of vitamin D has not yet been investigated in HT patients. In an intervention and control group, patients received 50000 IU cholecalciferol or placebo every week for three months, respectively. The CD4+ T cells of 40 patients were isolated, and the mRNA expression levels of vitamin D receptor (VDR), peroxisome proliferator-activated receptors (PPAR)-α, and PPAR-γ genes were determined by real-time PCR. ELISA method was used to determine serum levels of vitamin D, tumor necrosis factor-alpha (TNF-α), IFN-γ, and IP10. Vitamin D levels in the intervention group were significantly higher than in the placebo group after supplementation. PPAR-α and PPAR-γ gene expression levels did not differ significantly between the two groups. The serum levels of IP10, IFNγ, and TNF-α decreased significantly in the vitamin D group, as well as in the placebo group.  During this study, vitamin D levels significantly increased in the intervention group and inflammatory factors decreased. Based on the similar results obtained in the placebo group, further studies with larger sample sizes and longer intervention times are recommended.


Subject(s)
Hashimoto Disease , Thyroxine , Chemokine CXCL10/therapeutic use , Cholecalciferol/therapeutic use , Dietary Supplements , Double-Blind Method , Female , Hashimoto Disease/drug therapy , Humans , Interferon-gamma , Peroxisome Proliferator-Activated Receptors/therapeutic use , RNA, Messenger , Receptors, Calcitriol/genetics , Receptors, Calcitriol/therapeutic use , Thyroxine/therapeutic use , Tumor Necrosis Factor-alpha , Vitamin D/therapeutic use
10.
Medicina (Kaunas) ; 58(10)2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36295635

ABSTRACT

Guidelines have increasingly stressed the concept that adequate glycemic control is required to prevent or decrease the macro- and microvascular complications of type 2 diabetes mellitus (T2DM). PPAR-gamma agonists ("glitazones") are no longer prioritized due to their effects on heart failure. However, the association between these drugs and innovative therapies could be a valuable tool to attenuate the risk factors of the metabolic syndrome. Glitazones are used for the treatment of diabetes and associated comorbidities. There is substantial scientific evidence demonstrating the effect of glitazones at a cardiometabolic level, as well as on hematological and neurological pathologies that point to their usefulness. The use of glitazones has always been controversial both for the type of patients who must take these drugs and for the side effects associated with them. Unfortunately, the recent guidelines do not include them among the preferred drugs for the treatment of hyperglycemia and rosiglitazone is out of the market in many countries due to an adverse cardiovascular risk profile. Even though real-life studies have proven otherwise, and their pleiotropic effects have been highlighted, they have been unable to achieve primacy in the choice of antihyperglycemic drugs. It would be appropriate to demonstrate the usefulness of pioglitazone and its therapeutic benefit with further cardiovascular safety studies.


Subject(s)
Diabetes Mellitus, Type 2 , Thiazolidinediones , Humans , Thiazolidinediones/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Pioglitazone/therapeutic use , Rosiglitazone/therapeutic use , Peroxisome Proliferator-Activated Receptors/therapeutic use , Hypoglycemic Agents/adverse effects
11.
Acta Biomater ; 152: 519-531, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36055610

ABSTRACT

Modern lifestyle has led to an increase in the incidence of obesity as a public health concern; however, current anti-obesity medications often show limited efficacy with severe side effects. Therapeutic drugs that are selectively delivered to adipose tissue and accelerate energy consumption are promising strategies to overcome the limitations of existing anti-obesity treatment approaches. Herein, a drug delivery platform based on a macrophage cell membrane (Ma)-camouflaged recombinant high-density lipoprotein (rHDL) that was further decorated with a P3 peptide was fabricated to realize targeted drug delivery to adipose tissue. By co-delivering rosiglitazone (Rosi), a peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonist, and sildenafil (Sild), a phosphodiesterase type 5 (PDE5) inhibitor, a synergistic therapeutic outcome was achieved in the regulation of diet-induced obesity in a mice model. Body weight reduction and the metabolic status of obese mice were significantly improved after 28 days of treatment. More importantly, a sustainable self-reinforcement effect in multidose therapy was found after using this delivery system. The continuous treatment increased prohibitin (PHB) expression and capillary density in adipose tissue, which in turn improved the accumulation of the drugs in subsequent administration. Taken together, this constructed drug delivery system showed high effectiveness with good safety by combining two anti-obesity therapeutic agents, which exhibits promising research potential for adipose-targeted delivery. STATEMENT OF SIGNIFICANCE: Therapeutic strategies that directly target adipose tissue to increase energy consumption and regulate metabolism are promising but challenging. Herein, an adipose tissue-targeted delivery system was developed using a reconstituted high-density lipoprotein (rHDL) coated by a P3 peptide-decorated macrophage membrane. For the first time, we combined rosiglitazone (Rosi) and sildenafil (Sild) in the system and achieved synergy of adipose browning and angiogenesis for anti-obesity treatment. The therapy induced prohibitin expression and angiogenesis, which improved drug accumulation in adipose tissue in subsequent administrations. This resulted in a sustainable self-reinforcement effect with improved capacity for diet-induced obesity regulation. This study highlights the combination of adipose browning and angiogenesis in anti-obesity treatment and provides an innovative concept of enhancing adipose-targeted delivery.


Subject(s)
Nanoparticles , Peroxisome Proliferator-Activated Receptors , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/therapeutic use , Hypoglycemic Agents/therapeutic use , Lipoproteins , Lipoproteins, HDL , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Nanoparticles/therapeutic use , Obesity/drug therapy , Peroxisome Proliferator-Activated Receptors/therapeutic use , Rosiglitazone/therapeutic use , Sildenafil Citrate/therapeutic use , Treatment Outcome
12.
Neurotherapeutics ; 19(5): 1546-1565, 2022 09.
Article in English | MEDLINE | ID: mdl-35917087

ABSTRACT

Abnormal productions of amyloid beta (Aß) plaque and chronic neuroinflammation are commonly observed in the brain of patients with Alzheimer's disease, and both of which induce neuronal cell death, loss of memory, and cognitive dysfunction. However, many of the drugs targeting the production of Aß peptides have been unsuccessful in treating Alzheimer's disease. In this study, we identified synthetic novel peroxisome proliferator-activating receptor (PPAR) agonist, DTMB, which can ameliorate the chronic inflammation and Aß pathological progression of Alzheimer's disease. We discovered that DTMB attenuated the proinflammatory cytokine production of microglia by reducing the protein level of NF-κB. DTMB also improved the learning and memory defects and reduced the amount of Aß plaque in the brain of 5xFAD mice. This reduction in Aß pathology was attributed to the changes in gliosis and chronic inflammation level. Additionally, bulk RNA-sequencing showed that genes related to inflammation and cognitive function were changed in the hippocampus and cortex of DTMB-treated mice. Our findings demonstrate that DTMB has the potential to be a novel therapeutic agent for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Receptors, Artificial , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Microglia/metabolism , Amyloid beta-Peptides/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Mice, Transgenic , NF-kappa B/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Peroxisome Proliferators/therapeutic use , Receptors, Artificial/metabolism , Receptors, Artificial/therapeutic use , Disease Models, Animal , Plaque, Amyloid/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism , RNA/metabolism , RNA/pharmacology , RNA/therapeutic use
13.
Aliment Pharmacol Ther ; 56(2): 209-223, 2022 07.
Article in English | MEDLINE | ID: mdl-35661191

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors known to regulate glucose and fatty acid metabolism, inflammation, endothelial function and fibrosis. PPAR isoforms have been extensively studied in metabolic diseases, including type 2 diabetes and cardiovascular diseases. Recent data extend the key role of PPARs to liver diseases coursing with vascular dysfunction, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). AIM: This review summarises and discusses the pathobiological role of PPARs in cardiovascular diseases with a special focus on their impact and therapeutic potential in NAFLD and NASH. RESULTS AND CONCLUSIONS: PPARs may be attractive for the treatment of NASH due to their liver-specific effects but also because of their efficacy in improving cardiovascular outcomes, which may later impact liver disease. Assessment of cardiovascular disease in the context of NASH trials is, therefore, of the utmost importance, both from a safety and efficacy perspective.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Liver , Non-alcoholic Fatty Liver Disease/drug therapy , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use
14.
Clin Mol Hepatol ; 28(4): 725-738, 2022 10.
Article in English | MEDLINE | ID: mdl-35286799

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease with a global prevalence of about 55% in people with type 2 diabetes mellitus (T2DM). T2DM, obesity and NAFLD are three closely inter-related pathological conditions. In addition, T2DM is one of the strongest clinical risk factors for the faster progression of NAFLD to non-alcoholic steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma. Increasing evidence suggests that newer classes of glucose-lowering drugs, such as peroxisome proliferator-activated receptor agonists, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors or sodium-glucose cotransporter-2 inhibitors, could reduce the rates of NAFLD progression. This narrative review aims to briefly summarize the recent results from randomized controlled trials testing the efficacy and safety of old and new glucose-lowering drugs for the treatment of NAFLD or NASH in adults both with and without coexisting T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Sodium-Glucose Transporter 2 Inhibitors , Humans , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/epidemiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Glucose/therapeutic use , Peroxisome Proliferator-Activated Receptors/therapeutic use , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/therapeutic use , Sodium
15.
Sci Bull (Beijing) ; 66(15): 1571-1580, 2021 08 15.
Article in English | MEDLINE | ID: mdl-36654286

ABSTRACT

Chiglitazar (Carfloglitazar) is a novel non-thiazolidinedione (TZD) structured peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes in previous clinical studies. This randomized phase 3 trial aimed to compare the efficacy and safety of chiglitazar with placebo in patients with type 2 diabetes with insufficient glycemic control by strict diet and exercise alone. Eligible patients were randomly assigned to receive chiglitazar 32 mg (n = 167), chiglitazar 48 mg (n = 166), or placebo (n = 202) once daily. The primary endpoint was the change in glycosylated hemoglobin A1c (HbA1c) at week 24 with superiority of chiglitazar over placebo. The results showed that both chiglitazar 32 and 48 mg resulted in significant and clinically meaningful reductions in HbA1c, and placebo-adjusted estimated treatment differences at week 24 for chiglitazar 32 and 48 mg were -0.87% (95% confidential interval (CI): -1.10 to -0.65; P < 0.0001) and -1.05% (95% CI: -1.29 to -0.81; P < 0.0001), respectively. Secondary efficacy parameters including glycemic control, insulin sensitivity and triglyceride reduction were also significantly improved in the chiglitazar groups. The overall frequency of adverse events and study discontinuation attributable to adverse events were similar among the groups. Low incidences of mild edema and body weight gain were reported in the chiglitazar dose groups. The results from this phase 3 trial demonstrated that the PPAR pan-agonist chiglitazar possesses an overall good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/drug therapy , Peroxisome Proliferator-Activated Receptors/therapeutic use , Hypoglycemic Agents/adverse effects , Carbazoles
16.
Crit Rev Ther Drug Carrier Syst ; 35(1): 65-97, 2018.
Article in English | MEDLINE | ID: mdl-29611471

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear transcription factors. They exist in three isoforms (PPAR-α, PPAR-ß/δ, and PPAR-Υ) in humans, but mainly PPAR-Υ, and they are expressed in retinal epithelial pigment. PPARs are involved in mediating numerous pathological implications in eye such as diabetic retinopathy (DR), choroidal neovascularization (CNV), glaucoma, diabetic macular edema, and other retinal diseases. Peroxisome proliferator-activated receptors are key players in various biological pathways like lipid degeneration, immune regulation, and reactive oxygen species regulation, regulation of vascular endothelial growth factor, matrixmetalloproteinase-9, and docosahexaenoic acid pathway. Based on evidence from clinical investigations, the drugs meant for PPARs could be promising candidates for intraocular therapy. Anti-VEGF therapy, including bevacizumab, ranibizumab, and aptamers (pegaptanib), has been approved for wet age-related macular degeneration (ARMD). Recently, researchers have explored the role of PPAR-γ in ocular pathophysiological processes and PPAR-γ agonists as novel adjuvants in the treatment of eye diseases. PPAR-γ exhibits potential benefits to improve or prevent various vision-threatening eye diseases such as age-related macular degeneration (ARMD), diabetic retinopathy (DR), keratitis, and optic neuropathy. However, PPAR-γ presents challenges and offers opportunities for ocular scientists to bring better outcomes.


Subject(s)
Eye Diseases/drug therapy , Eye Diseases/physiopathology , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Apoptosis/physiology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/physiopathology , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/physiopathology , Drug Delivery Systems/methods , Glycation End Products, Advanced/metabolism , Humans , Keratitis/drug therapy , Keratitis/physiopathology , Optic Nerve Diseases/drug therapy , PPAR gamma/agonists , Protein Kinase C/antagonists & inhibitors , Vascular Endothelial Growth Factors/antagonists & inhibitors , Wet Macular Degeneration/drug therapy , Wet Macular Degeneration/physiopathology
17.
Am J Med ; 130(9): 1068-1075, 2017 09.
Article in English | MEDLINE | ID: mdl-28344139

ABSTRACT

BACKGROUND: Worsening renal function during hospitalization for an acute coronary syndrome is strongly predictive of in-hospital and long-term outcome. However, the role of post-discharge worsening renal function has never been investigated in this setting. METHODS: We considered the placebo cohort of the AleCardio trial comparing aleglitazar with standard medical therapy among patients with type 2 diabetes mellitus and a recent acute coronary syndrome. Patients who had died or had been admitted to hospital for heart failure before the 6-month follow-up, as well as patients without complete renal function data, were excluded, leaving 2776 patients for the analysis. Worsening renal function was defined as a >20% reduction in estimated glomerular filtration rate from discharge to 6 months, or progression to macroalbuminuria. The Cox regression analysis was used to determine the prognostic impact of 6-month renal deterioration on the composite of all-cause death and hospitalization for heart failure. RESULTS: Worsening renal function occurred in 204 patients (7.34%). At a median follow-up of 2 years the estimated rates of death and hospitalization for heart failure per 100 person-years were 3.45 (95% confidence interval [CI], 2.46-6.36) for those with worsening renal function, versus 1.43 (95% CI, 1.14-1.79) for patients with stable renal function. At the adjusted analysis worsening renal function was associated with the composite endpoint (hazard ratio 2.65; 95% CI, 1.57-4.49; P <.001). CONCLUSIONS: Post-discharge worsening renal function is not infrequent among patients with type 2 diabetes and acute coronary syndromes with normal or mildly depressed renal function, and is a strong predictor of adverse cardiovascular events.


Subject(s)
Acute Coronary Syndrome/mortality , Diabetes Complications/mortality , Heart Failure/mortality , Oxazoles/therapeutic use , Renal Insufficiency, Chronic/mortality , Thiophenes/therapeutic use , Acute Coronary Syndrome/epidemiology , Acute Coronary Syndrome/physiopathology , Acute Coronary Syndrome/therapy , Aged , Albuminuria , Analysis of Variance , Biomarkers , Cause of Death , Comorbidity , Creatinine/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Disease Progression , Female , Follow-Up Studies , Glomerular Filtration Rate , Humans , Male , Oxazoles/administration & dosage , Oxazoles/adverse effects , Patient Discharge/statistics & numerical data , Percutaneous Coronary Intervention , Peroxisome Proliferator-Activated Receptors/administration & dosage , Peroxisome Proliferator-Activated Receptors/adverse effects , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/therapeutic use , Predictive Value of Tests , Proportional Hazards Models , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/etiology , Risk Factors , Thiophenes/administration & dosage , Thiophenes/adverse effects
18.
Nat Rev Endocrinol ; 13(1): 36-49, 2017 01.
Article in English | MEDLINE | ID: mdl-27636730

ABSTRACT

Obesity is a worldwide epidemic that predisposes individuals to cardiometabolic complications, such as type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD), which are all related to inappropriate ectopic lipid deposition. Identification of the pathogenic molecular mechanisms and effective therapeutic approaches are highly needed. The peroxisome proliferator-activated receptors (PPARs) modulate several biological processes that are perturbed in obesity, including inflammation, lipid and glucose metabolism and overall energy homeostasis. Here, we review how PPARs regulate the functions of adipose tissues, such as adipogenesis, lipid storage and adaptive thermogenesis, under healthy and pathological conditions. We also discuss the clinical use and mechanism of PPAR agonists in the treatment of obesity comorbidities such as dyslipidaemia, T2DM and NAFLD. First generation PPAR agonists, primarily those acting on PPARγ, are associated with adverse effects that outweigh their clinical benefits, which led to the discontinuation of their development. An improved understanding of the physiological roles of PPARs might, therefore, enable the development of safe, new PPAR agonists with improved therapeutic potential.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/therapy , Dyslipidemias/pathology , Dyslipidemias/therapy , Humans , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/therapy , Peroxisome Proliferator-Activated Receptors/therapeutic use
19.
Clín. investig. arterioscler. (Ed. impr.) ; 25(2): 63-73, abr.-jun. 2013. ilus
Article in Spanish | IBECS | ID: ibc-114543

ABSTRACT

Introducción El consumo excesivo de alimentos hipercalóricos y de alto contenido en grasas saturadas produce una dislipidemia aterogénica. En este estudio hemos analizado los efectos del activador de PPARβ/δ GW501516 sobre la hipertrigliceridemia inducida por una dieta rica en grasas. Metodología Ratones macho fueron distribuidos aleatoriamente en 3 grupos: control (dieta estándar), dieta grasa (high fat diet [HFD], 35% grasa en peso, 58% kcal procedentes de grasa) y dieta grasa más GW501516 (3 mg/kg/día). La duración del tratamiento fue de 3 semanas. Resultados La HFD causó hipertrigliceridemia acompañada de una reducción de los niveles hepáticos de la proteína AMPK fosforilada y de los niveles de ARNm Pgc-1α y lipina 1. Estos efectos fueron revertidos por el tratamiento con GW501516. El mantenimiento de la AMPK fosforilada tras el tratamiento con GW501516 podría deberse al aumento de la relación AMP/ATP. GW501516 incrementó los niveles de proteína lipina 1 nuclear acompañado por una amplificación de la vía PGC-1α-PPARα y un aumento de la actividad de unión al ADN de PPARα, así como el incremento en la expresión de los genes diana de PPARα implicados en la β-oxidación de ácidos grasos. GW501516 también aumentó los niveles plasmáticos de β-hidroxibutirato, producto final de la β-oxidación hepática. Finalmente, GW501516 incrementó los niveles del ligando endógeno de PPARα, 16:0/18:1-fosfatidilcolina, y aumentó la expresión del receptor de las VLDL en hígado. Conclusión El efecto hipotrigliceridemiante de GW501516 en ratones sometidos a HFD se acompaña de un aumento de los niveles de la AMPK fosforilada y de un aumento de la vía PGC-1α-lipina 1-PPARα (AU)


Introduction Excessive consume of hypercaloric and high in saturated fat food causes an atherogenic dyslipidemia. In this study we analyzed the effects of PPARβ/δ activator GW501516 on the hypertriglyceridemia induced by a high-fat diet. Methods Male mice were randomized in three groups: control (standard chow), high fat diet (HFD, 35% fat by weight, 58% Kcal from fat) and high fat diet plus GW501516 (3 mg/Kg/day). Treatment duration was three weeks. Results HFD-induced hypertriglyceridemia was accompanied by a reduction in hepatic levels of phospho-AMPK and in PGC-1α and Lipin 1 mRNA levels. All these effects were reversed by GW501516 treatment. The lack of changes in phospho-AMPK levels after GW501516 treatment in HFD-fed animals could be the result of an increase in the AMP/ATP ratio. GW501516 treatment also increased Lipin 1 protein levels in the nucleus, led to the amplification of the PGC-1α-PPARα pathway and increased PPARα DNA-binding activity, as well as the expression of PPARα-target genes involved in fatty acid oxidation. GW501516 also increased β-hydroxibutirate plasmatic levels, a hepatic β-oxidation end product. Finally, GW501516 increased the hepatic levels of the PPARα endogenous ligand 16:0/18:1-PC and the expression of the VLDL receptor. Conclusion These data indicate that the hypotriglyceridemic effect of GW501516 in mice subjected to HFD-fed mice is accompanied by an increase in phospho-AMPK levels and the amplification of the PGC-1α-Lipin 1-PPARα pathway (AU)


Subject(s)
Animals , Mice , PPAR-beta/agonists , Hypertriglyceridemia/prevention & control , Dyslipidemias/drug therapy , Dietary Fats/adverse effects , Peroxisome Proliferator-Activated Receptors/therapeutic use , Adenosine Monophosphate/therapeutic use
20.
Curr Opin Crit Care ; 18(6): 651-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104069

ABSTRACT

PURPOSE OF REVIEW: Spinal cord injury is a devastating acute neurological condition with loss of function and poor long-term prognosis. This review summarizes current management strategies and innovative concepts on the horizon. RECENT FINDINGS: The routine use of steroids in patients with spinal cord injuries has been largely abandoned and considered a 'harmful standard of care'. Prospective trials have shown that early spine stabilization within 24  h results in decreased secondary complication rates. Neuronal plasticity and axonal regeneration in the adult spinal cord are limited due to myelin-associated inhibitory molecules, such as Nogo-A. The experimental inhibition of Nogo-A ameliorates axonal sprouting and functional recovery in animal models. SUMMARY: General management strategies for acute spinal cord injury consist of protection of airway, breathing, oxygenation and control of blood loss with maintenance of blood pressure. Unstable spine fractures should be stabilized early to allow unrestricted mobilization of patients with spinal cord injuries and to decrease preventable complications. Steroids are largely considered obsolete and have been abandoned in clinical guidelines. Nogo-A represents a promising new pharmacological target to promote sprouting of injured axons and restore function. Prospective clinical trials of Nogo-A inhibition in patients with spinal cord injuries are currently under way.


Subject(s)
Spinal Cord Injuries/drug therapy , Central Nervous System/immunology , Combined Modality Therapy/methods , Female , Growth Inhibitors/therapeutic use , Humans , Hypothermia, Induced , Immobilization , Male , Myelin Proteins/therapeutic use , Nogo Proteins , Peroxisome Proliferator-Activated Receptors/therapeutic use , Spinal Cord Injuries/classification , Spinal Cord Injuries/complications , Spinal Cord Injuries/physiopathology , Steroids/therapeutic use , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...