Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Am J Physiol Endocrinol Metab ; 316(5): E908-E921, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30807216

ABSTRACT

The prevalence of cardiometabolic syndrome (CMS) is increased in women after menopause. While hormone replacement therapy has been prescribed to relieve several components of CMS in postmenopausal women, some aspects of cardiometabolic dysfunction cannot be completely restored. The present study examined the effectiveness of estrogen replacement alone and in combination with exercise by voluntary wheel running (VWR) for alleviating the risks of CMS, insulin-mediated skeletal muscle glucose transport, and hepatic fat accumulation in ovariectomized Sprague-Dawley rats fed a high-fat high-fructose diet (OHFFD). We compared a sham-operated group with OHFFD rats that were subdivided into a sedentary, estradiol replacement (E2), and E2 plus VWR for 12 wk. E2 prevented the development of insulin resistance in skeletal muscle glucose transport and decreased hepatic fat accumulation in OHFFD rats. Furthermore, E2 treatment decreased visceral fat mass and low-density lipoprotein (LDL)-cholesterol in OHFFD rats, while VWR further decreased LDL-cholesterol and increased the ratio of high-density lipoprotein-cholesterol to total cholesterol to a greater extent. Although E2 treatment alone did not reduce serum triglyceride levels in OHFFD rats, the combined intervention of E2 and VWR lowered serum triglycerides in E2-treated OHFFD rats. The addition of VWR to E2-treated OHFFD rats led to AMPK activation and upregulation of peroxisome proliferator-activated receptor-γ (PPARγ) coactivator-1α and PPARδ in skeletal muscle along with increased fatty acid oxidation and suppressed fatty acid synthesis in the liver. Collectively, our findings indicate that, to achieve greater health benefits, physical exercise is required for E2-treated individuals under ovarian hormone deprivation with high-energy consumption.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Fatty Liver/metabolism , Liver/drug effects , Metabolic Syndrome/metabolism , Motor Activity , Muscle, Skeletal/drug effects , Animals , Cholesterol, HDL/drug effects , Cholesterol, HDL/metabolism , Cholesterol, LDL/drug effects , Cholesterol, LDL/metabolism , Diet, High-Fat , Dietary Sugars , Estrogen Replacement Therapy , Female , Fructose , Glucose/metabolism , Insulin Resistance , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Menopause , Muscle, Skeletal/metabolism , Ovariectomy , PPAR delta/drug effects , PPAR delta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Rats, Sprague-Dawley
2.
Gerontology ; 65(4): 397-406, 2019.
Article in English | MEDLINE | ID: mdl-31096217

ABSTRACT

BACKGROUND: Androgen production following exercise has been suggested to contribute anabolic actions of muscle. However, the underlying mechanisms of the androgen receptor (AR) in androgen's action are still unclear. OBJECTIVE: In the present study, we examined androgen/AR-mediated action in exercise, especially for the suppression of myostatin, a potent negative regulator of muscle mass. METHODS: To examine the effects of exercise, we employed low-intensity exercise in mice and electric pulse stimulation (EPS) in C2C12 myotubes. Androgen production by C2C12 myotubes was measured by enzyme-linked immunosorbent assay. To block the action of AR, we pretreated C2C12 myotubes with flutamide. Quantitative real-time polymerase chain reaction was used to determine the expression levels of proteolytic genes including CCAAT/enhancer-binding protein delta (C/EBPδ), myostatin and muscle E3 ubiquitin ligases, as well as myogenic genes such as myogenin and PGC1α. The activation of 5'-adenosine-activated protein kinase and STAT3 was determined by Western blot analysis. RESULTS: Both mRNA and protein levels of AR significantly increased in skeletal muscle of low-intensity exercised mice and C2C12 myotubes exposed to EPS. Production of testosterone and dihydrotestosterone from EPS-treated C2C12 myotubes was markedly increased. Of interest, we found that myostatin was clearly inhibited by EPS, and its inhibition was significantly abrogated when AR was blocked by flutamide. To test how AR suppresses myostatin, we examined the effects of EPS on C/EBPδ because the promoter region of myostatin has several C/EBP recognition sites. C/EBPδ expression was decreased by EPS, and this decrease was negated by flutamide. IL-6 and phospho-STAT3 (pSTAT3) expression, the downstream pathway of myostatin, were decreased by EPS and this was also reversed by flutamide. Similar downregulation of C/EBPδ, myostatin, and IL-6 was seen in skeletal muscle of low-intensity exercised mice. CONCLUSIONS: Muscle AR expression and androgen production were increased by exercise and EPS treatment. As a mechanistical insight, it is suggested that AR inhibited myostatin expression transcriptionally by C/EBPδ suppression, which negatively influences IL-6/pSTAT3 expression and consequently contributes to the prevention of muscle proteolysis during exercise.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/genetics , Muscle Fibers, Skeletal/metabolism , Myostatin/genetics , Physical Conditioning, Animal , Receptors, Androgen/genetics , Androgen Antagonists/pharmacology , Animals , CCAAT-Enhancer-Binding Protein-delta/drug effects , Dihydrotestosterone/metabolism , Electric Stimulation , Flutamide/pharmacology , In Vitro Techniques , Interleukin-6/metabolism , Mice , Muscle Fibers, Skeletal/drug effects , Myogenin/drug effects , Myogenin/genetics , Myostatin/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction , Testosterone/metabolism , Transcriptome
3.
Med Sci Monit ; 25: 1220-1231, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30765684

ABSTRACT

BACKGROUND PGC-1α can be activated by deacetylation reactions catalyzed by SIRT1. Resveratrol is currently known as a potent activator of SIRT1. However, it is unknown whether the renal-protective effect of resveratrol is further related to activation of the podocyte SIRT1/PGC-1α pathway. MATERIAL AND METHODS High glucose was used to stimulate mouse podocytes. Resveratrol and PGC-1α siRNA transfection were used to perform co-intervention treatments. The protein and mRNA expression levels of SIRT1, PGC-1α, NRF1, and TFAM were detect by immunofluorescence, Western blot analysis, and qRT-PCR in the podocytes, respectively. DCHF-DA and MitoSOX™ staining were used to monitor the total ROS and mitochondrial ROS levels, respectively. The specific activities of complexes I and III were measured using Complex I and III Assay Kits. Mitochondrial membrane potential and cell apoptosis were measured using JC-1 staining and Annexin V-FITC/PI double-staining, respectively. RESULTS We found that high-glucose stimulation results in time-dependent decreases in the expression of SIRT1, PGC-1α, and its downstream genes NRF1 and mitochondrial transcription factor A (TFAM) for mouse podocytes, and increases ROS levels in cells and mitochondria. Moreover, the expression of nephrin was downregulated and the cell apoptotic rate was increased. Resveratrol treatment can improve abnormalities caused by high-glucose stimulation. In addition, it can also reduce the release of mitochondrial cytochrome C and DIABLO proteins to the cytoplasm and increase respiratory chain complex I and III activity and mitochondrial membrane potential. CONCLUSIONS Resveratrol can reduce the oxidative damage and apoptosis of podocytes induced by high-glucose stimulation via SIRT1/PGC-1α-mediated mitochondrial protection.


Subject(s)
Oxidative Stress/drug effects , Podocytes/drug effects , Resveratrol/pharmacology , Animals , Apoptosis/drug effects , Cell Culture Techniques , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Membrane Potential, Mitochondrial , Mice , Mitochondria , Mitochondrial Proteins , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Protein Processing, Post-Translational , RNA, Small Interfering , Signal Transduction/drug effects , Sirtuin 1/drug effects , Transcription Factors
4.
PLoS Pathog ; 12(10): e1005954, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27764247

ABSTRACT

Chronic chagasic cardiomyopathy (CCM) is presented by increased oxidative/inflammatory stress and decreased mitochondrial bioenergetics. SIRT1 senses the redox changes and integrates mitochondrial metabolism and inflammation; and SIRT1 deficiency may be a major determinant in CCM. To test this, C57BL/6 mice were infected with Trypanosoma cruzi (Tc), treated with SIRT1 agonists (resveratrol or SRT1720), and monitored during chronic phase (~150 days post-infection). Resveratrol treatment was partially beneficial in controlling the pathologic processes in Chagas disease. The 3-weeks SRT1720 therapy provided significant benefits in restoring the left ventricular (LV) function (stroke volume, cardiac output, ejection fraction etc.) in chagasic mice, though cardiac hypertrophy presented by increased thickness of the interventricular septum and LV posterior wall, increased LV mass, and disproportionate synthesis of collagens was not controlled. SRT1720 treatment preserved the myocardial SIRT1 activity and PGC1α deacetylation (active-form) that were decreased by 53% and 9-fold respectively, in chagasic mice. Yet, SIRT1/PGC1α-dependent mitochondrial biogenesis (i.e., mitochondrial DNA content, and expression of subunits of the respiratory complexes and mtDNA replication machinery) was not improved in chronically-infected/SRT1720-treated mice. Instead, SRT1720 therapy resulted in 2-10-fold inhibition of Tc-induced oxidative (H2O2 and advanced oxidation protein products), nitrosative (inducible nitric oxide synthase, 4-hydroxynonenal, 3-nitrotyrosine), and inflammatory (IFNγ, IL1ß, IL6 and TNFα) stress and inflammatory infiltrate in chagasic myocardium. These benefits were delivered through SIRT1-dependent inhibition of NFκB transcriptional activity. We conclude that Tc inhibition of SIRT1/PGC1α activity was not a key mechanism in mitochondrial biogenesis defects during Chagas disease. SRT1720-dependent SIRT1 activation led to suppression of NFκB transcriptional activity, and subsequently, oxidative/nitrosative and inflammatory pathology were subdued, and antioxidant status and LV function were enhanced in chronic chagasic cardiomyopathy.


Subject(s)
Chagas Cardiomyopathy/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Oxidative Stress/drug effects , Sirtuin 1/metabolism , Stilbenes/pharmacology , Animals , Antioxidants/pharmacology , Blotting, Western , Disease Models, Animal , Heart/drug effects , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Resveratrol , Signal Transduction/physiology , Sirtuin 1/drug effects , Transcriptome , Trypanosoma cruzi
5.
Cell Mol Neurobiol ; 38(4): 929-939, 2018 May.
Article in English | MEDLINE | ID: mdl-29159732

ABSTRACT

Oxidative stress is a great challenge to neurons following cerebral ischemia. PGC-1α has been shown to act as a potent modulator of oxidative metabolism. In this study, the effects of ZLN005, a small molecule that activate PGC-1α, against oxygen-glucose deprivation (OGD)- or ischemia-induced neuronal injury in vitro and in vivo were investigated. Transient middle cerebral artery occlusion (tMCAO) was performed in rats and ZLN005 was administered intravenously at 2 h, 4 h, or 6 h after ischemia onset. Infarct volume and neurological deficit score were detected to evaluate the neuroprotective effects of ZLN005. Well-differentiated PC12 cells, which were subjected to OGD for 2 h followed by reoxygenation for 22 h, were used as an in vitro ischemic model. Changes in expression of PGC-1α, its related genes, and antioxidant genes were determined by real-time quantitative PCR. The results showed that ZLN005 reduced cerebral infarct volume and improved the neurological deficit in rat with tMCAO, and significantly protected OGD-induced neuronal injury in PC12 cells. Furthermore, ZLN005 enhanced expression of PGC-1α in PC12 cells and in the ipsilateral hemisphere of rats with tMCAO. Additionally, ZLN005 increased antioxidant genes, including SOD1 and HO-1, and significantly prevented the ischemia-induced decrease in SOD activity. Taking together, the PGC-1α activator ZLN005 exhibits neuroprotective effects under ischemic conditions and molecular mechanisms possibly involve activation of PGC-1α signaling pathway and cellular antioxidant systems.


Subject(s)
Benzimidazoles/pharmacology , Brain Ischemia/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Animals , Antioxidants/pharmacology , Brain Ischemia/metabolism , Cell Survival/drug effects , Ischemia/drug therapy , Ischemia/metabolism , Male , Neurons/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
Am J Physiol Heart Circ Physiol ; 312(1): H128-H140, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27836895

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is associated with progressive cardiac pathology; however, the SIRT1/PGC1-α activator quercetin may cardioprotect dystrophic hearts. We tested the extent to which long-term 0.2% dietary quercetin enrichment attenuates dystrophic cardiopathology in Mdx/Utrn+/- mice. At 2 mo, Mdx/Utrn+/- mice were fed quercetin-enriched (Mdx/Utrn+/--Q) or control diet (Mdx/Utrn+/-) for 8 mo. Control C57BL/10 (C57) animals were fed a control diet for 10 mo. Cardiac function was quantified by MRI at 2 and 10 mo. Spontaneous physical activity was quantified during the last week of treatment. At 10 mo hearts were excised for histological and biochemical analysis. Quercetin feeding improved various physiological indexes of cardiac function in diseased animals. Mdx/Utrn+/--Q also engaged in more high-intensity physical activity than controls. Histological analyses of heart tissues revealed higher expression and colocalization of utrophin and α-sarcoglycan. Lower abundance of fibronectin, cardiac damage (Hematoxylin Eosin-Y), and MMP9 were observed in quercetin-fed vs. control Mdx/Utrn+/- mice. Quercetin evoked higher protein abundance of PGC-1α, cytochrome c, ETC complexes I-V, citrate synthase, SOD2, and GPX compared with control-fed Mdx/Utrn+/- Quercetin decreased abundance of inflammatory markers including NFκB, TGF-ß1, and F4/80 compared with Mdx/Utrn+/-; however, P-NFκB, P-IKBα, IKBα, CD64, and COX2 were similar between groups. Dietary quercetin enrichment improves cardiac function in aged Mdx/Utrn+/- mice and increases mitochondrial protein content and dystrophin glycoprotein complex formation. Histological analyses indicate a marked attenuation in pathological cardiac remodeling and indicate that long-term quercetin consumption benefits the dystrophic heart. NEW & NOTEWORTHY: The current investigation provides first-time evidence that quercetin provides physiological cardioprotection against dystrophic pathology and is associated with improved spontaneous physical activity. Secondary findings suggest that quercetin-dependent outcomes are in part due to PGC-1α pathway activation.


Subject(s)
Antioxidants/pharmacology , Heart/drug effects , Muscular Dystrophy, Animal/physiopathology , Quercetin/pharmacology , Animals , Antigens, Differentiation/drug effects , Antigens, Differentiation/metabolism , Blotting, Western , Citrate (si)-Synthase/drug effects , Citrate (si)-Synthase/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Cytochromes c/drug effects , Cytochromes c/metabolism , Disease Models, Animal , Electron Transport Chain Complex Proteins/drug effects , Electron Transport Chain Complex Proteins/metabolism , Fibronectins/metabolism , Food, Fortified , Heart/diagnostic imaging , Heart/physiopathology , Magnetic Resonance Imaging , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred mdx , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Motor Activity , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne , Myocardium/metabolism , Myocardium/pathology , NF-KappaB Inhibitor alpha/drug effects , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation , Receptors, IgG/drug effects , Receptors, IgG/metabolism , Sarcoglycans/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Utrophin/genetics , Utrophin/metabolism
7.
J Cell Biochem ; 117(1): 239-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26105159

ABSTRACT

The activation of AMP-activated protein kinase (AMPK) is known to repress the expression of the insulin gene and glucose-stimulated insulin secretion (GSIS). However, the mechanisms by which this occurs, as well as the effects of AMPK activation on glucolipotoxicity-induced ß-cell dysfunction, have not been elucidated. To investigate the effects of 5-amino-4-imidazolecarboxamide ribonucleotide (AICAR) and peroxisome proliferator-activated receptorγ-coactivator-1α (PGC-1α) on ß-cell-specific genes under glucolipotoxic conditions, we performed real-time PCR and measured insulin secretion by primary islets. To study these effects in vivo, we administered AICAR for 10 days (1 mg/g body weight) to 90% pancreatectomized hyperglycemic mice. The exposure of isolated rat and human islets to glucolipotoxic conditions and the overexpression of PGC-1α suppressed insulin and NEUROD1 mRNA expression. However, the expression of these genes was preserved by AICAR treatment and by PGC-1α inhibition. Exposure of isolated islets to glucolipotoxic conditions for 3 days decreased GSIS, which was also well maintained by AICAR treatment and by PGC-1α inhibition. The administration of AICAR to 90% pancreatectomized hyperglycemic mice improved glucose tolerance and insulin secretion. These results indicate that treatment of islets with an AMPK agonist under glucolipotoxic conditions protects against glucolipotoxicity-induced ß-cell dysfunction. A better understanding of the functions of molecules such as PGC-1α and AMPK, which play key roles in intracellular fuel regulation, could herald a new era for the treatment of patients with type 2 diabetes mellitus by providing protection against glucolipotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/pharmacology , Insulin/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology
8.
Am J Physiol Endocrinol Metab ; 311(1): E246-51, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27245337

ABSTRACT

The transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α is recognized as the master regulator of mitochondrial biogenesis. However, recently a novel isoform, PGC-1α4, that specifically regulates muscle hypertrophy was discovered. Because stimulation of mechanistic target of rapamycin complex 1 (mTORC1) activity is tightly coupled to hypertrophy, we hypothesized that activation of this pathway would upregulate PGC-1α4. Eight male subjects performed heavy resistance exercise (10 × 8-12 repetitions at ∼75% of 1 repetition maximum in leg press) on four different occasions, ingesting in random order a solution containing essential amino acids (EAA), branched-chain amino acids (BCAA), leucine, or flavored water (placebo) during and after the exercise. Biopsies were taken from the vastus lateralis muscle before and immediately after exercise, as well as following 90 and 180 min of recovery. Signaling through mTORC1, as reflected in p70S6 kinase phosphorylation, was stimulated to a greater extent by the EAA and BCAA than the leucine or placebo supplements. Unexpectedly, intake of EAA or BCAA attenuated the stimulatory effect of exercise on PGC-1α4 expression by ∼50% (from a 10- to 5-fold increase with BCAA and EAA, P < 0.05) 3 h after exercise, whereas intake of leucine alone did not reduce this response. The 60% increase (P < 0.05) in the level of PGC-1α1 mRNA 90 min after exercise was uninfluenced by amino acid intake. Muscle glycogen levels were reduced and AMP-activated protein kinase α2 activity and phosphorylation of p38 mitogen-activated protein kinase enhanced to the same extent with all four supplements. In conclusion, induction of PGC-1α4 does not appear to regulate the nutritional (BCAA or EAA)-mediated activation of mTORC1 in human muscle.


Subject(s)
Amino Acids, Branched-Chain/pharmacology , Amino Acids, Essential/pharmacology , Leucine/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Quadriceps Muscle/metabolism , RNA, Messenger/drug effects , Resistance Training , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Adult , Exercise , Glycogen/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phosphorylation , Protein Isoforms/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Am J Physiol Endocrinol Metab ; 311(5): E836-E849, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27677502

ABSTRACT

Alcohol ingestion decreases postexercise rates of muscle protein synthesis, but the mechanism(s) (e.g., increased protein breakdown) underlying this observation is unknown. Autophagy is an intracellular "recycling" system required for homeostatic substrate and organelle turnover; its dysregulation may provoke apoptosis and lead to muscle atrophy. We investigated the acute effects of alcohol ingestion on autophagic cell signaling responses to a bout of concurrent (combined resistance- and endurance-based) exercise. In a randomized crossover design, eight physically active males completed three experimental trials of concurrent exercise with either postexercise ingestion of alcohol and carbohydrate (12 ± 2 standard drinks; ALC-CHO), energy-matched alcohol and protein (ALC-PRO), or protein (PRO) only. Muscle biopsies were taken at rest and 2 and 8 h postexercise. Select autophagy-related gene (Atg) proteins decreased compared with rest with ALC-CHO (P < 0.05) but not ALC-PRO. There were parallel increases (P < 0.05) in p62 and PINK1 commensurate with a reduction in BNIP3 content, indicating a diminished capacity for mitochondria-specific autophagy (mitophagy) when alcohol and carbohydrate were coingested. DNA fragmentation increased in both alcohol conditions (P < 0.05); however, nuclear AIF accumulation preceded this apoptotic response with ALC-CHO only (P < 0.05). In contrast, increases in the nuclear content of p53, TFEB, and PGC-1α in ALC-PRO were accompanied by markers of mitochondrial biogenesis at the transcriptional (Tfam, SCO2, and NRF-1) and translational (COX-IV, ATPAF1, and VDAC1) level (P < 0.05). We conclude that alcohol ingestion following exercise triggers apoptosis, whereas the anabolic properties of protein coingestion may stimulate mitochondrial biogenesis to protect cellular homeostasis.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Central Nervous System Depressants/pharmacology , Dietary Carbohydrates/pharmacology , Dietary Proteins/pharmacology , Ethanol/pharmacology , Exercise/physiology , Muscle Fibers, Skeletal/drug effects , Adolescent , Adult , Alcohol Drinking , Apoptosis/physiology , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cross-Over Studies , DNA Fragmentation/drug effects , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/metabolism , Healthy Volunteers , Humans , Male , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/drug effects , Mitochondrial Proton-Translocating ATPases/metabolism , Mitophagy/drug effects , Mitophagy/physiology , Molecular Chaperones/drug effects , Molecular Chaperones/metabolism , Muscle Fibers, Skeletal/physiology , Nuclear Respiratory Factor 1/drug effects , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Kinases/drug effects , Protein Kinases/metabolism , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/drug effects , Transcription Factors/metabolism , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Voltage-Dependent Anion Channel 1/drug effects , Voltage-Dependent Anion Channel 1/metabolism , Young Adult
10.
Chem Biol Interact ; 388: 110850, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38135199

ABSTRACT

Acute kidney injury (AKI) in sepsis is a vital and dangerous organ failure caused by an infection-induced dysregulation of the host reaction. Malvidin possesses significant anti-inflammatory and antioxidant bioactivities. This study explored the critical roles of malvidin in sepsis AKI and the crosstalk among mitochondrial function, nucleotide-binding oligomerization-like receptor 3 (NLRP3) inflammasome and nuclear factor erythroid 2 (Nrf2) signaling pathway. First, C57BL/6 mice were administered lipopolysaccharide intraperitoneally for 6 h to create an AKI model of sepsis. Hematoxylin-eosin staining and serum biomarker assays showed that malvidin protected from AKI in sepsis. Real-time fluorescence quantitative polymerase chain reaction analysis revealed that malvidin was able to inhibit inflammatory cytokines and mediators. Western blot assays indicated that malvidin suppressed NLRP3 inflammasome activation and enhanced antioxidant properties. Additionally, human renal tubular epithelial cells were stimulated by lipopolysaccharide/adenosine triphosphate to establish an NLRP3 inflammasome activation model in vitro, and in line with findings in vivo, malvidin significantly inhibited NLRP3 inflammasome activation. Furthermore, our data indicate that malvidin restored mitochondrial quality and function, reduced reactive oxygen species production, increased mitochondrial membrane potential, enhanced mitochondrial DNA copy number, and promoted peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) nuclear translocation. Moreover, inhibitor blockade assays indicated that both PGC-1α and Nrf2 affected the inhibition of the NLRP3 inflammasome by malvidin. Finally, immunoprecipitation assays showed that malvidin promoted PGC-1α and Nrf2 interactions. Overall, malvidin alleviated lipopolysaccharide-induced sepsis AKI, improved mitochondrial function and mitochondrial biogenesis, and inhibited the NLRP3 inflammasome through the PGC-1α/Nrf2 signaling pathway, suggesting that malvidin might translate into clinical applications for sepsis AKI therapy.


Subject(s)
Acute Kidney Injury , Anthocyanins , Sepsis , Animals , Humans , Mice , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Anthocyanins/pharmacology , Anthocyanins/therapeutic use , Antioxidants/pharmacology , Antioxidants/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/complications , Sepsis/drug therapy , Signal Transduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
11.
Biomed Environ Sci ; 37(4): 367-376, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38727159

ABSTRACT

Objective: This study aimed to clarify the intervention effect of salidroside (SAL) on lung injury caused by PM 2.5 in mice and illuminate the function of SIRT1-PGC-1ɑ axis. Methods: Specific pathogen-free (SPF) grade male C57BL/6 mice were randomly assigned to the following groups: control group, SAL group, PM 2.5 group, SAL+PM 2.5 group. On the first day, SAL was given by gavage, and on the second day, PM 2.5 suspension was given by intratracheal instillation. The whole experiment consist of a total of 10 cycles, lasting 20 days. At the end of treatment, blood samples and lung tissues were collected and analyzed. Observation of pathological changes in lung tissue using inverted microscopy and transmission electron microscopy. The expression of inflammatory, antioxidants, apoptosis, and SIRT1-PGC-1ɑ proteins were detected by Western blotting. Results: Exposure to PM 2.5 leads to obvious morphological and pathologica changes in the lung of mice. PM 2.5 caused a decline in levels of antioxidant-related enzymes and protein expressions of HO-1, Nrf2, SOD2, SIRT1 and PGC-1ɑ, and an increase in the protein expressions of IL-6, IL-1ß, Bax, caspase-9 and cleaved caspase-3. However, SAL reversed the aforementioned changes caused by PM 2.5 by activating the SIRT1-PGC-1α pathway. Conclusion: SAL can activate SIRT1-PGC-1ɑ to ameliorate PM 2.5-induced lung injury.


Subject(s)
Glucosides , Lung Injury , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenols , Sirtuin 1 , Animals , Mice , Glucosides/pharmacology , Glucosides/therapeutic use , Lung/drug effects , Lung/pathology , Lung/metabolism , Lung Injury/drug therapy , Particle Size , Particulate Matter/toxicity , Particulate Matter/adverse effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/drug effects , Sirtuin 1/genetics , Sirtuin 1/metabolism
12.
Toxicol Lett ; 350: 121-132, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34252510

ABSTRACT

Silicosis is characterized by pulmonary interstitial fibrosis that arises as a result of chronic exposure to silica. The few available treatments only delay its progression. As α-lipoic acid (ALA) has been shown to have various beneficial effects, including mitoprotective, antioxidant, and anti-inflammatory effects, we hypothesized that it may exhibit therapeutic effects in pulmonary fibrosis. Therefore, in the present study, we used a murine model of silicosis to investigate whether supplementation with exogenous ALA could attenuate silica-induced pulmonary fibrosis by improving mitochondrial function. ALA was administered to the model mice via continuous intragastric administration for 28 days, and then the antioxidant and mitoprotective effects of ALA were evaluated. The results showed that ALA decreased the production of reactive oxygen species, protected mitochondria from silica-induced dysfunction, and inhibited extracellular matrix deposition. ALA also decreased hyperglycemia and hyperlipidemia. Activation of the mitochondrial AMPK/PGC1α pathway might be responsible for these ALA-mediated anti-fibrotic effects. Exogenous ALA blocked oxidative stress by activating NRF2. Taken together, these findings demonstrate that exogenous ALA effectively prevents the progression of silicosis in a murine model, likely by stimulating mitochondrial biogenesis and endogenous antioxidant responses. Therefore, ALA can potentially delay the progression of silica-induced pulmonary fibrosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Silicon Dioxide/adverse effects , Silicosis/drug therapy , Thioctic Acid/therapeutic use , AMP-Activated Protein Kinases/drug effects , Animals , Antioxidants/metabolism , Antioxidants/therapeutic use , Humans , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Models, Animal , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Pulmonary Fibrosis/metabolism , Silicosis/metabolism , Silicosis/physiopathology , Thioctic Acid/metabolism
13.
Curr Pharm Biotechnol ; 22(5): 609-621, 2021.
Article in English | MEDLINE | ID: mdl-33198615

ABSTRACT

BACKGROUND: Osteoporosis, characterized by bone loss, usually occurs with the increased bone resorption and decreased bone formation. H2O2-induced MC3T3-E1 cells are commonly used for the study of osteoblastic activities, which play a crucial role in bone formation. OBJECTIVE: This study aimed to investigate the effects of Phosphocreatine (PCr) on the osteoblastic activities in H2O2-induced MC3T3-E1 cells and elaborate on the possible molecular mechanism. METHODS: The Osteoprotegerin (OPG)/Receptor Activator of NF-κB Ligand (RANKL) ratio and osteogenic markers were detected to investigate the effects of PCr on osteoblastic activities, and the osteoblastic apoptosis was detected using Hochest staining. Moreover, oxidative stress, Adenosine Triphosphate (ATP) generation and the expression of Sirtuin 1 (SIRT1), Forkhead Box O 1 (FOXO1) and Peroxisome Proliferator-Activated Receptor Γ Coactivator-1α (PGC-1α) were also examined to uncover the possible molecular mechanism in H2O2-induced MC3T3-E1 cells. RESULT: The results showed that PCr promoted the osteoblastic differentiation by increasing the expression levels of osteogenic markers of Alkaline Phosphatase (ALP) and Runt-related transcription factor 2 (Runx2), as well as increased the OPG/RANKL ratio and suppressed the osteoblastic apoptosis in H2O2-induced MC3T3-E1 cells. Moreover, treatment with PCr suppressed reactive oxygen species (ROS) over-generation and promoted the ATP production as well as increased the PGC-1α, FOXO1 and SIRT1 protein expression levels in H2O2-induced MC3T3-E1 cells. CONCLUSION: PCr treatment could promote osteoblastic activities via suppressing oxidative stress and increasing the ATP generation in H2O2-induced MC3T3-E1 cells. In addition, the positive effects of PCr on osteoblasts might be regulated by SIRT1/FOXO1/ PGC-1α signaling pathway.


Subject(s)
Forkhead Box Protein O1/drug effects , Hydrogen Peroxide/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Phosphocreatine/pharmacology , Signal Transduction/drug effects , Sirtuin 1/drug effects , 3T3 Cells , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/drug effects , Animals , Apoptosis/drug effects , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/drug effects , Mice , Osteoprotegerin/drug effects , Osteoprotegerin/metabolism , Oxidative Stress , RANK Ligand/drug effects , RANK Ligand/metabolism , Reactive Oxygen Species
14.
Circ Heart Fail ; 14(1): e005890, 2021 01.
Article in English | MEDLINE | ID: mdl-33356364

ABSTRACT

BACKGROUND: We recently reported that treatment with rhBDNF (recombinant human brain-derived neurotrophic factor) improved the reduced exercise capacity of mice with heart failure (HF) after myocardial infarction (MI). Since BDNF is reported to enhance fatty acid oxidation, we herein conducted an in vivo investigation to determine whether the improvement in exercise capacity is due to the enhancement of the fatty acid oxidation of skeletal muscle via the AMPKα-PGC1α (adenosine monophosphate-activated protein kinase-ɑ-proliferator-activated receptor-r coactivator-1ɑ) axis. METHODS: MI and sham operations were conducted in C57BL/6J mice. Two weeks postsurgery, we randomly divided the MI mice into groups treated with rhBDNF or vehicle for 2 weeks. AMPKα-PGC1α signaling and mitochondrial content in the skeletal muscle of the mice were evaluated by Western blotting and transmission electron microscopy. Fatty acid ß-oxidation was examined by high-resolution respirometry using permeabilized muscle fiber. BDNF-knockout mice were treated with 5-aminoimidazole-4-carboxamide-1-beta-d-riboruranoside, an activator of AMPK. RESULTS: The rhBDNF treatment significantly increased the expressions of phosphorylated AMPKα and PGC1α protein and the intermyofibrillar mitochondrial density in the MI mice. The lowered skeletal muscle mitochondrial fatty acid oxidation was significantly improved in the rhBDNF-treated MI mice. The reduced exercise capacity and mitochondrial dysfunction of the BDNF-knockout mice were improved by 5-aminoimidazole-4-carboxamide-1-beta-d-riboruranoside. CONCLUSIONS: Beneficial effects of BDNF on the exercise capacity of mice with HF are mediated through an enhancement of fatty acid oxidation via the activation of AMPKα-PGC1α in skeletal muscle. BDNF may become a therapeutic option to improve exercise capacity as an alternative or adjunct to exercise training.


Subject(s)
AMP-Activated Protein Kinases/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Exercise Tolerance/drug effects , Fatty Acids/metabolism , Heart Failure/metabolism , Muscle, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Recombinant Proteins , Ribonucleosides/pharmacology
15.
Neuropharmacology ; 196: 108685, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34175325

ABSTRACT

Targeting the common molecular mechanism of type 2 diabetes mellitus and Alzheimer's disease (AD), including dysregulation of glucose metabolism, insulin resistance, and neuroinflammation, might be an efficient treatment strategy for AD. Previous studies have shown that 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), an endogenous PPARγ agonist, has anti-inflammatory, insulin sensitizing and anti-diabetic effects. However, whether 15d-PGJ2 has beneficial effects on AD remains to be elucidated. In the present study, we found that intranasal administration of 15d-PGJ2 (300 ng/30 µL/day) for 3 months significantly inhibited Aß plaques, suppressed neuroinflammation, and attenuated cognitive deficits in APP/PS1 transgenic mice. Interestingly, 15d-PGJ2 treatment could increase brain glucose uptake, as detected by 18F-FDG microPET imaging, and co-localization of GLUT4 and NeuN in the hippocampus of APP/PS1 mice. Furthermore, 15d-PGJ2 markedly increased the expression of PPARγ and PGC-1α, upregulated GLUT4, and decreased the phosphorylation of IRS-1 (Ser616) in the hippocampus of APP/PS1 mice. Importantly, co-administration of a PPARγ antagonist GW9662 abrogated these protective effects of 15d-PGJ2. Collectively, intranasal 15d-PGJ2 conferred protective effects against AD by activating PPARγ-dependent PGC-1α/GLUT4 signalling. The PPARγ agonist 15d-PGJ2 might be a potential therapeutic drug for AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/drug effects , Glucose/metabolism , Immunologic Factors/pharmacology , Neurons/drug effects , PPAR gamma/drug effects , Prostaglandin D2/analogs & derivatives , Administration, Intranasal , Alzheimer Disease/genetics , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Anilides/pharmacology , Animals , Behavior, Animal/drug effects , Brain/diagnostic imaging , Brain/metabolism , Cytokines/drug effects , Cytokines/metabolism , Fluorodeoxyglucose F18 , Glucose Transporter Type 4/drug effects , Glucose Transporter Type 4/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Transgenic , Morris Water Maze Test , Neurons/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Positron-Emission Tomography , Presenilin-1/genetics , Prostaglandin D2/pharmacology , Radiopharmaceuticals , Signal Transduction
16.
Aging (Albany NY) ; 12(8): 7411-7430, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32343674

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have great potential in biomedical applications. However, the immature state of cardiomyocytes obtained using existing protocols limits the application of hPSC-CMs. Unlike adult cardiac myocytes, hPSC-CMs generate ATP through an immature metabolic pathway-aerobic glycolysis, instead of mitochondrial oxidative phosphorylation (OXPHOS). Hence, metabolic switching is critical for functional maturation in hPSC-CMs. Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) is a key regulator of mitochondrial biogenesis and metabolism, which may help promote cardiac maturation during development. In this study, we investigated the effects of PGC-1α and its activator ZLN005 on the maturation of human embryonic stem cell-derived cardiomyocyte (hESC-CM). hESC-CMs were generated using a chemically defined differentiation protocol and supplemented with either ZLN005 or DMSO (control) on differentiating days 10 to 12. Biological assays were then performed around day 30. ZLN005 treatment upregulated the expressions of PGC-1α and mitochondrial function-related genes in hESC-CMs and induced more mature energy metabolism compared with the control group. In addition, ZLN005 treatment increased cell sarcomere length, improved cell calcium handling, and enhanced intercellular connectivity. These findings support an effective approach to promote hESC-CM maturation, which is critical for the application of hESC-CM in disease modeling, drug screening, and engineering cardiac tissue.


Subject(s)
Benzimidazoles/pharmacology , Energy Metabolism/drug effects , Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Cell Differentiation , Cells, Cultured , Heat-Shock Proteins , Human Embryonic Stem Cells/metabolism , Humans , Hypoglycemic Agents , Myocytes, Cardiac/drug effects , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Tissue Engineering
17.
Aging (Albany NY) ; 13(1): 493-515, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33259334

ABSTRACT

Mitochondrial calcium uptake 1 (MICU1) is a pivotal molecule in maintaining mitochondrial homeostasis under stress conditions. However, it is unclear whether MICU1 attenuates mitochondrial stress in angiotensin II (Ang-II)-induced cardiac hypertrophy or if it has a role in the function of melatonin. Here, small-interfering RNAs against MICU1 or adenovirus-based plasmids encoding MICU1 were delivered into left ventricles of mice or incubated with neonatal murine ventricular myocytes (NMVMs) for 48 h. MICU1 expression was depressed in hypertrophic myocardia and MICU1 knockdown aggravated Ang-II-induced cardiac hypertrophy in vivo and in vitro. In contrast, MICU1 upregulation decreased cardiomyocyte susceptibility to hypertrophic stress. Ang-II administration, particularly in NMVMs with MICU1 knockdown, led to significantly increased reactive oxygen species (ROS) overload, altered mitochondrial morphology, and suppressed mitochondrial function, all of which were reversed by MICU1 supplementation. Moreover, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α)/MICU1 expression in hypertrophic myocardia increased with melatonin. Melatonin ameliorated excessive ROS generation, promoted mitochondrial function, and attenuated cardiac hypertrophy in control but not MICU1 knockdown NMVMs or mice. Collectively, our results demonstrate that MICU1 attenuates Ang-II-induced cardiac hypertrophy by inhibiting mitochondria-derived oxidative stress. MICU1 activation may be the mechanism underlying melatonin-induced protection against myocardial hypertrophy.


Subject(s)
Antioxidants/pharmacology , Calcium-Binding Proteins/genetics , Cardiomegaly/genetics , Melatonin/pharmacology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Angiotensin II/toxicity , Animals , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Heart/drug effects , In Vitro Techniques , Mice , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reactive Oxygen Species/metabolism , Vasoconstrictor Agents/toxicity
18.
J Diabetes Res ; 2020: 9084567, 2020.
Article in English | MEDLINE | ID: mdl-32090125

ABSTRACT

BACKGROUND: The role of exendin-4 in brown adipose tissue (BAT) activation was not very clear. This study is to verify the role of BAT involved in renal benefits of exendin-4 in diabetes mellitus (DM). METHODS: In vivo, C57BL/6 mice were randomly divided into nondiabetic (control) and diabetic groups (DM). The diabetic mice were randomized into a control group (DM-Con), BAT-excision group (DM+Exc), exendin-4-treated group (DM+E4), and BAT-excision plus exendin-4-treated group (DM+Exc+E4). The weight, blood glucose and lipids, 24 h urine albumin and 8-OH-dG, and renal fibrosis were analyzed. In vitro, we investigated the role of exendin-4 in the differentiation process of 3T3-L1 and brown preadipocytes and its effect on the rat mesangial cells induced by oleate. RESULTS: The expressions of UCP-1, PGC-1α, ATGL, and CD36 in BAT of DM mice were all downregulated, which could be upregulated by exendin-4 treatment with significant effects on ATGL and CD36. BAT-excision exacerbated high blood glucose (BG) with no significant effect on the serum lipid level. Exendin-4 significantly lowered the level of serum triglycerides (TG) and low-density lipoprotein- (LDL-) c, 24 h urine albumin, and 8-OH-dG; improved renal fibrosis and lipid accumulation; and activated renal AMP-activated protein kinase (AMPK) in diabetic mice regardless of BAT excision. In vitro, there was no significant effect of exendin-4 on brown or white adipogenesis. However, exendin-4 could improve lipid accumulation and myofibroblast-like phenotype transition of mesangial cells induced by oleate via activating the AMPK pathway. CONCLUSIONS: Exendin-4 could decrease the renal lipid deposit and improve diabetic nephropathy via activating the renal AMPK pathway independent of BAT activation.


Subject(s)
Adipose Tissue, Brown/drug effects , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Exenatide/pharmacology , Incretins/pharmacology , Kidney/drug effects , 3T3-L1 Cells , 8-Hydroxy-2'-Deoxyguanosine/urine , Adenylate Kinase/metabolism , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipogenesis/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/surgery , Albuminuria , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , CD36 Antigens/drug effects , CD36 Antigens/genetics , Cholesterol, HDL/drug effects , Cholesterol, HDL/metabolism , Cholesterol, LDL/drug effects , Cholesterol, LDL/metabolism , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Disease Models, Animal , Fibrosis , Gene Expression/drug effects , Kidney/pathology , Lipase/drug effects , Lipase/genetics , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Random Allocation , Rats , Real-Time Polymerase Chain Reaction , Triglycerides/metabolism , Uncoupling Protein 1/drug effects , Uncoupling Protein 1/genetics
19.
Neurotox Res ; 38(2): 461-477, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32394056

ABSTRACT

In this study, we aim to assess the phytomedicinal potential of perillyl alcohol (PA), a dietary monoterpenoid, in a unilateral 6-hydroxydopamine (6-OHDA) lesion rat model of Parkinson's disease (PD). We observed that PA supplementation alleviated behavioural abnormalities such as loss of coordination, reduced rearing and motor asymmetry in lesioned animals. We also observed that PA-treated animals exhibited reduced oxidative stress, DNA fragmentation and caspase 3 activity indicating alleviation of apoptotic cell death. We found reduced mRNA levels of pro-apoptotic regulator BAX and pro-inflammatory mediators IL18 and TNFα in PA-treated animals. Further, PA treatment successfully increased mRNA and protein levels of Bcl2, mitochondrial biogenesis regulator PGC1α and tyrosine hydroxylase (TH) in lesioned animals. We observed that PA treatment blocked BAX and Drp1 translocation to mitochondria, an event often associated with the inception of apoptosis. Further, 6-OHDA exposure reduced expression of electron transport chain complexes I and IV, thereby disturbing energy metabolism. Conversely, expression levels of both complexes were upregulated with PA treatment in lesioned rats. Finally, we found that protein levels of Nrf2, the transcription factor responsible for antioxidant gene expression, were markedly reduced in cytosolic and nuclear fraction on 6-OHDA exposure, and PA increased expression of Nrf2 in both fractions. We believe that our data hints towards PA having the ability to provide cytoprotection in a hemiparkinsonian rat model through alleviation of motor deficits, oxidative stress, mitochondrial dysfunction and apoptosis.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Monoterpenes/pharmacology , Movement/drug effects , Oxidative Stress/drug effects , Parkinsonian Disorders/metabolism , Animals , Behavior, Animal/drug effects , Caspase 3/drug effects , Caspase 3/metabolism , DNA Fragmentation/drug effects , Dynamins/drug effects , Dynamins/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/physiopathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Sympatholytics/toxicity , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/genetics , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
20.
Brain Res ; 1714: 126-132, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30826352

ABSTRACT

Growing evidence indicates links between type 2 diabetes and Parkinson's disease. The glucagon-like peptide 1 analogue, liraglutide, a commonly used anti-diabetic drug, has protective effects on neurons. The goal of this study was to determine whether long-term liraglutide treatment could reduce the risk of adult type 2 diabetic mice developing Parkinson's disease. Male diabetic db/db mice (12 weeks old) were injected daily with liraglutide (n = 8), or saline (n = 8), and non-diabetic m/m littermates (n = 6) were included as controls. Motor function was assessed every 4 weeks and all mice were sacrificed after 8 weeks of drug intervention for further analysis. The results revealed that long-term treatment of liraglutide protected the db/db mice against the motor function decay and the dopaminergic neuron loss. Liraglutide also restored the impaired AMP kinase (AMPK)/peroxisome proliferator-activated receptor-γ coactivator 1a (PGC-1a) signaling in the striatum of db/db mice. Further experiments in SH-SY5Y cells supported that AMPK is involved in the neuroprotective effect of liraglutide. In summary, long-term liraglutide ameliorated motor dysfunction and dopaminergic neuron impairment in type 2 diabetic mice, probably via enhancing AMPK/PGC-1a signaling.


Subject(s)
Liraglutide/pharmacology , Motor Disorders/drug therapy , Parkinson Disease/prevention & control , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Glucagon-Like Peptide 1/analogs & derivatives , Hypoglycemic Agents , Liraglutide/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL