Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Chin J Integr Med ; 28(11): 975-982, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34874519

ABSTRACT

OBJECTIVE: To explore the protective effect and underlying mechanism of Lycium barbarum polysaccharides (LBP) in a non-alcoholic fatty liver disease (NAFLD) cell model. METHODS: Normal human hepatocyte LO2 cells were treated with 1 mmol/L free fatty acids (FFA) mixture for 24 h to induce NAFLD cell model. Cells were divided into 5 groups, including control, model, low-, medium- and high dose LBP (30,100 and 300 µg/mL) groups. The monosaccharide components of LBP were analyzed with high performance liquid chromatography. Effects of LBP on cell viability and intracellular lipid accumulation were assessed by cell counting Kit-8 assay and oil red O staining, respectively. Triglyceride (TG), alanine aminotransferase (ALT), aspartate aminotransferase (AST), adenosine triphosphate (ATP) and oxidative stress indicators were evaluated. Energy balance and mitochondrial biogenesis related mRNA and proteins were determined by quantitative real-time polymerase chain reaction and Western blot, respectively. RESULTS: Heteropolysaccharides with mannose and glucose are the main components of LBP. LBP treatment significantly decreased intracellular lipid accumulation as well as TG, ALT, AST and malondialdehyde levels (P<0.05 or P<0.01), increased the levels of superoxide dismutase, phospholipid hydroperoxide glutathione peroxidase, catalase, and ATP in NAFLD cell model (P<0.05). Meanwhile, the expression of uncoupling protein 2 was down-regulated and peroxisome proliferator-activated receptor gamma coactivator-1α/nuclear respiratory factor 1/mitochondrial transcription factor A pathway was up-regulated (P<0.05). CONCLUSION: LBP promotes mitochondrial biogenesis and improves energy balance in NAFLD cell model.


Subject(s)
Drugs, Chinese Herbal , Lycium , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Lycium/chemistry , Lycium/metabolism , Catalase/metabolism , Organelle Biogenesis , Alanine Transaminase , Uncoupling Protein 2 , Fatty Acids, Nonesterified , Mannose , Nuclear Respiratory Factor 1/metabolism , PPAR gamma/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Drugs, Chinese Herbal/pharmacology , Malondialdehyde/metabolism , Superoxide Dismutase/metabolism , Polysaccharides/pharmacology , Triglycerides , RNA, Messenger , Aspartate Aminotransferases , Glucose , Adenosine Triphosphate
2.
Mar Drugs ; 19(4)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810216

ABSTRACT

Mitochondrial dysfunction contributes to the pathogenesis of kidney injury related with cardiovascular disease. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) protects renal tubular cells by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2). AMP-activated protein kinase (pAMPK)-mediated phosphorylation and sirtuin 1/3 (SIRT1/3)-mediated deacetylation are required for PGC-1α activation. In the present study, we aimed to investigate whether omega-3 fatty acids (FAs) regulate the expression of mediators of mitochondrial biogenesis in 5/6 nephrectomy (Nx) rats. Male Sprague-Dawley rats were assigned to the following groups: sham control, Nx, and Nx treated with omega-3 FA. The expression of PGC-1α, phosphorylated PGC-1α (pPGC-1α), acetylated PGC-1α, and factors related to mitochondrial biogenesis was examined through Western blot analysis. Compared to the control group, the expression of PGC-1α, pAMPK, SIRT1/3, Nrf1, mTOR, and Nrf2 was significantly downregulated, and that of Keap 1, acetylated PGC-1α, and FoxO1/3, was significantly upregulated in the Nx group. These changes in protein expression were rescued in the omega-3 FA group. However, the expression of pPGC-1α was similar among the three groups. Omega-3 FAs may involve mitochondrial biogenesis by upregulating Nrf1 and Nrf2. This protective mechanism might be attributed to the increased expression and deacetylation of PGC-1α, which was triggered by SIRT1/3.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Kidney Diseases/drug therapy , Kidney/drug effects , Mitochondria/drug effects , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/metabolism , Sirtuins/metabolism , Acetylation , Animals , Disease Models, Animal , Kidney/enzymology , Kidney/pathology , Kidney Diseases/enzymology , Kidney Diseases/etiology , Kidney Diseases/pathology , Male , Mitochondria/enzymology , Mitochondria/pathology , NF-E2-Related Factor 2/metabolism , Nephrectomy , Organelle Biogenesis , Protein Processing, Post-Translational , Rats, Sprague-Dawley , Signal Transduction
3.
Nat Commun ; 12(1): 256, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431871

ABSTRACT

In humans, inactivating mutations in MLL4, which encodes a histone H3-lysine 4-methyltransferase, lead to Kabuki syndrome (KS). While dwarfism is a cardinal feature of KS, the underlying etiology remains unclear. Here we report that Mll4 regulates the development of growth hormone-releasing hormone (GHRH)-producing neurons in the mouse hypothalamus. Our two Mll4 mutant mouse models exhibit dwarfism phenotype and impairment of the developmental programs for GHRH-neurons. Our ChIP-seq analysis reveals that, in the developing mouse hypothalamus, Mll4 interacts with the transcription factor Nrf1 to trigger the expression of GHRH-neuronal genes. Interestingly, the deficiency of Mll4 results in a marked reduction of histone marks of active transcription, while treatment with the histone deacetylase inhibitor AR-42 rescues the histone mark signature and restores GHRH-neuronal production in Mll4 mutant mice. Our results suggest that the developmental dysregulation of Mll4-directed epigenetic control of transcription plays a role in the development of GHRH-neurons and dwarfism phenotype in mice.


Subject(s)
Growth Hormone-Releasing Hormone/biosynthesis , Histone-Lysine N-Methyltransferase/metabolism , Hypothalamus/cytology , Neurons/metabolism , Animals , Base Sequence , Dwarfism/metabolism , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Hypothalamus/embryology , Male , Mice, Knockout , Models, Biological , Nuclear Respiratory Factor 1/metabolism , Phenylbutyrates/pharmacology , Transcription Factors/metabolism
4.
Free Radic Biol Med ; 163: 141-152, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33276082

ABSTRACT

Targeting energy metabolism holds the potential to effectively treat a variety of malignant diseases, and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) is a key regulator of energy metabolism. However, PGC1α's role in cancer, especially in hepatocellular carcinoma (HCC) remains largely unknown. In the present study, we reported that PGC1α was significantly downregulated in HCC cell lines and specimens. Moreover, reduced expression of PGC1α in tumor cells was correlated with poor prognosis. PGC1α overexpression substantially inhibited cell proliferation and induced apoptosis in vitro and in vivo. On the contrary, the knockdown of PGC1α produced the opposite effect. The mechanism was at least partially due to the upregulation of mitochondrial pyruvate carrier 1 (MPC1) caused by PGC1α, which promoted mitochondrial biogenesis by binding to nuclear respiratory factor 1 (NRF1). Consequently, the production of cellular reactive oxygen species (ROS) caused by mitochondrial oxidation was elevated above a critical threshold for survival. Furthermore, we found that PGC1α could enhance the antitumor activity of sorafenib and doxorubicin in HCC through ROS accumulation-mediated cell death. These results indicate that PGC1α/NRF1-MPC1 axis is involved in HCC progression and could be a promising target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Doxorubicin/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sorafenib/pharmacology
5.
Med Sci Monit Basic Res ; 26: e928648, 2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33361744

ABSTRACT

BACKGROUND Excessive reactive oxygen species (ROS) stimulate mitochondrial damage that causes degenerative diseases such as cardiovascular disease (CVD). ß-carotene (BC), a natural antioxidant able to counteract free radicals, acts as a cytoprotective agent. However, knowledge of the role of BC on cardiomyoblasts is limited. In this study, we explored its role on COX4, Tom20, Nfr1, Nrf2, Nf-kappaB, LC3, p62, caspase 3, and caspase 9 and its association with cardiomyoblast viability and survival. MATERIAL AND METHODS H9C2 cell lines were seeded, cultivated until 90% to 100% confluency, and treated with various doses of BC: 10 µM, 1 µM, 0.1 µM, and 0.01 µM. After 24 h, the cells were harvested, lyzed, and tested for specific related protein expressions from each dose. RESULTS Low-dose BC induced autophagy most effectively at 1 µM, 0.1 µM, and 0.01 µM, as indicated by a decrease of LC3II and p62 levels. We observed that Nf-kB protein levels were suppressed; Nrf2 was stimulated, but Nrf1 was not altered significantly. Further, low-dose BC might stimulate cell viability by reducing apoptotic signals of caspase 3 and 9. Notably, low-dose BC also showed potential to increase Tom20 protein levels. CONCLUSIONS Low-dose BC supplementation shows beneficial effects, especially at 0.01 µM, by reducing inflammation through the suppression of Nf-kappaB and increase of Nrf2 level. Autophagy as a cellular maintenance mechanism was also stimulated, and the amount of the mitochondria marker Tom20 increased. Taken together, results showed that specific low-dose BC is effective and might improve cell viability by stimulating autophagy, inhibiting proinflammatory factors, and suppressing apoptosis.


Subject(s)
Autophagy , Caspases/metabolism , Inflammation/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction , beta Carotene/pharmacology , Animals , Autophagy/drug effects , Biomarkers/metabolism , Cell Line , Cell Shape/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nuclear Respiratory Factor 1/metabolism , Rats , Signal Transduction/drug effects
6.
Am J Chin Med ; 48(3): 631-650, 2020.
Article in English | MEDLINE | ID: mdl-32329640

ABSTRACT

The loss of skeletal muscle mass and function is a serious consequence of chronic diseases and aging. BST204 is a purified ginseng (the root of Panax ginseng) extract that has been processed using ginsenoside-ß-glucosidase and acid hydrolysis to enrich ginsenosides Rg3 and Rh2 from the crude ginseng. BST204 has a broad range of health benefits, but its effects and mechanism on muscle atrophy are currently unknown. In this study, we have examined the effects and underlying mechanisms of BST204 on myotube formation and myotube atrophy induced by tumor necrosis factor-α (TNF-α). BST204 promotes myogenic differentiation and multinucleated myotube formation through Akt activation. BST204 prevents myotube atrophy induced by TNF-α through the activation of Akt/mTOR signaling and down-regulation of muscle-specific ubiquitin ligases, MuRF1, and Atrogin-1. Furthermore, BST204 treatment in atrophic myotubes suppresses mitochondrial reactive oxygen species (ROS) production and regulates mitochondrial transcription factors such as NRF1 and Tfam, through enhancing the activity and expression of peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α). Collectively, our findings indicate that BST204 improves myotube formation and PGC1α-mediated mitochondrial function, suggesting that BST204 is a potential therapeutic or neutraceutical remedy to intervene muscle weakness and atrophy.


Subject(s)
Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Panax/chemistry , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Animals , Atrophy/chemically induced , Atrophy/drug therapy , Humans , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Plant Extracts/isolation & purification , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Stimulation, Chemical , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha
7.
Molecules ; 25(4)2020 Feb 16.
Article in English | MEDLINE | ID: mdl-32079067

ABSTRACT

Red ginseng has been reported to elicit various therapeutic effects relevant to cancer, diabetes, neurodegenerative diseases, and inflammatory diseases. However, the effect of red ginseng on exercise endurance and skeletal muscle function remains unclear. Herein, we sought to investigate whether red ginseng could affect exercise endurance and examined its molecular mechanism. Mice were fed with red ginseng extract (RG) and undertook swimming exercises to determine the time to exhaustion. Animals fed with RG had significantly longer swimming endurance. RG treatment was also observed to enhance ATP production levels in myoblasts. RG increased mRNA expressions of mitochondrial biogenesis regulators, NRF-1, TFAM, and PGC-1α, which was accompanied by an elevation in mitochondrial DNA, suggesting an enhancement in mitochondrial energy-generating capacity. Importantly, RG treatment induced phosphorylation of p38 and AMPK and upregulated PGC1α expression in both myoblasts and in vivo muscle tissue. In addition, RG treatment also stimulated C2C12 myogenic differentiation. Our findings show that red ginseng improves exercise endurance, suggesting that it may have applications in supporting skeletal muscle function and exercise performance.


Subject(s)
Mitochondria/drug effects , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Panax/chemistry , Physical Endurance/drug effects , Plant Extracts/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Cell Differentiation/drug effects , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , Physical Endurance/physiology , Plant Extracts/isolation & purification , Swimming/physiology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Oncol Rep ; 42(3): 1214-1224, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31322174

ABSTRACT

Tetramethylpyrazine (TMP; an extract of the Chinese herbal medicine, Chuanxiong) has been shown to exert remarkable antiretinoblastoma (RB) effects. Based on our previous study, the target gene was found to be C­X­C chemokine receptor type 4 (CXCR4). CXCR4 is a prognostic marker in various types of cancer, but the exact mechanisms underlying the regulation of CXCR4 expression by TMP in WERI­Rb1 cells have yet to be fully elucidated. In the present study, it was revealed that TMP significantly downregulated CXCR4 expression and inhibited CXCR4 promoter activity in WERI­Rb1 cells, indicating that TMP inhibits CXCR4 expression in WERI­Rb1 cells through transcriptional regulatory mechanisms. Among the numerous transcription factors involved in CXCR4 function, including Yin Yang 1 (YY1), nuclear respiratory factor­1 (Nrf­1), Krüppel­like Factor 2 (KLF2), specificity protein 1 (SP1), and nuclear factor­κB subunit 1 (NF­κB1), only TMP led to a significant downregulation of Nrf­1 expression. Chromatin immunoprecipitation assays further indicated that Nrf­1 directly binds to the promoter region of CXCR4, and silencing Nrf­1 via siRNA transfection notably inhibited CXCR4 expression in WERI­Rb1 cells. In addition, the expression levels of both Nrf­1 and CXCR4 increased concomitantly with WERI­Rb1 cell density. Furthermore, the downregulation of Nrf­1 and CXCR4 expression in RB by TMP was confirmed in vivo. Taken together, the results of the present study have uncovered a novel mechanism in which CXCR4 expression is regulated by Nrf­1 in WERI­Rb1 cells, thereby identifying novel potential targets for the treatment of RB, and providing evidence for the clinical application of TMP in adjuvant retinoblastoma therapy.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Nuclear Respiratory Factor 1/metabolism , Pyrazines/pharmacology , Receptors, CXCR4/genetics , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Transcription, Genetic/drug effects , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Nuclear Respiratory Factor 1/genetics , Receptors, CXCR4/metabolism , Retinal Neoplasms/drug therapy , Retinal Neoplasms/genetics , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Tumor Cells, Cultured , Vasodilator Agents/pharmacology , Xenograft Model Antitumor Assays
9.
Cell Physiol Biochem ; 50(5): 1726-1739, 2018.
Article in English | MEDLINE | ID: mdl-30384366

ABSTRACT

BACKGROUND/AIMS: Shengmai San (SMS), prepared from Panax ginseng, Ophiopogon japonicus, and Schisandra chinensisin, has been widely used to treat ischemic disease. In this study, we investigated whether SMS may exert a beneficial effect in diabetic cardiomyopathy through improvement of mitochondrial lipid metabolism. METHODS: A leptin receptor-deficient db/db mouse model was utilized, and lean age-matched C57BLKS mice served as non-diabetic controls. Glucose and lipid profiles, myocardial structure, dimension, and function, and heart weight to tibial length ratio were determined. Myocardial ultrastructural morphology was observed with transmission electron microscopy. Protein expression and activity of oxidative phosphorylation (OXPHOS) complex were assessed using western blotting and microplate assay kits. We also observed cellular viability, mitochondrial membrane potential, OXPHOS complex activity, and cellular ATP level in palmitic acid-stimulated H9C2 cardiomyocytes. Changes in the sirtuin (SIRT1)/AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) pathway and mitochondrial uncoupling signaling were assessed using western blotting and quantitative real-time PCR. RESULTS: Leptin receptor-deficient db/db mice exhibit obesity, hyperglycemia, and hyperlipidemia, accompanied by distinct myocardial hypertrophy and diastolic dysfunction. SMS at a dose of 3 g/kg body weight contributed to a recovery of diabetes-induced myocardial hypertrophy and diastolic dysfunction. SMS administration led to an effective restoration of mitochondrial structure and function both in vivo and in vitro. Furthermore, SMS markedly enhanced SIRT1 and p-AMPKα protein levels and decreased the expression of acetylated-PGC-1α and uncoupling protein 2 protein. SMS also restored the depletion of NRF1 and TFAM levels in diabetic hearts and H9C2 cardiomyocytes. CONCLUSION: The results indicate that SMS may alleviate diabetes-induced myocardial hypertrophy and diastolic dysfunction by improving mitochondrial lipid metabolism.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Mitochondria/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/veterinary , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Drug Combinations , Drugs, Chinese Herbal/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nuclear Respiratory Factor 1/metabolism , Oxidative Phosphorylation/drug effects , Palmitic Acid/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Signal Transduction/drug effects , Sirtuin 1/metabolism , Uncoupling Protein 2/metabolism
10.
Invest Ophthalmol Vis Sci ; 59(5): 2133-2141, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29801148

ABSTRACT

Purpose: Tetramethylpyrazine (TMP) is the active ingredient extracted from the Chinese herb Chuanxiong. The purpose of our study was to identify the mechanism of therapeutic TMP suppression of pathologic chemokine receptor 4 (CXCR4) transcription. Methods: C57BL/6J mice with alkali-burned corneas were treated with either TMP eye drops (1.5 mg/mL) or PBS. Corneal neovascularization (CNV) was measured and a clinical assessment was made by slit lamp microscopy. Expression of CXCR4 and the transcription factors nuclear respiratory factor-1 (NRF-1), nuclear factor kappa B (NFκB), forkhead box C1, and yin yang 1 were tracked by real-time RT-PCR and immunofluorescence staining of murine corneas. Western blot, real-time PCR, and immunofluorescence evaluated expression of related genes in human umbilical vein endothelial cells (HUVECs) after 200-µmol/L TMP treatment. In addition, plasmid transfection and chromatin immunoprecipitation assays elucidated the relationship among NRF-1, NFκB, and CXCR4. Results: Corneas treated with TMP had smaller areas of neovascularization and scored better in clinical assessments. Injured corneas showed significantly elevated expressions of NRF-1, NFκB, and CXCR4 that were normalized in vivo by TMP treatment. Similarly, in HUVECs in vitro, TMP decreased expression of NRF-1, NFκB, and CXCR4. Overexpression of NFκB or NRF-1 raised the expression of CXCR4 in HUVECs, but not synergistically. Chromatin immunoprecipitation assays detected only NRF-1 bound to the CXCR4 promoter region, suggesting NFκB controls CXCR4 expression by upregulating NRF-1. Together, our data suggest TMP downregulates CXCR4 by repressing NRF-1 expression in CNV, likely indirectly by downregulating NFκB. Conclusions: Our results implicate a novel mechanism wherein TMP inhibits neovascularization via an NFκB/NRF-1/CXCR4 circuit.


Subject(s)
Burns, Chemical/drug therapy , Corneal Neovascularization/prevention & control , Eye Burns/chemically induced , NF-kappa B/metabolism , Nuclear Respiratory Factor 1/metabolism , Pyrazines/therapeutic use , Receptors, CXCR4/metabolism , Animals , Blotting, Western , Burns, Chemical/metabolism , Corneal Neovascularization/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Nuclear Respiratory Factor 1/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Sodium Hydroxide , Vasodilator Agents/therapeutic use
11.
J Nutr Biochem ; 38: 125-133, 2016 12.
Article in English | MEDLINE | ID: mdl-27736732

ABSTRACT

Previously, it was reported that conjugated linoleic acid (CLA) with exercise training potentially improved endurance capacity via the peroxisome proliferator-activated receptor δ (PPARδ)-mediated mechanism in mice. This study determined the role of exercise and/or CLA in endurance capacity and PPARδ-associated regulators. Male 129Sv/J mice were fed either control (soybean oil) or CLA (0.5%) containing diets for 4 weeks and were further divided into sedentary or training regimes. CLA supplementation significantly reduced body weight and fat mass independent of exercise during the experimental period. Endurance capacity was significantly improved by CLA supplementation, while no effect of exercise was observed. Similarly, CLA treatment significantly increased expressions of sirtuin 1 and PPARγ coactivator-1α, up-stream regulators of PPARδ, in both sedentary and trained animals. With respect to downstream markers of PPARδ, CLA up-regulated the key biomarker needed to stimulate mitochondrial biogenesis, nuclear respiratory factor 1. Moreover, CLA supplementation significantly induced overall genes associated with muscle fibers, such as type I (slow-twitch) and type II (fast twitch). Taken together, it suggests that CLA improves endurance capacity independent of mild-intensity exercise via PPARδ-mediated mechanism.


Subject(s)
Dietary Supplements , Gene Expression Regulation , Linoleic Acids, Conjugated/administration & dosage , Muscle, Skeletal/metabolism , PPAR gamma/agonists , Performance-Enhancing Substances/administration & dosage , Physical Endurance , Adiposity , Animals , Biomarkers/metabolism , Dietary Supplements/adverse effects , Energy Intake , Energy Metabolism , Linoleic Acids, Conjugated/adverse effects , Male , Mice, 129 Strain , Mitochondria, Muscle/metabolism , Mitochondrial Dynamics , Nuclear Respiratory Factor 1/agonists , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , PPAR gamma/genetics , PPAR gamma/metabolism , Performance-Enhancing Substances/adverse effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Random Allocation , Sirtuin 1/chemistry , Sirtuin 1/genetics , Sirtuin 1/metabolism , Weight Gain
12.
Exp Cell Res ; 347(1): 222-231, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27515002

ABSTRACT

Acquired tamoxifen (TAM) resistance is a significant clinical problem in treating patients with estrogen receptor α (ERα)+ breast cancer. We reported that ERα increases nuclear respiratory factor-1 (NRF-1), which regulates nuclear-encoded mitochondrial gene transcription, in MCF-7 breast cancer cells and NRF-1 knockdown stimulates apoptosis. Whether NRF-1 and target gene expression is altered in endocrine resistant breast cancer cells is unknown. We measured NRF-1and metabolic features in a cell model of progressive TAM-resistance. NRF-1 and its target mitochondrial transcription factor A (TFAM) were higher in TAM-resistant LCC2 and LCC9 cells than TAM-sensitive MCF-7 cells. Using extracellular flux assays we observed that LCC1, LCC2, and LCC9 cells showed similar oxygen consumption rate (OCR), but lower mitochondrial reserve capacity which was correlated with lower Succinate Dehydrogenase Complex, Subunit B in LCC1 and LCC2 cells. Complex III activity was lower in LCC9 than MCF-7 cells. LCC1, LCC2, and LCC9 cells had higher basal extracellular acidification (ECAR), indicating higher aerobic glycolysis, relative to MCF-7 cells. Mitochondrial bioenergetic responses to estradiol and 4-hydroxytamoxifen were reduced in the endocrine-resistant cells compared to MCF-7 cells. These results suggest the acquisition of altered metabolic phenotypes in response to long term antiestrogen treatment may increase vulnerability to metabolic stress.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Energy Metabolism , Nuclear Respiratory Factor 1/metabolism , Tamoxifen/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Respiration/drug effects , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Electron Transport Complex III/metabolism , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation/drug effects , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Tamoxifen/analogs & derivatives , Transcription Factors/metabolism
13.
Biomed Pharmacother ; 79: 201-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27044829

ABSTRACT

PURPOSE: Aloe vera is compound which has strong antioxidant and anti-inflammatory effects. We investigated the neuroprotective role of aloe vera treatment in rats with experimental sciatic nerve ischemia/reperfusion injury. METHODS: Twenty-eight male Wistar Albino rats were divided equally into 4 groups. Groups; Control group (no surgical procedure or medication), sciatic nerve ischemia/reperfusion group, sciatic nerve ischemia/reperfusion+aloe vera group and sciatic nerve ischemia/reperfusion+methylprednisolone group. Ischemia was performed by clamping the infrarenal abdominal aorta. 24 hours after ischemia, all animals were sacrificed. Sciatic nerve tissues were also examined histopathologically and biochemically. RESULTS: Ischemic fiber degeneration significantly decreased in the pre-treated with aloe vera and treated with methylprednisolone groups, especially in the pre-treated with aloe vera group, compared to the sciatic nerve ischemia/reperfusion group (p<0.05). A significant decrease in MDA, an increase in NRF1 level and SOD activity were observed in the groups which obtained from the AV and MP groups when compared to the sciatic nerve ischemia/reperfusion group. When all results were analysed it was seen that the aloe vera group was not statistically different compared to the MP group (p>0.05). CONCLUSIONS: Aloe vera is effective neuroprotective against sciatic nerve ischemia/reperfusion injury via antioxidant and anti-inflammatory properties. Also aloe vera was found to be as effective as MP.


Subject(s)
Aloe/chemistry , Plant Extracts/therapeutic use , Reperfusion Injury/drug therapy , Sciatic Nerve/pathology , Animals , Axons/drug effects , Axons/pathology , Male , Malondialdehyde/metabolism , Myelin Sheath/drug effects , Myelin Sheath/pathology , NF-kappa B/metabolism , Nuclear Respiratory Factor 1/metabolism , Plant Extracts/pharmacology , Rats, Wistar , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/drug effects , Superoxide Dismutase/metabolism
14.
J Pharm Pharmacol ; 68(1): 119-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26806698

ABSTRACT

OBJECTIVES: The aim of this study was to determine the protective mechanisms of wild ginseng cambial meristematic cells (CMCs) on non-alcoholic fatty liver disease in high-fat diet (HFD)-fed mice. METHODS: Male C57BL/6 mice received either normal-fat diet or HFD for 10 weeks along with wild ginseng CMCs (75, 150 and 300 mg/kg) or vehicle (0.5% carboxyl methyl cellulose) by oral administration once a day. Triglyceride and total cholesterol contents were measured in liver and serum samples. Parameters for hepatic lipid metabolism and mitochondria biogenesis were assessed. KEY FINDINGS: Treatment with wild ginseng CMCs markedly attenuated body weight, serum and hepatic lipid contents, and serum aminotransferase activity. While wild ginseng CMCs attenuated the increases in sterol regulatory element-binding transcription factor 1 (SREBP-1) and carbohydrate-responsive element-binding protein (ChREBP) expression, it enhanced the increases in carnitine palmitoyltransferase 1A (CPT1A) and peroxisome proliferator-activated receptor alpha (PPAR-α) expression. HFD decreased glutamate dehydrogenase activity and glutathione content, and increased lipid peroxidation, which were all attenuated by wild ginseng CMCs. Furthermore, wild ginseng CMCs enhanced mitochondrial biogenesis-related factors, including peroxisome proliferator-activated receptor-γ co activator 1α (PGC1α), nuclear respiratory factor 1 (NRF1) and mitochondrial transcription factor A (TFAM). CONCLUSIONS: Wild ginseng CMCs protect against HFD-induced liver injury, which prevents lipid accumulation and mitochondrial oxidative stress, and enhances mitochondrial biogenesis.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Mitochondrial Diseases/drug therapy , Panax/chemistry , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Body Weight/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Cholesterol/blood , Cholesterol/metabolism , DNA-Binding Proteins/metabolism , Fatty Liver/blood , Fatty Liver/metabolism , Glutathione/metabolism , High Mobility Group Proteins/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondrial Diseases/blood , Mitochondrial Diseases/metabolism , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Nuclear Proteins/metabolism , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , PPAR alpha/metabolism , RNA-Binding Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transaminases/metabolism , Transcription Factors/metabolism , Triglycerides/blood
15.
Int J Clin Exp Pathol ; 8(3): 2515-24, 2015.
Article in English | MEDLINE | ID: mdl-26045757

ABSTRACT

OBJECTIVE: Previous studies have shown that Astragalus polysaccharides (APS) can be used to ameliorate cardiotoxicity due to chemotherapy and improve the cardiac function. However, the mechanism by which APS mediate this effect is unclear. In the present study, the effects of APS, which suppressed ROS-mediated apoptosis through Nrf1 accumulation in human cardiac myocytes (HCMs), was investigated. METHODS: The cell viability was detected by the CCK8 assay. The cell apoptosis was assessed by annexin V-PI double-labeling staining. Expression of genes and proteins were analyzed by real-time PCR and western blotting respectively. Nrf1 gene was overexpressed using a lentiviral expression vector in HCMs in vitro, in order to explore the mechanism by which the Nrf1 promoted cell growth. RESULTS: CCK8 and Annexin V-PI double-labeling showed that PAL induced cell death in a concentration-dependent manner, and suppressed HCMs proliferation. The combination PAL with APS was significantly decreased the percentage of the early phase of apoptosis cells. ROS levels were increased in HCMs by exposure to PAL. APS treatment significantly inhibited generation of ROS in response to palmitate. Moreover, PAL administration significantly decreased the mRNA and proteins expression of Bcl-2 as well as increased the mRNA expression of BAX and the protein expression of caspase-3 and caspase-8 as compare to those of control group, but APS treatment could reverse PA-induced HCMs apoptosis. The levels of reactive oxygen species (ROS), which was an oxidative stress marker, was significantly increased in cardiomyocytes by exposure to PAL, but overexpressing Nrf1 could ameliorate ROS-induced cardiomyocyte toxicity and increase the expression of SOD1 and SOD2 in HCMs by overexpressing Nrf1. CONCLUSIONS: This study demonstrated that the PAL could induce HCMs apoptosis. However, APS could reverse PAL-induced cardiomyocyte toxicity, at least partially, through suppression ROS and Nrf1 accumulation in HCMs.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Myocytes, Cardiac/drug effects , Nuclear Respiratory Factor 1/metabolism , Reactive Oxygen Species/metabolism , Astragalus Plant , Blotting, Western , Cell Line , Cell Survival/drug effects , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Palmitates/toxicity , Plant Roots , Polysaccharides/pharmacology , Real-Time Polymerase Chain Reaction , Transcriptome , Transfection
16.
Food Funct ; 6(2): 401-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25554947

ABSTRACT

Mulberry extract (ME) has been shown to possess beneficial effects towards obesity, but its mechanism is still unclear. In small mammals, mitochondria enriched brown adipose tissue (BAT) is known to convert protein's electrochemical energy to heat and maintain a constant body temperature. Improving the mitochondrial function or increasing the number of mitochondria could promote the metabolism of carbohydrate and fat. Thus, this study was designed to investigate the mitochondrial function regulated by ME and mulberry wine extract (MWE) during the brown adipogenesis. The C3H10T1/2 mesenchymal stem cell was treated with ME and MWE, both of which significantly (p < 0.05) increased the expression levels of fatty acid oxidation related genes such as peroxisome proliferator-activated receptor-γ coactivator-1α, PR domain-containing 16 and carnitine palmitoyltransferase 1α during brown adipogenesis. These changes were accompanied with increases in mitochondrial oxidative complex proteins upon ME and/or MWE exposure. Notably, ME and/or MWE also significantly (p < 0.05) increased the expression of the transcription factor A and the nuclear respiratory factor-1, which are the key transcription factors of mitochondrial biogenesis. In parallel, the mitochondrial copy number and brown adipose tissue specific gene-uncoupling protein-1 expression were dramatically (p < 0.05) elevated after ME or MWE treatment. Cyanidin-3-glucoside (Cy-3-glu) was found to be one of the most abundant anthocyanins in ME and MWE. Therefore, the BAT regulatory activity of ME and MWE might be, at least in part, due to the effect of Cy-3-glu. These results suggested that ME and MWE could ameliorate metabolic disease through an improvement in mitochondrial functions.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue, Brown/metabolism , Mitochondria/drug effects , Morus/chemistry , Plant Extracts/pharmacology , Wine/analysis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Animals , Anthocyanins/pharmacology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Glucosides/pharmacology , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C3H , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , Oxygen Consumption/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Exp Gerontol ; 56: 77-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24704714

ABSTRACT

In this paper we analyzed changes in hepatocyte mitochondrial mass and ultrastructure as well as in mitochondrial markers of fission/fusion and biogenesis in mice subjected to 40% calorie restriction (CR) for 18 months versus ad libitum-fed controls. Animals subjected to CR were separated into three groups with different dietary fats: soybean oil (also in controls), fish oil and lard. Therefore, the effect of the dietary fat under CR was studied as well. Our results show that CR induced changes in hepatocyte and mitochondrial size, in the volume fraction occupied by mitochondria, and in the number of mitochondria per hepatocyte. Also, mean number of mitochondrial cristae and lengths were significantly higher in all CR groups compared with controls. Finally, CR had no remarkable effects on the expression levels of fission and fusion protein markers. However, considerable differences in many of these parameters were found when comparing the CR groups, supporting the idea that dietary fat plays a relevant role in the modulation of CR effects in aged mice.


Subject(s)
Aging/pathology , Caloric Restriction , Dietary Fats/administration & dosage , Hepatocytes/ultrastructure , Mitochondria, Liver/ultrastructure , Age Factors , Aging/metabolism , Animals , Biomarkers/metabolism , Cell Size , Fish Oils/administration & dosage , Hepatocytes/metabolism , Lipid Peroxides/metabolism , Male , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Dynamics , Mitochondrial Size , Mitochondrial Turnover , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Soybean Oil/administration & dosage , Time Factors , Transcription Factors/metabolism
18.
J Appl Physiol (1985) ; 116(8): 1078-87, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24610533

ABSTRACT

While exercise benefits have been well documented in patients with chronic diseases, the mechanistic understanding of cachectic muscle's response to contraction is essentially unknown. We previously demonstrated that treadmill exercise training attenuates the initiation of cancer cachexia and the development of metabolic syndrome symptoms (Puppa MJ, White JP, Velazquez KT, Baltgalvis KA, Sato S, Baynes JW, Carson JA. J Cachexia Sarcopenia Muscle 3: 117-137, 2012). However, cachectic muscle's metabolic signaling response to a novel, acute bout of low-frequency contraction has not been determined. The purpose of this study was to determine whether severe cancer cachexia disrupts the acute contraction-induced response to low-frequency muscle contraction [low-frequency stimulation (LoFS)]. Metabolic gene expression and signaling was examined 3 h after a novel 30-min bout of contraction (10 Hz) in cachectic Apc(Min/+) (Min) and C57BL/6 (BL-6) mice. Pyrrolidine dithiocarbamate, a STAT/NF-κB inhibitor and free radical scavenger, was administered systemically to a subset of mice to determine whether this altered the muscle contraction response. Although glucose transporter-4 mRNA was decreased by cachexia, LoFS increased muscle glucose transporter-4 mRNA in both BL-6 and Min mice. LoFS also induced muscle peroxisome proliferator-activated receptor-γ and peroxisome proliferator-activated receptor-α coactivator-1 mRNA. However, in Min mice, LoFS was not able to induce muscle proliferator-activated receptor-α coactivator-1 targets nuclear respiratory factor-1 and mitochondrial transcription factor A mRNA. LoFS induced phosphorylated-S6 in BL-6 mice, but this induction was blocked by cachexia. Administration of pyrrolidine dithiocarbamate for 24 h rescued LoFS-induced phosphorylated-S6 in cachectic muscle. LoFS increased muscle phosphorylated-AMP-activated protein kinase and p38 in BL-6 and Min mice. These data demonstrate that cachexia alters the muscle metabolic response to acute LoFS, and combination therapies in concert with muscle contraction may be beneficial for improving muscle mass and function during cachexia.


Subject(s)
Cachexia/physiopathology , Cachexia/therapy , Electric Stimulation Therapy/methods , Muscle, Skeletal/physiopathology , AMP-Activated Protein Kinases/metabolism , Animals , Cachexia/genetics , Cytochromes c/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation , Genes, APC , Hand Strength/physiology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pyrrolidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Thiocarbamates/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Mol Nutr Food Res ; 57(11): 1931-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23818290

ABSTRACT

SCOPE: Lipid homeostasis in vertebrates is regulated at many levels including synthesis, degradation, and distribution. MicroRNAs (miRNAs) are key regulators of lipid homeostasis. The use of phytochemicals to target miRNA (miR) could provide new therapeutic approaches to human diseases. Thus, we investigated the regulation of lipid metabolism by the flavonoid fisetin during experimental analysis of hepatic miRs in mice. METHODS AND RESULTS: Mice were separated into three groups. One group was maintained on the normal diet and the other two groups were fed either a high-fat (HF) diet or HF supplemented with fisetin. We found that fisetin lowered hepatic fat accumulation in HF mice and reversed abnormal expressions of lipid metabolism genes. The co-expression of miR-378 and its host gene PGC-1ß was significantly induced by HF, whereas fisetin prevented the induction of both genes. We also identified nuclear respiratory factor-1 (NRF-1), a critical regulator of the mitochondrial function, as a direct target of miR-378. CONCLUSION: Dietary fisetin protects against hepatosteatosis in association with modulation of lipid metabolism genes and miR-378 in mice. These observations suggest that the use of fisetin to target miRs could be an effective prevention or intervention against metabolic diseases.


Subject(s)
Dietary Supplements , Fatty Liver/prevention & control , Flavonoids/administration & dosage , Liver/drug effects , MicroRNAs/metabolism , Obesity/prevention & control , Animals , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Flavonols , Gene Expression Regulation , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Obesity/etiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Triglycerides/metabolism
20.
Basic Res Cardiol ; 108(3): 329, 2013 May.
Article in English | MEDLINE | ID: mdl-23460046

ABSTRACT

Impaired mitochondrial biogenesis causes skeletal muscle damage in diabetes. However, whether and how mitochondrial biogenesis is impaired in the diabetic heart remains largely unknown. Whether adiponectin (APN), a potent cardioprotective molecule, regulates cardiac mitochondrial function has also not been previously investigated. In this study, electron microscopy revealed significant mitochondrial disorders in ob/ob cardiomyocytes, including mitochondrial swelling and cristae disorientation and breakage. Moreover, mitochondrial biogenesis of ob/ob cardiomyocytes is significantly impaired, as evidenced by reduced Ppargc-1a/Nrf-1/Tfam mRNA levels, mitochondrial DNA content, ATP content, citrate synthase activity, complexes I/III/V activity, AMPK phosphorylation, and increased PGC-1α acetylation. Since APN is an upstream activator of AMPK and APN plasma levels are significantly reduced in ob/ob mice, we further tested the hypothesis that reduced APN in ob/ob mice is causatively related to mitochondrial biogenesis impairment. One week of APN treatment of ob/ob mice activated AMPK, reduced PGC-1α acetylation, increased mitochondrial biogenesis, and attenuated mitochondrial disorders. In contrast, knocking out APN inhibited AMPK-PGC-1α signaling and impaired both mitochondrial biogenesis and function. The ob/ob mice exhibited lower survival rates and exacerbated myocardial injury after MI, when compared to controls. APN supplementation improved mitochondrial biogenesis and attenuated MI injury, an effect that was almost completely abrogated by the AMPK inhibitor compound C. In high glucose/high fat treated neonatal rat ventricular myocytes, siRNA-mediated knockdown of PGC-1α blocked gAd-enhanced mitochondrial biogenesis and function and attenuated protection against hypoxia/reoxygenation injury. In conclusion, hypoadiponectinemia impaired AMPK-PGC-1α signaling, resulting in dysfunctional mitochondrial biogenesis that constitutes a novel mechanism for rendering diabetic hearts more vulnerable to enhanced MI injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Diabetes Complications/enzymology , Mitochondria, Heart/enzymology , Mitochondrial Turnover , Myocardial Infarction/enzymology , Myocardium/enzymology , Signal Transduction , Trans-Activators/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Adiponectin/deficiency , Adiponectin/genetics , Animals , Animals, Newborn , Cells, Cultured , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Diabetes Complications/genetics , Diabetes Complications/pathology , Diabetes Complications/physiopathology , Diabetes Complications/prevention & control , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism , High Mobility Group Proteins/metabolism , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondria, Heart/pathology , Mitochondrial Swelling , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardium/pathology , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Trans-Activators/genetics , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL