Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
1.
J Neuroinflammation ; 18(1): 226, 2021 Oct 13.
Article En | MEDLINE | ID: mdl-34645465

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) is a severe anoxic brain injury that leads to premature mortality or long-term disabilities in infants. Neuroinflammation is a vital contributor to the pathogenic cascade post-HIE and a mediator to secondary neuronal death. As a plasma membrane G-protein-coupled receptor, GPR39, exhibits anti-inflammatory activity in several diseases. This study aimed to explore the neuroprotective function of GPR39 through inhibition of inflammation post-hypoxic-ischemic (HI) injury and to elaborate the contribution of sirtuin 1(SIRT1)/peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α)/nuclear factor, erythroid 2 like 2(Nrf2) in G-protein-coupled receptor 39 (GPR39)-mediated protection. METHODS: A total of 206 10-day-old Sprague Dawley rat pups were subjected to HIE or sham surgery. TC-G 1008 was administered intranasally at 1 h, 25 h, 49 h, and 73 h post-HIE induction. SIRT1 inhibitor EX527, GPR39 CRISPR, and PGC-1α CRISPR were administered to elucidate the underlying mechanisms. Brain infarct area, short-term and long-term neurobehavioral tests, Nissl staining, western blot, and immunofluorescence staining were performed post-HIE. RESULTS: The expression of GPR39 and pathway-related proteins, SIRT1, PGC-1α and Nrf2 were increased in a time-dependent manner, peaking at 24 h or 48-h post-HIE. Intranasal administration of TC-G 1008 reduced the percent infarcted area and improved short-term and long-term neurological deficits. Moreover, TC-G 1008 treatment significantly increased the expression of SIRT1, PGC-1α and Nrf2, but downregulated the expressions of IL-6, IL-1ß, and TNF-α. GPR39 CRISPR EX527 and PGC-1α CRISPR abolished GPR39's neuroprotective effects post-HIE. CONCLUSIONS: TC-G 1008 attenuated neuroinflammation in part via the SIRT1/PGC-1α/Nrf2 pathway in a neonatal rat model of HIE. TC-G 1008 may be a novel therapeutic target for treatment post-neonatal HIE injury.


Hypoxia-Ischemia, Brain/metabolism , NF-E2-Related Factor 2/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/biosynthesis , Sirtuin 1/biosynthesis , Sulfonamides/pharmacology , Animals , Animals, Newborn , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/therapeutic use
2.
Neurotox Res ; 39(5): 1393-1404, 2021 Oct.
Article En | MEDLINE | ID: mdl-34251648

SIRT1 is a deacetylase with multiple physiological functions by targeting histones and non-histone proteins. It has been shown that SIRT1 activation is involved in neuroprotection in Parkinson's disease (PD) models. In the present study, we provided direct evidences showing the neuroprotective roles of SIRT1 in dopaminergic neurons. Our data showed that increased expression of SIRT1 plays beneficial roles against MPP+ insults in SH-SY5Y cells and primary dopaminergic neurons, including increased cell viability, reduced LDH release, improved the mitochondrial membrane potential (MMP), and attenuated cell apoptosis. On the contrary, knockdown of SIRT1 further aggravated cell injuries induced by MPP+. Moreover, mutated SIRT1 without deacetylase activity (SIRT1 H363Y) failed to protect dopaminergic neurons from MPP+ injuries. Mechanistically, SIRT1 improved PGC-1α expression and mitochondrial biogenesis. Knockdown of PGC-1α almost completely abolished the neuroprotective roles of SIRT1 in SH-SY5Y cells. Collectively, our data indicate that SIRT1 has neuroprotective roles in dopaminergic neurons, which is dependent upon PGC-1α-mediated mitochondrial biogenesis. These findings suggest that SIRT1 may hold great therapeutic potentials for treating dopaminergic neuron loss associated disorders such as PD.


Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Parkinsonian Disorders/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Sirtuin 1/biosynthesis , 1-Methyl-4-phenylpyridinium/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Mitochondria/pathology , Neuroprotection/drug effects , Neuroprotection/physiology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/prevention & control , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Sirtuin 1/genetics
3.
Neurotox Res ; 39(4): 1274-1284, 2021 Aug.
Article En | MEDLINE | ID: mdl-33939098

Thimerosal (THIM) induces neurotoxic changes including neuronal death and releases apoptosis inducing factors from mitochondria to cytosol. THIM alters the expression level of factors involved in apoptosis. On the other hand, the anti-apoptotic effects of exercise have been reported. In this study, we aimed to discover the effect of three protocols of treadmill exercise on the expression level of mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), BCL-2-associated death (BAD), BCL-2-associated X (BAX), BCL-XL, and BCL-2 (a pro-survival BCL-2 protein) in the hippocampus of control and THIM-exposed rats. Male Wistar rats were used in this research. Real-time PCR was applied to assess genes expression. The results showed that THIM increased the expression of pro-apoptotic factors (BAD and BAX), decreased the expression of anti-apoptotic factors (BCL-2 and BCL-XL), and decreased the expression of factors involved in mitochondrial biogenesis (TFAM and PGC-1α). Treadmill exercise protocols reversed the effect of THIM on all genes. In addition, treadmill exercise protocols decreased the expression of BAD and BAX, increased the expression of BCL-2, and increased the expression of TFAM and PGC-1α in control rats. In conclusion, THIM induced a pro-apoptotic effect and disturbed mitochondrial biogenesis and stability, whereas treadmill exercise reversed these effects.


Exercise Test/methods , Hippocampus/drug effects , Physical Conditioning, Animal/physiology , Thimerosal/toxicity , Animals , Gene Expression , Hippocampus/metabolism , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Physical Conditioning, Animal/methods , Preservatives, Pharmaceutical/toxicity , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Transcription Factors/biosynthesis , Transcription Factors/genetics , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/genetics , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
4.
Biochem Pharmacol ; 188: 114559, 2021 06.
Article En | MEDLINE | ID: mdl-33872571

Retinoid X receptor beta (RXRß) has been poorly studied in atherosclerosis. The aim of the present study is to explore the function of RXRß in oxidized low density lipoprotein (ox-LDL)-induced inflammation in endothelial cells and the underlying mechanism. The protein expression of RXRß in the aorta of atherosclerotic mice was detected. A lentivirus vector for RXRß overexpression and RNA interference for RXRß downregulation were constructed and transfected into human aortic endothelial cells (HAECs). The results showed that RXRß protein expression was downregulated in aorta of high fat diet (HFD)-fed LDLr-/- mice and ox-LDL-treated HAECs. The ox-LDL-induced production of pro-inflammatory cytokines and activations of TLR9/NF-κB and NLRP3/caspase-1 inflammasome pathway were significantly decreased by RXRß overexpression but increased by RXRß knockdown in HAECs. The ox­LDL­induced mitochondrial damage indicated as the increased generation of mitochondrial ROS, decreased mitochondrial membrane potential and increased mitochondrial DNA release was abolished by RXRß overexpression but aggravated by RXRß knockdown. Treatment with mito-TEMPO significantly reduced the increased production of pro-inflammatory cytokines and activations of TLR9/NF-κB and NLRP3/caspase-1 inflammasome induced by RXRß knockdown in ox-LDL treated HAECs. Moreover, peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α) protein expression was reduced in HFD-fed LDLr-/- mice. RXRß could interact with PGC1α in HAECs. Ox-LDL-induced reduction of PGC1α was significantly inhibited by RXRß overexpression and aggravated by RXRß downregulation. Our further study showed that transfection of PGC1α siRNA abrogated the alleviative effects of RXRß overexpression on mitochondrial damage and inflammation in ox-LDL treated cells. The present study indicates that RXRß exerted protective effects against the ox-LDL-induced inflammation may through regulating PGC1α-dependent mitochondrial homeostasis.


DNA-Binding Proteins/biosynthesis , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Lipoproteins, LDL/toxicity , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Animals , Cell Line , DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Endothelial Cells/drug effects , Gene Expression Regulation , Homeostasis/drug effects , Homeostasis/physiology , Humans , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
5.
Inflammation ; 44(5): 1771-1781, 2021 Oct.
Article En | MEDLINE | ID: mdl-33847873

Cardiac dysfunction in severe sepsis is associated with increased mortality. However, the molecular mechanisms underlying septic heart dysfunction remain unclear. Expression of peroxisome proliferator-activated receptor-γ coactivator 1α (Pgc-1α), concentrations of inflammatory factors, and activation of the nuclear factor kappa-B (NF-κB) signaling pathway were examined in H9c2 cells after a 24-h lipopolysaccharide (LPS) stimulation period using qPCR, enzyme-linked immunosorbent assays (ELISAs), and western blots (WBs), respectively. Pgc-1α was overexpressed and suppressed in cells using a lentivirus vector and siRNA, respectively. The effects of Pgc-1α dysfunction on the release of inflammatory factors and apoptosis were analyzed. Pgc-1α expression was increased after LPS induction for 0.5 h and returned to the pre-induction level at 2 h. Levels of IL-1ß, IL-6, and TNF-α increase after LPS induction for 0.5 h and accumulated in the culture supernatants over time. The WBs revealed the highest Pgc-1α and phospho (p)-p65 protein levels after LPS induction for 0.5 h, followed by a decrease; moreover, the cleaved-caspase-3 level increased after LPS induction for 0.5 h and increased gradually thereafter. A functional analysis of Pgc-1α revealed that overexpression of this protein enhanced LPS-induced inflammatory factors and p-p65 levels and inhibited apoptosis during the early stage after LPS induction (0.5 and 4 h). In contrast, the inhibition of Pgc-1α expression inhibited the LPS expression-associated increases in inflammatory factors and p-p65 and promoted apoptosis. Pgc-1α promoted LPS-induced p65 phosphorylation and inflammatory factor release while inhibiting apoptosis.


Apoptosis/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Animals , Apoptosis/physiology , Cell Line , Phosphorylation/drug effects , Phosphorylation/physiology , Rats
6.
Neurotox Res ; 39(3): 872-885, 2021 Jun.
Article En | MEDLINE | ID: mdl-33025359

Inflammation plays an important role in depression pathology, making it a promising target for ameliorating depression-like behaviors. The peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional coactivator being able to constrain inflammatory events through NF-κB signaling. However, the role of PGC-1α in depression is not yet clear. This study was designed to investigate the role of PGC-1α in depression and explore the underlying mechanisms. Mice modeled with chronic unpredictable mild stimulation (CUMS) were explored for the relationship between depression-like behaviors and PGC-1α. Baicalin was used to evaluate the effect regulating PGC-1α. Furthermore, the anti-neuroinflammatory effect of baicalin was investigated both in BV2-SH-SY5Y co-culture system and in mice by LPS challenge. The role of PGC-1α in neuroinflammation was explored in cell co-culture systems under gene silencing conditions targeting NF-κB signaling. We found that the expression of PGC-1α was inhibited in the hippocampus of mice exposed to CUMS or LPS, while baicalin could increase the expression of PGC-1α and alleviate the depression-like behaviors. Furthermore, baicalin attenuated neuroinflammation in the hippocampus of mice and BV2-SH-SY5Y co-culture system by LPS challenge via regulating NF-κB signaling; however, knockdown of the PGC-1α could reverse the effect of baicalin on neuroinflammation and NF-κB signaling. Our results revealed a vital role for PGC-1α in attenuating neuroinflammation in depression, indicating that PGC-1α might be a therapeutic target for depression.


Depression/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Up-Regulation/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Line, Tumor , Depression/drug therapy , Depression/psychology , Dose-Response Relationship, Drug , Flavonoids/pharmacology , Flavonoids/therapeutic use , Hindlimb Suspension/adverse effects , Hindlimb Suspension/psychology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/psychology , Male , Mice , Mice, Inbred ICR , Up-Regulation/drug effects
7.
Neurosci Lett ; 744: 135598, 2021 01 23.
Article En | MEDLINE | ID: mdl-33373677

Amyloid-ß (Aß) is the core component of amyloid plaques of Alzheimer's disease (AD). Recent evidence has confirmed that Aß triggers neurodegeneration by dramatically suppressing vitamin D receptor (VDR) expression. Thus far, the onset mechanisms and means of preventing AD are largely unknown. Perioxisome proliferator-activated receptor-γ coactivator (PGC-1α), as a transcriptional coactivator of VDR could protect cells against oxidative stress. Thus, upregulation of PGC-1α is a candidate therapeutic strategy for AD. To investigate the effect of PGC-1α in AD, and to illuminate the precise involvement of VDR in the neuroprotective strategy, the varies of molecular of PGC-1α and VDR were studied in APP/PS-1 double transgenic (2xTg-AD) mice at 6 months of age, significant reduction in the expression of PGC-1α and VDR was found in their hippocampus and the cortex. Besides, a specific mouse line, Dlx5/6-Cre:PGC-1αfl/fl in which the PGC-1α deficiency was limited to the hippocampus and the cortex, was used to study the target intervention of PGC-1α, decreased expression of VDR and increased oxidative damage were observed in AD-related brain regions by PGC-1α deficiency. To explore the function and therapeutic strategy of PGC-1α in AD, an adeno-associated virus (AAV) was used to induce PGC-1α overexpressed in the hippocampus of 2xTg-AD mice. Overexpressed PGC-1α results in a remarkable increase in the levels of VDR associated with a significant reduction in the expression of Aß plaques and of 8-oxo-dG in 2xTg-AD mice. These data may have ramifications for neuroprotective strategies targeting overexpression of PGC-1α in Alzheimer's disease.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Receptors, Calcitriol/biosynthesis , Alzheimer Disease/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Animals , Gene Expression , Hippocampus/metabolism , Mice , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, Calcitriol/genetics
8.
Inflammation ; 43(5): 1806-1820, 2020 Oct.
Article En | MEDLINE | ID: mdl-32529514

Mitochondrial damage is considered one of the main pathogenetic mechanisms in septic cardiomyopathy. Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) is critical for maintaining energy homeostasis in different organs and in various physiological and pathological states. It is also a key regulator gene in mitochondrial metabolism. In this study, we investigated whether regulation of the PGC-1α gene had protective effects on septic cardiomyopathy. We developed a rat model of septic cardiomyopathy. H9c2 myocardiocytes were treated with lipopolysaccharide (LPS) and PGC-1α expression measured. PGC-1α-overexpressing lentivirus was used to transfect H9c2 cells. ZLN005 was used to activate PGC-1α. The effect of the inhibition of PGC-1α expression on myocardial cell injury and its underlying mechanisms were also explored. Cell viability was measured by CCK-8 assay. Mitochondrial damage was determined by measuring cellular ATP, reactive oxygen species, and the mitochondrial membrane potential. An apoptosis analysis kit was used to measure cellular apoptosis. Mitochondrial DNA was extracted and real-time PCR performed. LC3B, mitochondrial transcription factor A (TFA), P62, Bcl2, and Bax were determined by immunofluorescence. LC3B, TFA, P62, Parkin, PTEN-induced putative kinase 1, and PGC-1α proteins were determined by Western blotting. We found mitochondrial damage and apoptotic cells in the myocardial tissue of rats with septic cardiomyopathy and in LPS-treated cardiomyocytes. PGC-1α expression was decreased in the late phase of septic cardiomyopathy and in LPS-treated cardiomyocytes. PGC-1α activation by ZLN005 and PGC-1α overexpression reduced apoptosis in myocardiocytes after LPS incubation. PGC-1α gene overexpression alleviated LPS-induced cardiomyocyte mitochondrial damage by activating mitochondrial biogenesis and autophagy functions. Our study indicated that mitochondrial damage and apoptosis occurred in septic cardiomyopathy and LPS-treated cardiomyocytes. The low expression level of PGC-1α protein may have contributed to this damage. By activating the expression of PGC-1α, apoptosis was reduced in cardiomyocytes. The underlying mechanism may be that PGC-1α can activate mitochondrial biogenesis and autophagy functions, reducing mitochondrial damage and thereby reducing apoptosis.


Apoptosis/drug effects , Lipopolysaccharides/toxicity , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Animals , Apoptosis/physiology , Dose-Response Relationship, Drug , Gene Expression , Male , Mitochondria/drug effects , Mitochondria/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Rats , Rats, Wistar , Sepsis/chemically induced , Sepsis/metabolism , Sepsis/pathology
9.
Life Sci ; 254: 117762, 2020 Aug 01.
Article En | MEDLINE | ID: mdl-32437795

AIMS: Patients with nonalcoholic fatty liver disease (NAFLD) have less tolerance to ischemia-reperfusion injury (IRI) of the liver than those with the healthy liver; hence have a higher incidence of severe complications after surgery. This study aimed to investigate the dynamics of the liver and mitochondrial damage and the impact of TLR4 knockout (TLR4KO) on Mfn2 expression in the composite model of NAFLD and IRI. MAIN METHODS: We performed high-fat diet (HFD) feeding and ischemia reperfusion (IR) on wild type (WT) and TLR4 knockout TLR4KO mice. KEY FINDINGS: The degree of structural and functional injuries to the liver and mitochondria (NAFLD and IRI) is greater than that caused by a single factor (NAFLD or IRI) or a simple superposition of both. The IL-6 and TNF-α expressions were significantly suppressed (P < .05), while PGC-1α and Mfn2 expressions were up-regulated considerably (P < .05) after TLR4KO. Furthermore, mitochondrial fusion increased, while ATP consumption and ROS production decreased significantly after TLR4KO (P < .05). The degree of reduction of compound injury by TLR4KO is more significant than the reduction degree of single factor injury. Also, TNF-α and IL-6 levels can be used predictive markers for mitochondrial damage and liver tolerance to NAFLD and IRI. SIGNIFICANCE: TLR4KO upregulates the expression of Mfn2 and PGC-1α in the composite model of NAFLD and IRI. This pathway may be related to IL-6 and TNF-α. This evidence provides theoretical and experimental basis for the subsequent Toll-like receptor 4 (TLR4) receptor targeted therapy.


GTP Phosphohydrolases/biosynthesis , Liver/blood supply , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Reperfusion Injury/metabolism , Toll-Like Receptor 4/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Liver/metabolism , Liver/pathology , Liver Transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reperfusion , Reperfusion Injury/pathology , Signal Transduction/physiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
10.
Aging (Albany NY) ; 12(8): 7334-7349, 2020 04 26.
Article En | MEDLINE | ID: mdl-32335547

The pathological effects of obesity are often severe in aging condition. Although exercise training is found to be advantageous, the intensity of exercise performed is limited in aging condition. Therefore in this study we assessed the effect of a combined treatment regimen with a short-peptide IF isolated from alcalase potato-protein hydrolysates and a moderate exercise training for 15 weeks in a 6 month old HFD induced obese senescence accelerated mouse-prone 8 (SAMP8) mice model. Animals were divided into 6 groups (n=6) (C:Control+BSA); (HF:HFD+BSA); (EX:Control+ BSA+Exercise); (HF+IF:HFD+ IF); (HF+EX:HFD+Exercise); (HF+EX+IF:HFD+Exercise+IF). A moderate incremental swimming exercise training was provided for 6 weeks and after 3 weeks of exercise, IF was orally administered (1 mg/kg body Weight). The results show that combined administration of IF and exercise provides a better protection to aging animals by reducing body weight and regulated tissue damage. IF intake and exercise training provided protection against cardiac hypertrophy and maintains the tissue homeostasis in the heart and liver sections. Interestingly, IF and exercise training showed an effective upregulation in pAMPK/ SIRT1/ PGC-1α/ pFOXO3 mechanism of cellular longevity. Therefore, exercise training with IF intake is a possible strategy for anti-obesity benefits and superior cardiac and hepatic protection in aging condition.


Aging , Forkhead Box Protein O3/genetics , Gene Expression Regulation , Obesity/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RNA/genetics , Sirtuin 1/genetics , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Forkhead Box Protein O3/biosynthesis , Male , Mice , Obesity/metabolism , Obesity/therapy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Physical Conditioning, Animal , Sirtuin 1/biosynthesis
11.
Cell Death Dis ; 11(4): 226, 2020 04 08.
Article En | MEDLINE | ID: mdl-32269221

An imbalance in mitochondrial dynamics induced by oxidative stress may lead to hepatocyte epithelial mesenchymal transition (EMT) and liver fibrosis. However, the underlying molecular mechanisms have not been fully elucidated. This study investigated the role of mitochondrial dynamics in hepatocyte EMT and liver fibrosis using an in vitro human (L-02 cells, hepatic cell line) and an in vivo mouse model of liver fibrosis. Findings showed that oxidative stress-induced mitochondrial DNA damage was associated with abnormal mitochondrial fission and hepatocyte EMT. The reactive oxygen species (ROS) scavengers apocynin and mito-tempo effectively attenuated carbon tetrachloride (CCl4)-induced abnormal mitochondrial fission and liver fibrosis. Restoring mitochondrial biogenesis attenuated hepatocyte EMT. Oxidative stress-induced abnormal hepatocyte mitochondrial fission events by a mechanism that involved the down regulation of PGC-1α. PGC-1α knockout mice challenged with CCl4 had increased abnormal mitochondrial fission and more severe liver fibrosis than wild type mice. These results indicate that PGC-1α has a protective role in oxidative stress-induced-hepatocyte EMT and liver fibrosis.


Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Transcription Factors/biosynthesis , Animals , Cell Line , Disease Models, Animal , Epithelial-Mesenchymal Transition , Hepatocytes/pathology , Humans , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Dynamics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Transfection
12.
Aging (Albany NY) ; 12(8): 7411-7430, 2020 04 28.
Article En | MEDLINE | ID: mdl-32343674

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.


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
13.
Aging (Albany NY) ; 12(8): 7299-7312, 2020 04 19.
Article En | MEDLINE | ID: mdl-32305957

Cardiac ischemia/reperfusion injury is associated with reduced mitochondrial turnover and regeneration. There is currently no effective approach to stimulate mitochondrial biogenesis in the reperfused myocardium. In this study, we investigated whether melatonin could increase mitochondrial biogenesis and thus promote mitochondrial homeostasis in cardiomyocytes. Cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) injury with or without melatonin treatment, and various mitochondrial functions were measured. H/R injury repressed mitochondrial biogenesis in cardiomyocytes, whereas melatonin treatment restored mitochondrial biogenesis through the 5' adenosine monophosphate-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1α) pathway. Melatonin enhanced mitochondrial metabolism, inhibited mitochondrial oxidative stress, induced mitochondrial fusion and prevented mitochondrial apoptosis in cardiomyocytes subjected to H/R injury. The melatonin-induced improvement in mitochondrial biogenesis was associated with increased cardiomyocyte survival during H/R injury. On the other hand, silencing of PGC1α attenuated the protective effects of melatonin on cardiomyocyte viability, thereby impairing mitochondrial bioenergetics, disrupting the mitochondrial morphology, and activating mitochondrial apoptosis. Thus, H/R injury suppressed mitochondrial biogenesis, while melatonin activated the AMPK/PGC1α pathway and restored mitochondrial biogenesis, ultimately protecting the reperfused heart.


AMP-Activated Protein Kinases/genetics , Gene Expression Regulation , Melatonin/pharmacology , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , AMP-Activated Protein Kinases/biosynthesis , Animals , Antioxidants/pharmacology , Disease Models, Animal , Male , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , RNA/genetics , RNA/metabolism , RNA-Binding Proteins , Rats
14.
Anticancer Res ; 40(2): 653-664, 2020 Feb.
Article En | MEDLINE | ID: mdl-32014906

BACKGROUND/AIM: Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is a master regulator of mitochondrial biogenesis and metabolism. We investigated the effect of PGC-1α knockdown in the human colorectal cancer cell line SW620, which highly expresses PGC-1α. MATERIALS AND METHODS: We established the PGC-1α shRNA-silenced SW620 stable cell line (PGC-1α shRNA-SW620 cells) and examined cell proliferation by cell counts and carboxyfluorescein succinimidyl ester (CFSE) staining, migration by wound-healing and transwell migration assay, and invasion by transwell assays. RESULTS: PGC-1α knockdown inhibited cell proliferation, migration, and invasion in SW620 cells. Western blot analysis showed that p-AKT, p-GSK-3ß, ß-catenin, N-cadherin and vimentin expression were all reduced, but E-cadherin had increased expression in PGC-1α shRNA-SW620 cells. We also examined cell proliferation, migration, invasion and the expression of p-AKT, p-GSK-3ß, ß-catenin, N-cadherin, vimentin, and E-cadherin in PGC-1α overexpressing SW480 cells (a low PGC-1α expressing line). We observed a complete reversal of the results seen in the knockdown. CONCLUSION: PGC-1α might regulate cell proliferation and invasion via AKT/GSK-3ß/ß-catenin pathway in SW620 and SW480 cells.


Glycogen Synthase Kinase 3 beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Knockdown Techniques , Humans , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Signal Transduction , Transfection
15.
Neurochem Res ; 45(5): 1086-1096, 2020 May.
Article En | MEDLINE | ID: mdl-32060774

Blood brain barrier (BBB) disruption plays an important role in brain injury after acute kidney injury (AKI). However, its underlying mechanisms remain poorly understood. Recent evidence has revealed that proper mitochondrial function is essential for BBB permeability. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a key factor in mitochondrial biogenesis and function. This study was designed to investigate the role of PGC-1α in BBB injury after AKI and its related mechanisms. Mice received recombinant adenovirus encoding murine PGC-1α (100 µl, 1.0 × 109PFU/ml) or vehicle 5 days before renal I/R or sham operation. Twenty-four hours after the operation, brain, kidney and serum samples were collected for assessments. We found that mice suffering from renal I/R injury showed decreased PGC-1α levels in both the kidney and BBB. PGC-1α transfection resulted in increased PGC-1α level and mitochondrial transcripts in BBB at 24 h after AKI. PGC-1α transfection improved renal function, systemic inflammation and BBB permeability via both the paracellular and transcellular pathways. Further study suggested that PGC-1α overexpression elevated fatty acid oxidation related gene expression. Our findings demonstrate the importance of PGC-1α in AKI-induced BBB injury and suggest that it could be a therapeutic target for BBB repair via the regulation of mitochondrial function.


Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Blood-Brain Barrier/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Acute Kidney Injury/pathology , Animals , Blood-Brain Barrier/pathology , Cytoprotection/physiology , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/pathology , Random Allocation
16.
J Clin Invest ; 130(2): 853-862, 2020 02 03.
Article En | MEDLINE | ID: mdl-31929186

Oncogene-targeted and immune checkpoint therapies have revolutionized the clinical management of malignant melanoma and now offer hope to patients with advanced disease. Intimately connected to patients' overall clinical risk is whether the initial primary melanoma lesion will metastasize and cause advanced disease, but underlying mechanisms are not entirely understood. A subset of melanomas display heightened peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) expression that maintains cell survival cues by promoting mitochondrial function, but also suppresses metastatic spread. Here, we show that PGC1α expression in melanoma cells was silenced by chromatin modifications that involve promoter H3K27 trimethylation. Pharmacological EZH2 inhibition diminished H3K27me3 histone markers, increased PGC1α expression, and functionally suppressed invasion within PGC1α-silenced melanoma cells. Mechanistically, PGC1α silencing activated transcription factor 12 (TCF12), to increase expression of WNT5A, which in turn stabilized YAP protein levels to promote melanoma migration and metastasis. Accordingly, inhibition of components of this transcription-signaling axis, including TCF12, WNT5A, or YAP, blocked melanoma migration in vitro and metastasis in vivo. These results indicate that epigenetic control of melanoma metastasis involved altered expression of PGC1α and an association with the inherent metabolic state of the tumor.


Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Histones/metabolism , Melanoma, Experimental/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Transcription Factors/metabolism , Wnt-5a Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Line, Tumor , HEK293 Cells , Histones/genetics , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Transcription Factors/genetics , Wnt-5a Protein/genetics , YAP-Signaling Proteins
17.
J Clin Invest ; 130(5): 2319-2331, 2020 05 01.
Article En | MEDLINE | ID: mdl-31961829

BACKGROUNDBeige adipose tissue is associated with improved glucose homeostasis in mice. Adipose tissue contains ß3-adrenergic receptors (ß3-ARs), and this study was intended to determine whether the treatment of obese, insulin-resistant humans with the ß3-AR agonist mirabegron, which stimulates beige adipose formation in subcutaneous white adipose tissue (SC WAT), would induce other beneficial changes in fat and muscle and improve metabolic homeostasis.METHODSBefore and after ß3-AR agonist treatment, oral glucose tolerance tests and euglycemic clamps were performed, and histochemical analysis and gene expression profiling were performed on fat and muscle biopsies. PET-CT scans quantified brown adipose tissue volume and activity, and we conducted in vitro studies with primary cultures of differentiated human adipocytes and muscle.RESULTSThe clinical effects of mirabegron treatment included improved oral glucose tolerance (P < 0.01), reduced hemoglobin A1c levels (P = 0.01), and improved insulin sensitivity (P = 0.03) and ß cell function (P = 0.01). In SC WAT, mirabegron treatment stimulated lipolysis, reduced fibrotic gene expression, and increased alternatively activated macrophages. Subjects with the most SC WAT beiging showed the greatest improvement in ß cell function. In skeletal muscle, mirabegron reduced triglycerides, increased the expression of PPARγ coactivator 1 α (PGC1A) (P < 0.05), and increased type I fibers (P < 0.01). Conditioned media from adipocytes treated with mirabegron stimulated muscle fiber PGC1A expression in vitro (P < 0.001).CONCLUSIONMirabegron treatment substantially improved multiple measures of glucose homeostasis in obese, insulin-resistant humans. Since ß cells and skeletal muscle do not express ß3-ARs, these data suggest that the beiging of SC WAT by mirabegron reduces adipose tissue dysfunction, which enhances muscle oxidative capacity and improves ß cell function.TRIAL REGISTRATIONClinicaltrials.gov NCT02919176.FUNDINGNIH: DK112282, P30GM127211, DK 71349, and Clinical and Translational science Awards (CTSA) grant UL1TR001998.


Acetanilides/administration & dosage , Adrenergic beta-3 Receptor Agonists/administration & dosage , Blood Glucose/metabolism , Obesity/blood , Thiazoles/administration & dosage , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adult , Aged , Biopsy , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/drug therapy , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis
18.
Metab Brain Dis ; 35(1): 183-192, 2020 01.
Article En | MEDLINE | ID: mdl-31773435

Cholestasis means impaired bile synthesis or secretion. In fact, it is a bile flow reduction following Bile Duct Ligation (BDL). Cholestasis has a main role in necrosis and apoptosis. Apoptosis is a form of programmed cell death that has intrinsic and extrinsic pathways. The intrinsic pathway is mediated by Bcl-2 (B cell lymphoma-2) proteins which integrate death and survival signals. Bcl-2 has anti-apoptotic and Bax has pro-apoptotic effects. Also, striatum is one of the brain regions that has high expressions of Bcl-2 proteins. Moreover, Tfam and Pgc-1α are involved in mitochondrial biogenesis. On the other hand, NeuroAid, is a drug that has neuroprotective and anti-apoptosis effects. In this study, using quantitative PCR, we measured the expression of all these genes in the striatum of male rats following BDL and NeuroAid administration. Results showed, BDL increased the expression of Bax and Tfam and decreased the expression of Bcl-2. NeuroAid restored the effect of BDL on the expression of Bax, while did not alter the effect of BDL on Bcl-2. In addition, it increased the expression of Tfam that was previously elevated by BDL and raised the expression of Tfam in normal rats. Both BDL and NeuroAid, had no effect on Pgc-1α. In conclusion, cholestasis increased the expression of Bax and decreased the expression of Bcl-2, and this effect may have related to enhanced susceptibility of mitochondrial pathways following oxidative stress. Tfam expression was increased following cholestasis and this effect may have related to cellular compensatory mechanisms against high accumulation of free radicals or mitochondrial biogenesis failure. Furthermore, NeuroAid may play a role against apoptosis and can be used to increase mitochondrial biogenesis.


Cholestasis/metabolism , Corpus Striatum/metabolism , Drugs, Chinese Herbal/therapeutic use , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transcription Factors/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cholestasis/drug therapy , Cholestasis/genetics , Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Gene Expression , Male , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Transcription Factors/genetics , Treatment Outcome , bcl-2-Associated X Protein/genetics
19.
Neural Plast ; 2019: 8798069, 2019.
Article En | MEDLINE | ID: mdl-31885537

Neuronal apoptosis induced by oxidative stress is a major pathological process that occurs after cerebral ischemia-reperfusion. Calycosin-7-O-ß-D-glucoside (CG) is a representative component of isoflavones in Radix Astragali (RA). Previous studies have shown that CG has potential neuroprotective effects. However, whether CG alleviates neuronal apoptosis through antioxidant stress after ischemia-reperfusion remains unknown. To investigate the positive effects of CG on oxidative stress and apoptosis of neurons, we simulated the ischemia-reperfusion process in vitro using an immortalized hippocampal neuron cell line (HT22) and oxygen-glucose deprivation/reperfusion (OGD/R) model. CG significantly improved cell viability and reduced oxidative stress and neuronal apoptosis. In addition, CG treatment upregulated the expression of SIRT1, FOXO1, PGC-1α, and Bcl-2 and downregulated the expression of Bax. In summary, our findings indicate that CG alleviates OGD/R-induced damage via the SIRT1/FOXO1/PGC-1α signaling pathway. Thus, CG maybe a promising therapeutic candidate for brain injury associated with ischemic stroke.


Apoptosis/physiology , Forkhead Box Protein O1/biosynthesis , Glucose/deficiency , Glucosides/pharmacology , Isoflavones/pharmacology , Oxidative Stress/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Sirtuin 1/biosynthesis , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Sci Signal ; 12(606)2019 11 05.
Article En | MEDLINE | ID: mdl-31690633

TFEB is a basic helix-loop-helix transcription factor that confers protection against metabolic diseases such as atherosclerosis by targeting a network of genes involved in autophagy-lysosomal biogenesis and lipid catabolism. In this study, we sought to characterize the role of TFEB in adipocyte and adipose tissue physiology and evaluate the therapeutic potential of adipocyte-specific TFEB overexpression in obesity. We demonstrated that mice with adipocyte-specific TFEB overexpression (Adipo-TFEB) were protected from diet-induced obesity, insulin resistance, and metabolic sequelae. Adipo-TFEB mice were lean primarily through increased metabolic rate, suggesting a role for adipose tissue browning and enhanced nonshivering thermogenesis in fat. Transcriptional characterization revealed that TFEB targeted genes involved in adipose tissue browning rather than those involved in autophagy. One such gene encoded PGC-1α, an established target of TFEB that promotes adipocyte browning. To dissect the role of PGC-1α in mediating the downstream effects of TFEB overexpression, we generated mice with adipocyte-specific PGC-1α deficiency and TFEB overexpression. Without PGC-1α, the ability of TFEB overexpression to brown adipose tissue and to elicit beneficial metabolic effects was blunted. Overall, these data implicate TFEB as a PGC-1α-dependent regulator of adipocyte browning and suggest its therapeutic potential in treating metabolic disease.


Adipocytes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Metabolic Diseases/prevention & control , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Adipocytes/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Mice, Transgenic , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
...