Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Metabolism ; 128: 154959, 2022 03.
Article in English | MEDLINE | ID: mdl-34953916

ABSTRACT

BACKGROUND: Mitochondrial biogenesis and dysfunction are associated with renal tubular epithelial cell injury and the pathophysiological development of diabetic nephropathy (DN). Adiponectin (APN) is a plasma hormone protein specifically secreted by adipocytes. In the present study, we studied the effects of APN on mitochondrial biogenesis and function in renal tubular epithelial cells and examined the mechanisms underlying its actions. MATERIALS: A rat model of type 2 diabetes mellitus (T2DM) was established using streptozotocin (STZ), and an NRK-52E culture model exposed to high glucose was also used. We found that APN treatment alleviated kidney histopathological injury in T2DM rats, reduced fasting blood glucose (FBG) and postprandial blood glucose (PBG) levels, maintained stable animal weight, promoted cell viability, inhibited apoptosis and the formation of autophagosomes, and also increased mitochondrial mass, mitochondrial DNA (mtDNA) content and mitochondrial membrane potential (MMP) in vivo and in vitro. RESULTS: We found that the expression of AdipoR1/CREB/PGC-1α/TFAM pathway proteins and respiratory chain complex subunits CO1, CO2, CO3, ATP6 and ATP8 were significantly increased after APN treatment. We also found that inhibition of cAMP response element binding protein (CREB) weakened the effects of APN in NRK-52E cells treated with high glucose. Coimmunoprecipitation experiments showed that AdipoR1 interacted with CREB. CONCLUSION: APN promoted mitochondrial biogenesis and function in renal tubular epithelial cells by regulating the AdipoR1/CREB/PGC-1α/TFAM pathway. APN has the potential to serve as an effective drug for the treatment of DN.


Subject(s)
Adiponectin/pharmacology , Diabetic Nephropathies/drug therapy , Kidney Tubules/drug effects , Mitochondria/drug effects , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/physiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Epithelial Cells/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/physiology , Male , Mitochondria/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Streptozocin , Transcription Factors/physiology
2.
Bull Exp Biol Med ; 172(2): 180-186, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34853966

ABSTRACT

The study demonstrated the crucial role of Sirt1 gene in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) related to the influence of Sirt1 on oxidative stress and glycolipid metabolism. The 16-week-old genetically diabetic db/db mice were characterized with downregulated expression of Sirt1 in the liver accompanied by hepatomegaly and a larger size of fat vacuoles in hepatocytes. In db/m mice, silencing Sirt1 gene induced hepatic steatosis and significantly increased serum AST. Additionally, the levels of triglycerides in blood and liver of these mice were elevated. However, all pathological changes in the liver of Sirt1-knockdown db/m mice were less pronounced than in 16-week-old db/db mice. Further experiments showed that oxidative stress and PGC-1α-mediated mitochondrial dysfunction are implicated in pathological changes of lipid metabolism in T2DM-induced NAFLD provoked by Sirt1 silencing. This study showed that down-regulation of Sirt1 expression plays the key role in pathological processes developed during T2DM-induced abnormalities of lipid metabolism in the liver. Thus, up-regulation of Sirt1 expression seems to be a promising strategy in early prevention of T2DM-induced NAFLD.


Subject(s)
Lipid Metabolism/genetics , Liver/metabolism , Sirtuin 1/genetics , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/physiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Signal Transduction/genetics
3.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34465622

ABSTRACT

Plasticity of cells, tissues, and organs is controlled by the coordinated transcription of biological programs. However, the mechanisms orchestrating such context-specific transcriptional networks mediated by the dynamic interplay of transcription factors and coregulators are poorly understood. The peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is a prototypical master regulator of adaptive transcription in various cell types. We now uncovered a central function of the C-terminal domain of PGC-1α to bind RNAs and assemble multiprotein complexes including proteins that control gene transcription and RNA processing. These interactions are important for PGC-1α recruitment to chromatin in transcriptionally active liquid-like nuclear condensates. Notably, such a compartmentalization of active transcription mediated by liquid-liquid phase separation was observed in mouse and human skeletal muscle, revealing a mechanism by which PGC-1α regulates complex transcriptional networks. These findings provide a broad conceptual framework for context-dependent transcriptional control of phenotypic adaptations in metabolically active tissues.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , RNA/metabolism , Animals , Cell Line , Chromatin/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Domains , Protein Interaction Domains and Motifs
4.
Mol Neurobiol ; 58(11): 5890-5906, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34415486

ABSTRACT

Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic ß-amyloid peptide (Aß). Aß induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aß to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aß deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aß, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Exercise , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Autophagy , Blood-Brain Barrier , Brain-Derived Neurotrophic Factor/physiology , Carrier Proteins/metabolism , Disease Models, Animal , Exercise/physiology , Fibronectins/physiology , Glymphatic System , Humans , Membrane Microdomains/physiology , Mice , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control , Neuroinflammatory Diseases/physiopathology , Peptide Hydrolases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Physical Conditioning, Animal , Proteolysis , Signal Transduction/physiology , Sirtuin 1/physiology , Unfolded Protein Response/physiology
5.
Cardiovasc Ther ; 2021: 6615400, 2021.
Article in English | MEDLINE | ID: mdl-34221126

ABSTRACT

Abdominal aortic aneurysm (AAA) is defined as a progressive segmental dilation of the abdominal aorta and is associated with high mortality. The characterized features of AAA indicate several underlying mechanisms of AAA formation and progression, including reactive oxygen species production, inflammation, and atherosclerosis. Mitochondrial functions are critical for determining cell fate, and mitochondrial dynamics, especially selective mitochondrial autophagy, which is termed as mitophagy, has emerged as an important player in the pathogenesis of several cardiovascular diseases. The PARKIN/PARIS/PGC1α pathway is associated with AAA formation and has been proposed to play a role in mitochondrial dynamics mediated by the PINK/PARKIN pathway in the pathogenesis underlying AAA. This review is aimed at deepening our understanding of AAA formation and progression, which is vital for the development of potential medical therapies for AAA.


Subject(s)
Aortic Aneurysm, Abdominal/physiopathology , Aortic Dissection/physiopathology , Mitochondrial Dynamics/physiology , Animals , Atherosclerosis/metabolism , Disease Models, Animal , Humans , Inflammation/metabolism , Male , Mitochondria/metabolism , Mitochondria/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/physiology
6.
Int J Mol Sci ; 22(13)2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34281248

ABSTRACT

Age-related macular degeneration (AMD), the main cause of vision loss in the elderly, is associated with oxidation in the retina cells promoting telomere attrition. Activation of telomerase was reported to improve macular functions in AMD patients. The catalytic subunit of human telomerase (hTERT) may directly interact with proteins important for senescence, DNA damage response, and autophagy, which are impaired in AMD. hTERT interaction with mTORC1 (mTOR (mechanistic target of rapamycin) complex 1) and PINK1 (PTEN-induced kinase 1) activates macroautophagy and mitophagy, respectively, and removes cellular debris accumulated over AMD progression. Ectopic expression of telomerase in retinal pigment epithelium (RPE) cells lengthened telomeres, reduced senescence, and extended their lifespan. These effects provide evidence for the potential of telomerase in AMD therapy. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) may be involved in AMD pathogenesis through decreasing oxidative stress and senescence, regulation of vascular endothelial growth factor (VEGF), and improving autophagy. PGC-1α and TERT form an inhibitory positive feedback loop. In conclusion, telomerase activation and its ectopic expression in RPE cells, as well as controlled clinical trials on the effects of telomerase activation in AMD patients, are justified and should be assisted by PGC-1α modulators to increase the therapeutic potential of telomerase in AMD.


Subject(s)
Macular Degeneration/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Telomerase/metabolism , Aging/metabolism , Autophagy/physiology , DNA Damage/physiology , DNA Repair/physiology , Humans , Macular Degeneration/physiopathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Phenotype , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Signal Transduction , Telomerase/physiology , Telomere/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Metab Brain Dis ; 36(7): 1501-1521, 2021 10.
Article in English | MEDLINE | ID: mdl-33988807

ABSTRACT

As progressive, chronic, incurable and common reasons for disability and death, neurodegenerative diseases (NDDs) are significant threats to human health. Besides, the increasing prevalence of neuronal gradual degeneration and death during NDDs has made them a global concern. Since yet, no effective treatment has been developed to combat multiple dysregulated pathways/mediators and related complications in NDDs. Therefore, there is an urgent need to create influential and multi-target factors to combat neuronal damages. Accordingly, the plant kingdom has drawn a bright future. Among natural entities, flavonoids are considered a rich source of drug discovery and development with potential biological and medicinal activities. Growing studies have reported multiple dysregulated pathways in NDDs, which among those mediator AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) play critical roles. In this line, critical role of flavonoids in the upregulation of AMPK/PGC-1α pathway seems to pave the road in the treatment of Alzheimer's disease (AD), Parkinson's disease (PD), aging, central nervous system (brain/spinal cord) damages, stroke, and other NDDs. In the present study, the regulatory role of flavonoids in managing various NDDs has been shown to pass through AMPK/PGC-1α signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/physiology , Flavonoids/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Aging/drug effects , Alzheimer Disease/drug therapy , Animals , Brain Injuries, Traumatic/drug therapy , Flavonoids/therapeutic use , Humans , Ischemic Stroke/drug therapy , Memory Disorders/drug therapy , Signal Transduction/drug effects
8.
Int J Mol Sci ; 22(7)2021 Mar 28.
Article in English | MEDLINE | ID: mdl-33800548

ABSTRACT

Parkinson's disease is one of the most common neurodegenerative disorders worldwide, characterized by a progressive loss of dopaminergic neurons mainly localized in the substantia nigra pars compacta. In recent years, the detailed analyses of both genetic and idiopathic forms of the disease have led to a better understanding of the molecular and cellular pathways involved in PD, pointing to the centrality of mitochondrial dysfunctions in the pathogenic process. Failure of mitochondrial quality control is now considered a hallmark of the disease. The peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1) family acts as a master regulator of mitochondrial biogenesis. Therefore, keeping PGC-1 level in a proper range is fundamental to guarantee functional neurons. Here we review the major findings that tightly bond PD and PGC-1s, raising important points that might lead to future investigations.


Subject(s)
Neurons/metabolism , Parkinson Disease/metabolism , Pars Compacta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Animals , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Dopaminergic Neurons/metabolism , Genome-Wide Association Study , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Organelle Biogenesis , Oxidative Stress , Phosphorylation , Protein Deglycase DJ-1/metabolism , Protein Kinases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins/metabolism , alpha-Synuclein/metabolism
9.
Int J Mol Sci ; 22(9)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33925111

ABSTRACT

Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) is a protein that promotes transcription of numerous genes, particularly those responsible for the regulation of mitochondrial biogenesis. Evidence for a key role of PGC1α in bone metabolism is very recent. In vivo studies showed that PGC1α deletion negatively affects cortical thickness, trabecular organization and resistance to flexion, resulting in increased risk of fracture. Furthermore, in a mouse model of bone disease, PGC1α activation stimulates osteoblastic gene expression and inhibits atrogene transcription. PGC1α overexpression positively affects the activity of Sirtuin 3, a mitochondrial nicotinammide adenina dinucleotide (NAD)-dependent deacetylase, on osteoblastic differentiation. In vitro, PGC1α overexpression prevents the reduction of mitochondrial density, membrane potential and alkaline phosphatase activity caused by Sirtuin 3 knockdown in osteoblasts. Moreover, PGC1α influences the commitment of skeletal stem cells towards an osteogenic lineage, while negatively affects marrow adipose tissue accumulation. In this review, we will focus on recent findings about PGC1α action on bone metabolism, in vivo and in vitro, and in pathologies that cause bone loss, such as osteoporosis and type 2 diabetes.


Subject(s)
Bone and Bones/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Animals , Bone and Bones/physiology , Cell Differentiation/genetics , Humans , Mice , Mitochondria/metabolism , Organelle Biogenesis , Osteoblasts/metabolism , Osteocytes/metabolism , Osteogenesis , Signal Transduction , Sirtuin 3/metabolism , Transcription Factors/metabolism
10.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925372

ABSTRACT

Mitochondria play vital roles, including ATP generation, regulation of cellular metabolism, and cell survival. Mitochondria contain the majority of cellular nicotinamide adenine dinucleotide (NAD+), which an essential cofactor that regulates metabolic function. A decrease in both mitochondria biogenesis and NAD+ is a characteristic of metabolic diseases, and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) orchestrates mitochondrial biogenesis and is involved in mitochondrial NAD+ pool. Here we discuss how PGC-1α is involved in the NAD+ synthesis pathway and metabolism, as well as the strategy for increasing the NAD+ pool in the metabolic disease state.


Subject(s)
Metabolic Diseases/metabolism , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Humans , Metabolic Diseases/physiopathology , Mitochondria/physiology , NAD/biosynthesis , NAD/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Signal Transduction/physiology , Sirtuins/metabolism , Transcription Factors/metabolism
11.
Int J Mol Sci ; 22(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567500

ABSTRACT

Age-related macular degeneration (AMD) is the most prevalent form of irreversible blindness worldwide in the elderly population. In our previous studies, we found that deficiencies in the nuclear factor, erythroid 2 like 2 (NFE2L2) and peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) genes caused AMD-like pathological phenotypes in mice. In the present work, we show hijacked epithelial-mesenchymal transition (EMT) due to the common loss of PGC-1α and NFE2L2 (double knock-out, dKO) genes in aged animals. The implanted area was assessed by histology, immunohistochemistry and transmission electron microscopy. Confocal microscopy revealed altered regions in the filamentous actin ring. This contrasted with hexagonal RPE morphology in wild-type mice. The ultrastructural RPE features here illustrated loss of apical microvilli, alteration of cell-cell contact, loss of basal in-folding with deposits on Bruch's membrane, and excessive lipofuscin deposition in dKO samples. We also found the expression of epithelial-mesenchymal transition transcription factors, such as Snail, Slug, collagen 1, vimentin and OB-cadherin, to be significantly different in dKO RPEs. An increased immunoreactivity of senescence markers p16, DEC1 and HMGB1 was also noted. These findings suggest that EMT and senescence pathways may intersect in the retinas of dKO mice. Both processes can be activated by damage to the RPE, which may be caused by increased oxidative stress resulting from the absence of NFE2L2 and PGC-1α genes, important for antioxidant defense. This dKO model may provide useful tools for studying AMD pathogenesis and evaluating novel therapies for this disease.


Subject(s)
Cellular Senescence , Epithelial-Mesenchymal Transition , Mitochondria/pathology , NF-E2-Related Factor 2/physiology , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Retinal Pigment Epithelium/pathology , Animals , Macular Degeneration , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Phenotype , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Signal Transduction
12.
Nat Rev Nephrol ; 17(5): 335-349, 2021 05.
Article in English | MEDLINE | ID: mdl-33547418

ABSTRACT

Kidney damage varies according to the primary insult. Different aetiologies of acute kidney injury (AKI), including kidney ischaemia, exposure to nephrotoxins, dehydration or sepsis, are associated with characteristic patterns of damage and changes in gene expression, which can provide insight into the mechanisms that lead to persistent structural and functional damage. Early morphological alterations are driven by a delicate balance between energy demand and oxygen supply, which varies considerably in different regions of the kidney. The functional heterogeneity of the various nephron segments is reflected in their use of different metabolic pathways. AKI is often linked to defects in kidney oxygen supply, and some nephron segments might not be able to shift to anaerobic metabolism under low oxygen conditions or might have remarkably low basal oxygen levels, which enhances their vulnerability to damage. Here, we discuss why specific kidney regions are at particular risk of injury and how this information might help to delineate novel routes for mitigating injury and avoiding permanent damage. We suggest that the physiological heterogeneity of the kidney should be taken into account when exploring novel renoprotective strategies, such as improvement of kidney tissue oxygenation, stimulation of hypoxia signalling pathways and modulation of cellular energy metabolism.


Subject(s)
Acute Kidney Injury/etiology , Kidney/physiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Hypoxia , Disease Susceptibility , Energy Metabolism , Gene Expression , Humans , Kidney/pathology , Mitochondria/physiology , Oxygen/metabolism , PPAR gamma/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology
13.
Neuropeptides ; 87: 102134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33639357

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and the deposition of Lewy bodies. Mitochondrial dysfunction, oxidative stress, and autophagy dysfunction are involved in the pathogenesis of PD. Ghrelin is a brain-gut peptide that has been reported that protected against 1-methyl-4-phenyl-1,2,3,6- tetrahydropyran (MPTP)/MPP+-induced toxic effects. In the present work, human neuroblastoma SH-SY5Y cells were exposed to rotenone as a PD model to explore the underlying mechanism of ghrelin. We found that ghrelin inhibited rotenone-induced cytotoxicity, mitochondrial dysfunction, and apoptosis by improving cell viability, increasing the ratio of red/green of JC-1, inhibiting the production of reactive oxidative species (ROS), and regulating Bcl-2, Bax, Cytochrome c, caspase-9, and caspase-3 expression. Besides, ghrelin promoted mitophagy accompanied by up-regulating microtubule-associated protein 1 Light Chain 3B-II/I(LC3B-II/I) and Beclin1 but decreasing the expression of p62. Moreover, ghrelin promoted PINK1/Parkin mitochondrial translocation. Additionally, we investigated that ghrelin activated the AMPK/SIRT1/PGC1α pathway and pharmacological inhibition of AMPK and SIRT1 abolished the cytoprotection of ghrelin, decreased the level of mitophagy, and PINK1/Parkin mitochondrial translocation. Taken together, our findings suggested that mitophagy and AMPK/SIRT1/PGC1α pathways were related to the cytoprotection of ghrelin. These findings provided novel insights into the underlying mechanisms of ghrelin, further mechanistic studies on preclinical and clinical levels are required to be conducted with ghrelin to avail and foresee it as a potential agent in the treatment and management of PD.


Subject(s)
Ghrelin/physiology , Mitochondria/drug effects , Mitophagy/physiology , Nerve Tissue Proteins/physiology , Rotenone/toxicity , Signal Transduction/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/physiology , Apoptosis/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Ghrelin/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neuroblastoma , Oxidative Stress/drug effects , Parkinson Disease , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Protein Kinases/metabolism , Protein Transport/drug effects , Reactive Oxygen Species , Rotenone/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/physiology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
14.
Microcirculation ; 28(4): e12675, 2021 05.
Article in English | MEDLINE | ID: mdl-33377581

ABSTRACT

BACKGROUND: Dexmedetomidine (Dex) exerts an effective therapeutic role in numerous diseases associated with ischemia/reperfusion (I/R) injury via its anti-apoptosis properties. Therefore, this study explores the cardioprotective effects of Dex in cardiac microvascular endothelial cells (CMECs) in response to oxygen-glucose deprivation and re-oxygenation (OGD/R) injury and its potential mechanism. MATERIAL AND METHODS: CMECs were pretreatment with different concentration of Dex, then exposed to OGD/R. Cell viability was measured with CCK-8 assay. Apoptosis was evaluated by flow cytometry, and apoptosis-related protein was determined by Western blot. Autophagy was assessed by transmission electron microscopy and autophagy-related proteins. Besides, the role peroxisome proliferator-activated receptors (PPARδ) in Dex-mediated anti-apoptosis property was validated with agonist and antagonist. RESULTS: OGD/R significantly decreased cell viability, increased reactive oxygen species, caused disorder of autophagy, and increased apoptosis in CMECs. Dex enhanced the viability of the OGD/R-treated CMECs and effectively decreased reactive oxygen species production. Autophagy in CMECs was activated by Dex, as evidenced by the increase in the ratio of LC3B-II/I, expression level of Beclin1 and number of autophagosomes in the OGD/R-induced CMECs. The mechanistic investigation indicated that PPARδ antagonist GW501516 aggravated cell damage following OGD/R, while PPARδ agonist GW6471 partly abolished the Dex-mediated protective effects. CONCLUSIONS: Dex activated the PPARδ-AMPK-PGC-1α pathway-mediated autophagy in CMECs, therefore to inhibit excessive apoptosis induced by OGD/R. Dex may potentially be a therapeutic intervention for myocardial I/R injury.


Subject(s)
Coronary Vessels , Dexmedetomidine , Endothelium, Vascular/drug effects , Myocardial Reperfusion Injury , PPAR delta , Protective Agents , AMP-Activated Protein Kinases/physiology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Autophagy/drug effects , Autophagy/physiology , Cells, Cultured , Coronary Circulation/drug effects , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Dexmedetomidine/pharmacology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Microcirculation/drug effects , Microvessels/drug effects , Microvessels/pathology , Microvessels/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , PPAR delta/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Protective Agents/pharmacology , Reactive Oxygen Species , Signal Transduction
15.
Biomed Pharmacother ; 131: 110684, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33152903

ABSTRACT

Marein, an active component of the Coreopsis tinctoria Nutt. plant, is known to improve diabetic nephropathy (DN). However, its anti-diabetic functions in DN and potential mechanisms remain unclear. The aim of this study was to elucidate the effects and mechanisms of Marein in diabetic db/db mice with DN, and in high glucose-treated HK-2 cells. In vivo, treating diabetic db/db mice with Marein for 12 consecutive weeks restored diabetes-induced hyperglycemia and dyslipidemia, and ameliorated renal function deterioration, glomerulosclerosis, and renal ectopic lipid deposition. Marein exerted renoprotective effects by directly inhibiting renal tubule sodium glucose transporter 2 (SGLT2) expression, and then activating the AMP-activated protein kinase (AMPK)/acetyl CoA carboxylase (ACC)/peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) pathway in db/db mice. Meanwhile, Marein ameliorated fibrosis and inflammation by suppressing the pro-inflammatory factors interleukin-6 (IL-6) and monocyte chemotactic protein-1 (MCP-1), and expression of the extracellular matrix proteins, fibronectin (FN) and collagen 1 (COL1) in diabetic mice. In vitro, MDCK monolayer cells were established to explore the characteristics of Marein transmembrane transport. Marein was found to be absorbed across the membrane at a medium level that involved active transport and this was mediated by SGLTs. In HK-2 cells, Marein decreased uptake of the fluorescent glucose analog, 2-NBDG, by 22 % by inhibiting SGLT2 expression. In high glucose-treated HK-2 cells, Marein decreased SGLT2 expression and increased phosphorylated (p)-AMPK/p-ACC to improve high glucose-induced cellular dysfunction. Furthermore, Marein treatment decreased SGLT2 expression in SGLT2-overexpressing HK-2 cells. In addition, molecular docking and dynamics analysis revealed that SGLT2 was a direct target of Marein. Collectively, our results demonstrated that Marein ameliorates DN by inhibiting renal SGLT2 and activating p-AMPK, suggesting Marein can potentially prevent DN by suppressing renal SGLT2 expression directly.


Subject(s)
AMP-Activated Protein Kinases/physiology , Chalcones/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Blood Glucose/analysis , Cells, Cultured , Chalcones/chemistry , Chalcones/pharmacokinetics , Chalcones/pharmacology , Diabetes Mellitus, Experimental/metabolism , Humans , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Phlorhizin/pharmacology , Signal Transduction/drug effects , Sodium-Glucose Transporter 2/chemistry
16.
Sci Rep ; 10(1): 16888, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060645

ABSTRACT

The arcuate nucleus (ARC) of the hypothalamus is a key regulator of food intake, brown adipose tissue (BAT) thermogenesis, and locomotor activity. Whole-body deficiency of the transcriptional coactivator peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1ß (PGC-1ß) disrupts mouse circadian locomotor activity and BAT-regulated thermogenesis, in association with altered gene expression at the central level. We examined whether PGC-1ß expression in the ARC is required for proper energy balance and locomotor behavior by generating mice lacking the PGC-1ß gene specifically in pro-opiomelanocortin (POMC) neurons. POMC neuron-specific deletion of PGC-1ß did not impact locomotor behavior, food intake, body composition, energy fuel utilization and metabolic rate in fed, 24-h fasted and 24-h refed conditions. In contrast, in the fed state, deletion of PGC-1ß in POMC cells elevated core body temperature during the nighttime period. Importantly, this higher body temperature is not associated with changes in BAT function and gene expression. Conversely, we provide evidence that mice lacking PGC-1ß in POMC neurons are more sensitive to the effect of leptin on heat dissipation. Our data indicate that PGC-1ß-expressing POMC neurons are part of a circuit controlling body temperature homeostasis and that PGC-1ß function in these neurons is involved in the thermoregulatory effect of leptin.


Subject(s)
Body Temperature Regulation/physiology , Neurons/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Adipose Tissue, Brown/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight , Energy Metabolism/drug effects , Gene Expression Regulation , Hypothalamus/metabolism , Leptin/metabolism , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/physiology , Thermogenesis/physiology , Transcription Factors/metabolism
17.
J Cell Mol Med ; 24(18): 10866-10875, 2020 09.
Article in English | MEDLINE | ID: mdl-32757458

ABSTRACT

Cardiomyocyte dysfunction is attributed to excess oxidative damage, but the molecular pathways involved in this process have not been completely elucidated. Evidence indicates that isosteviol sodium (STVNa) has cardioprotective effects. We therefore aimed to identify the effect of STVNa on cardiomyocytes, as well as the potential mechanisms involved in this process. We established two myocardial hypertrophy models by treating H9c2 cells with high glucose (HG) and isoprenaline (ISO). Our results showed that STVNa reduced H9c2 mitochondrial damage by attenuating oxidative damage and altering the morphology of mitochondria. The results also indicated that STVNa had a positive effect on HG- and ISO-induced damages via mitochondrial biogenesis. The protective effects of STVNa on cardiomyocytes were associated with the regulation of the SIRT1/PGC-1α signalling pathway. Importantly, the effects of STVNa involved different methods of regulation in the two models, which was confirmed by experiments using an inhibitor and activator of SIRT1. Together, the results provide the basis for using STVNa as a therapy for the prevention of cardiomyocyte dysfunctions.


Subject(s)
Cardiotonic Agents/pharmacology , Diterpenes, Kaurane/pharmacology , Myocytes, Cardiac/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Signal Transduction/drug effects , Sirtuin 1/physiology , Animals , Carbazoles/pharmacology , Cell Line , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/physiology , DNA, Mitochondrial/ultrastructure , Glucose/toxicity , Hypertrophy , Isoproterenol/toxicity , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Organelle Biogenesis , Rats , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology , Sirtuin 1/drug effects
18.
Ann Rheum Dis ; 79(9): 1227-1233, 2020 09.
Article in English | MEDLINE | ID: mdl-32482644

ABSTRACT

OBJECTIVES: Coactivators are a heterogeneous family of transcriptional regulators that are essential for modulation of transcriptional outcomes and fine-tune numerous cellular processes. The aim of the present study was to evaluate the role of the coactivator peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) in the pathogenesis of systemic sclerosis (SSc). METHODS: Expression of PGC-1α was analysed by real-time PCR, western blot and immunofluorescence. Modulation of autophagy was analysed by reporter studies by expression of autophagy-related genes. The effects of PGC-1α knockdown on collagen production and myofibroblast differentiation were analysed in cultured human fibroblasts and in two mouse models with fibroblast-specific knockout of PGC-1α. RESULTS: The expression of PGC-1α was induced in dermal fibroblasts of patients with SSc and experimental murine fibrosis. Transforming growth factor beta (TGFß), hypoxia and epigenetic mechanisms regulate the expression of PGC-1α in fibroblasts. Knockdown of PGC-1α prevented the activation of autophagy by TGFß and this translated into reduced fibroblast-to-myofibroblast differentiation and collagen release. Knockout of PGC-1α in fibroblasts prevented skin fibrosis induced by bleomycin and by overexpression of a constitutively active TGFß receptor type I. Moreover, pharmacological inhibition of PGC-1α by SR18292 induced regression of pre-established, bleomycin-induced skin fibrosis. CONCLUSION: PGC-1α is upregulated in SSc and promotes autophagy to foster TGFß-induced fibroblast activation. Targeting of PGC-1α prevents aberrant autophagy, inhibits fibroblast activation and tissue fibrosis and may over therapeutic potential.


Subject(s)
Autophagy/genetics , Fibroblasts/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Scleroderma, Systemic/genetics , Scleroderma, Systemic/pathology , Animals , Bleomycin/pharmacology , Blotting, Western , Collagen/biosynthesis , Disease Models, Animal , Fibrosis , Fluorescent Antibody Technique , Humans , Mice , Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Up-Regulation
19.
Appl Physiol Nutr Metab ; 45(9): 927-936, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32516539

ABSTRACT

The majority of human energy metabolism occurs in skeletal muscle mitochondria emphasizing the importance of understanding the regulation of myocellular mitochondrial function. The transcriptional co-activator peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) has been characterized as a major factor in the transcriptional control of several mitochondrial components. Thus, PGC-1α is often described as a master regulator of mitochondrial biogenesis as well as a central player in regulating the antioxidant defense. However, accumulating evidence suggests that PGC-1α is also involved in the complex regulation of mitochondrial quality beyond biogenesis, which includes mitochondrial network dynamics and autophagic removal of damaged mitochondria. In addition, mitochondrial reactive oxygen species production has been suggested to regulate skeletal muscle insulin sensitivity, which may also be influenced by PGC-1α. This review aims to highlight the current evidence for PGC-1α-mediated regulation of skeletal muscle mitochondrial function beyond the effects on mitochondrial biogenesis as well as the potential PGC-1α-related impact on insulin-stimulated glucose uptake in skeletal muscle. Novelty PGC-1α regulates mitochondrial biogenesis but also has effects on mitochondrial functions beyond biogenesis. Mitochondrial quality control mechanisms, including fission, fusion, and mitophagy, are regulated by PGC-1α. PGC-1α-mediated regulation of mitochondrial quality may affect age-related mitochondrial dysfunction and insulin sensitivity.


Subject(s)
Mitochondria/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Aging , Animals , Antioxidants/metabolism , Energy Metabolism , Gene Expression Regulation , Humans , Insulin Resistance , Muscle, Skeletal/physiology , Organelle Biogenesis
20.
Metabolism ; 109: 154280, 2020 08.
Article in English | MEDLINE | ID: mdl-32473155

ABSTRACT

OBJECTIVE: Obesity is recognized as the cause of multiple metabolic diseases and is rapidly increasing worldwide. As obesity is due to an imbalance in energy homeostasis, the promotion of energy consumption through browning of white adipose tissue (WAT) has emerged as a promising therapeutic strategy to counter the obesity epidemic. However, the molecular mechanisms of the browning process are not well understood. In this study, we investigated the effects of the GATA family of transcription factors on the browning process. METHODS: We used qPCR to analyze the expression of GATA family members during WAT browning. In order to investigate the function of GATA3 in the browning process, we used the lentivirus system for the ectopic expression and knockdown of GATA3. Western blot and real-time qPCR analyses revealed the regulation of thermogenic genes upon ectopic expression and knockdown of GATA3. Luciferase reporter assays, co-immunoprecipitation, and chromatin immunoprecipitation were performed to demonstrate that GATA3 interacts with proliferator-activated receptor-γ co-activator-1α (PGC-1α) to regulate the promoter activity of uncoupling protein-1 (UCP-1). Enhanced energy expenditure by GATA3 was confirmed using oxygen consumption assays, and the mitochondrial content was assessed using MitoTracker. Furthermore, we examined the in vivo effects of lentiviral GATA3 overexpression and knockdown in inguinal adipose tissue of mice. RESULTS: Gata3 expression levels were significantly elevated in the inguinal adipose tissue of mice exposed to cold conditions. Ectopic expression of GATA3 enhanced the expression of UCP-1 and thermogenic genes upon treatment with norepinephrine whereas GATA3 knockdown had the opposite effect. Luciferase reporter assays using the UCP-1 promoter region showed that UCP-1 expression was increased in a dose-dependent manner by GATA3 regardless of norepinephrine treatment. GATA3 was found to directly bind to the promoter region of UCP-1. Furthermore, our results indicated that GATA3 interacts with the transcriptional coactivator PGC-1α to increase the expression of UCP-1. Taken together, we demonstrate that GATA3 has an important role in enhancing energy expenditure by increasing the expression of thermogenic genes both in vitro and in vivo. CONCLUSION: GATA3 may represent a promising target for the prevention and treatment of obesity by regulating thermogenic capacity.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , GATA3 Transcription Factor/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Uncoupling Protein 1/metabolism , Animals , Cold Temperature , Energy Metabolism , GATA3 Transcription Factor/genetics , Humans , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Promoter Regions, Genetic , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...