Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Acta Pharmacol Sin ; 44(3): 499-512, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36229600

ABSTRACT

Cannabidiol (CBD) reportedly exerts protective effects against many psychiatric disorders and neurodegenerative diseases, but the mechanisms are poorly understood. In this study, we explored the molecular mechanism of CBD against cerebral ischemia. HT-22 cells or primary cortical neurons were subjected to oxygen-glucose deprivation insult followed by reoxygenation (OGD/R). In both HT-22 cells and primary cortical neurons, CBD pretreatment (0.1, 0.3, 1 µM) dose-dependently attenuated OGD/R-induced cell death and mitochondrial dysfunction, ameliorated OGD/R-induced endoplasmic reticulum (ER) stress, and increased the mitofusin-2 (MFN2) protein level in HT-22 cells and primary cortical neurons. Knockdown of MFN2 abolished the protective effects of CBD. CBD pretreatment also suppressed OGD/R-induced binding of Parkin to MFN2 and subsequent ubiquitination of MFN2. Overexpression of Parkin blocked the effects of CBD in reducing MFN2 ubiquitination and reduced cell viability, whereas overexpressing MFN2 abolished Parkin's detrimental effects. In vivo experiments were conducted on male rats subjected to middle cerebral artery occlusion (MCAO) insult, and administration of CBD (2.5, 5 mg · kg-1, i.p.) dose-dependently reduced the infarct volume and ER stress in the brains. Moreover, the level of MFN2 within the ischemic penumbra of rats was increased by CBD treatment, while the binding of Parkin to MFN2 and the ubiquitination of MFN2 was decreased. Finally, short hairpin RNA against MFN2 reversed CBD's protective effects. Together, these results demonstrate that CBD protects brain neurons against cerebral ischemia by reducing MFN2 degradation via disrupting Parkin's binding to MFN2, indicating that MFN2 is a potential target for the treatment of cerebral ischemia.


Subject(s)
Brain Ischemia , Cannabidiol , GTP Phosphohydrolases , Neuroprotective Agents , Reperfusion Injury , Animals , Male , Rats , Apoptosis , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cannabidiol/pharmacology , Glucose/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neuroprotection , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Reperfusion Injury/prevention & control , Ubiquitin-Protein Ligases/metabolism , GTP Phosphohydrolases/drug effects , GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism
2.
CNS Neurosci Ther ; 29(3): 783-788, 2023 03.
Article in English | MEDLINE | ID: mdl-36550598

ABSTRACT

In the ischemic brain, hypoxia leads to mitochondrial dysfunction, insufficient energy production, and astrocyte activation. Yet, most studies investigating mitochondrial dysfunction in cerebral ischemia have focused exclusively on neurons. This review will highlight the importance of the morphological, molecular, and functional heterogeneity of astrocytes in their role in brain injuries and explore how activated astrocytes exhibit calcium imbalance, reactive oxygen species overproduction, and apoptosis. In addition, special focus will be given to the role of the mitochondrial protein frataxin in activated astrocytes during ischemia and its putative role in the pharmacological management of cerebral ischemia.


Subject(s)
Brain Ischemia , Mitochondrial Proteins , Humans , Astrocytes/metabolism , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Mitochondria/physiology , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Frataxin
3.
Brain Res Bull ; 180: 12-23, 2022 03.
Article in English | MEDLINE | ID: mdl-34953929

ABSTRACT

Studies have found that Platonin has neuroprotective effect, but its molecular mechanism needs further study. We found that at the early stage of cerebral ischemia/reperfusion injury, Platonin treatment significantly reduced cerebral infarct lesions, improved neurological scores, and exerted neuroprotective effects. Our group has shown that NLRP3 inflammasomes activation is required to mediate neuronal injury during cerebral ischemia /reperfusion injury. The brain protective effect of Platonin is related to its ability to effectively regulate autophagy and NLRP3 inflammasomes-derived inflammation. Platonin treatment effectively induced autophagy (LC3II/I, p62) and reduced NLRP3 inflammasomes activation(NLRP3, cleaved-IL-1ß,cleaved-IL-18, cleaved-caspase1). However, 3-MA (15 mg/kg) treatment downregulated the inhibitory effect of Platonin on NLRP3 inflammasomes. We also studied the location of BNIP3 in Platonin-mediated neuroprotection and found that Platonin induced the expression of autophagic protein BNIP3 and enhanced the co-immunoprecipitation of BNIP3 with LC3, and double-labeled immunofluorescence also showed enhanced co-localization of BNIP3 with LC3. Finally, si-BNIP3 transfection attenuated the co localization of BNIP3 with LC3, decreased the autophagy activity to a certain extent and blocked the inhibition of NLRP3 inflammasomes-derived inflammation by Platonin. This study demonstrated that Platonin may play a neuroprotection role in cerebral I / R injury by inhibiting NLRP3 inflammasomes activation through upregulating autophagy via BNIP3 / LC3 pathway.


Subject(s)
Autophagy/drug effects , Inflammasomes/drug effects , Membrane Proteins/drug effects , Microtubule-Associated Proteins/drug effects , Mitochondrial Proteins/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Thiazoles/pharmacology , Animals , Disease Models, Animal , Rats
4.
Molecules ; 26(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34641427

ABSTRACT

O-GlcNAcylation is a nutrient-driven post-translational modification known as a metabolic sensor that links metabolism to cellular function. Recent evidences indicate that the activation of O-GlcNAc pathway is a potential pro-survival pathway and that acute enhancement of this response is conducive to the survival of cells and tissues. 2-(4-Methoxyphenyl)ethyl-2-acetamido-2-deoxy-ß-d-pyranoside (SalA-4g), is a salidroside analogue synthesized in our laboratory by chemical structure-modification, with a phenyl ring containing a para-methoxy group and a sugar ring consisting of N-acetylglucosamine. We have previously shown that SalA-4g elevates levels of protein O-GlcNAc and improves neuronal tolerance to ischemia. However, the specific target of SalA-4g regulating O-GlcNAcylation remains unknown. To address these questions, in this study, we have focused on mitochondrial network homeostasis mediated by O-GlcNAcylation in SalA-4g's neuroprotection in primary cortical neurons under ischemic-like conditions. O-GlcNAc-modified mitochondria induced by SalA-4g demonstrated stronger neuroprotection under oxygen glucose deprivation and reoxygenation stress, including the improvement of mitochondrial homeostasis and bioenergy, and inhibition of mitochondrial apoptosis pathway. Blocking mitochondrial protein O-GlcNAcylation with OSMI-1 disrupted mitochondrial network homeostasis and antagonized the protective effects of SalA-4g. Collectively, these data demonstrate that mitochondrial homeostasis mediated by mitochondrial protein O-GlcNAcylation is critically involved in SalA-4g neuroprotection.


Subject(s)
Acetylglucosamine/analogs & derivatives , Energy Metabolism , Ischemia/prevention & control , Mitochondria/drug effects , Mitochondrial Proteins/chemistry , Neurons/drug effects , Neuroprotective Agents/pharmacology , Acetylglucosamine/pharmacology , Animals , Glucose/metabolism , Glycosylation , Homeostasis , Ischemia/metabolism , Ischemia/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/drug effects , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley
5.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34080021

ABSTRACT

Colorectal cancer (CRC) ranks third in incidence and second in mortality among all types of cancer, and due to its insidious onset and lack of early symptoms, it is usually diagnosed at a later stage. Saponins, a class of compounds abundant in plants, have been reported to possess prominent anti­tumour properties. The use of ginsenoside Rg3 in the clinical setting was authorized by the National Medicinal Products Administration of China. In the present study, total saponins from Rhizoma Panacis Majoris (RPMTG) were prepared, and the pharmacological mechanisms underlying the anti­CRC effects of RPMTG were investigated. The effect of RPMTG on the proliferation, cell cycle progression and apoptosis of HCT116 and SW620 cells were detected by MTT, flow cytometry and western blotting assays, and it was demonstrated that RPMTG could inhibit the proliferation of HCT116 and SW620 cells with IC50 values of 315.8 and 355.1 µg/ml, respectively, induce cell cycle arrest in the S and G0/G1 phase, and trigger apoptosis by downregulating the expression of the anti­apoptotic proteins Bcl­2, Bcl­xL and induced myeloid leukaemia cell differentiation protein Mcl­1, and increasing the expression of the pro­apoptotic proteins Bax and Bad, cleaved caspased­3 and poly(ADP)­ribose polymerase. These findings suggested that RPMTG induced apoptosis through mitochondrial­related pathways. In addition, RPMTG also decreased the expression of phosphorylated (p)­extracellular signal­regulated kinase and increased p­c­Jun N­terminal kinase (p­JNK) and p­p38. Moreover, the effects of RPMTG on cell proliferation and apoptosis were partially reversed when the JNK and p38 mitogen­activated protein kinase (MAPK) pathways were inhibited, indicating that RPMTG triggered apoptosis mainly via regulating JNK and p38 MAPK signalling. Therefore, RPMTG may have potential as an anti­CRC agent, and further evaluations are needed.


Subject(s)
Colorectal Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , MAP Kinase Signaling System/drug effects , Panax/chemistry , Rhizome/chemistry , Saponins/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Drugs, Chinese Herbal/isolation & purification , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitochondrial Proteins/drug effects , Protein Kinase Inhibitors/pharmacology , Saponins/isolation & purification , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Exp Biol Med (Maywood) ; 246(18): 2019-2028, 2021 09.
Article in English | MEDLINE | ID: mdl-34053232

ABSTRACT

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent organic pollutant that is strongly associated with a number of human diseases and birth defects, including cleft palate. Transforming growth factor (TGF) plays a significant role during mammalian palatogenesis. However, the epigenetic mechanism of transforming growth factors in the process of TCDD-induced cleft palate is unclear. The purpose of this research was to investigate the relationship and potential mechanism between TGF-ß2/3 promoter DNA methylation and Smad signaling during TCDD-induced cleft palate. Pregnant C57BL/6N mice were exposed to 64 µg/kg TCDD on gestational day 10 (GD10) to establish the cleft palate model and palatal tissues of embryos were collected on GD13, GD14, and GD15 for subsequent experiments. TGF-ß2/3 mRNA expression, TGF-ß2/3 promoter methylation, and Smad signaling molecules expression were assessed in the palate of the two groups. The results showed that the incidence of cleft palate was 94.7% in the TCDD-treated group whereas no cleft palate was found in the control group. TCDD-treated group altered specific CpG sites of TGF-ß2/3 promoter methylation. Compared to the control group, the proliferation of mouse embryonic palate mesenchymal stromal cells (MEPM), the expressions of TGF-ß2/3, p-Smad2, and Smad4 were all reduced, while the expression of Smad7 was significantly increased in the atAR group. Smad signaling was downregulated by TCDD. Therefore, we suggest that TGF-ß2/3 promoter methylation and Smad signaling may be involved in TCDD-induced cleft palate formation in fetal mice.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , DNA Methylation/drug effects , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Animals , Down-Regulation , Mice, Inbred C57BL , Palate/drug effects , Polychlorinated Dibenzodioxins/metabolism , Promoter Regions, Genetic/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism
7.
J Neurochem ; 158(2): 262-281, 2021 07.
Article in English | MEDLINE | ID: mdl-33837559

ABSTRACT

Tissue accumulation and high urinary excretion of ethylmalonic acid (EMA) are found in ethylmalonic encephalopathy (EE), an inherited disorder associated with cerebral and cerebellar atrophy whose pathogenesis is poorly established. The in vitro and in vivo effects of EMA on bioenergetics and redox homeostasis were investigated in rat cerebellum. For the in vitro studies, cerebellum preparations were exposed to EMA, whereas intracerebellar injection of EMA was used for the in vivo evaluation. EMA reduced state 3 and uncoupled respiration in vitro in succinate-, glutamate-, and malate-supported mitochondria, whereas decreased state 4 respiration was observed using glutamate and malate. Furthermore, mitochondria permeabilization and succinate supplementation diminished the decrease in state 3 with succinate. EMA also inhibited the activity of KGDH, an enzyme necessary for glutamate oxidation, in a mixed manner and augmented mitochondrial efflux of α-ketoglutarate. ATP levels were markedly reduced by EMA, reflecting a severe bioenergetic disruption. Docking simulations also indicated interactions between EMA and KGDH and a competition with glutamate and succinate for their mitochondrial transporters. In vitro findings also showed that EMA decreased mitochondrial membrane potential and Ca2+ retention capacity, and induced swelling in the presence of Ca2+ , which were prevented by cyclosporine A and ADP and ruthenium red, indicating mitochondrial permeability transition (MPT). Moreover, EMA, at high concentrations, mildly increased ROS levels and altered antioxidant defenses in vitro and in vivo. Our data indicate that EMA-induced impairment of glutamate and succinate oxidation and MPT may contribute to the pathogenesis of the cerebellum abnormalities in EE.


Subject(s)
Cerebellum/drug effects , Cerebellum/metabolism , Energy Metabolism/drug effects , Glutamates/metabolism , Malonates/toxicity , Mitochondrial Permeability Transition Pore , Succinates/metabolism , Animals , Ketoglutaric Acids/metabolism , Malates/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Molecular Docking Simulation , Oxidation-Reduction , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Succinates/pharmacology
8.
Autophagy ; 17(8): 1934-1946, 2021 08.
Article in English | MEDLINE | ID: mdl-32722981

ABSTRACT

Mitophagy, the elimination of damaged mitochondria through autophagy, promotes neuronal survival in cerebral ischemia. Previous studies found deficient mitophagy in ischemic neurons, but the mechanisms are still largely unknown. We determined that BNIP3L/NIX, a mitophagy receptor, was degraded by proteasomes, which led to mitophagy deficiency in both ischemic neurons and brains. BNIP3L exists as a monomer and homodimer in mammalian cells, but the effects of homodimer and monomer on mitophagy are unclear. Site-specific mutations in the transmembrane domain of BNIP3L (S195A and G203A) only formed the BNIP3L monomer and failed to induce mitophagy. Moreover, overexpression of wild-type BNIP3L, in contrast to the monomeric BNIP3L, rescued the mitophagy deficiency and protected against cerebral ischemic injury. The macroautophagy/autophagy inhibitor 3-MA and the proteasome inhibitor MG132 were used in cerebral ischemic brains to identify how BNIP3L was reduced. We found that MG132 blocked the loss of BNIP3L and subsequently promoted mitophagy in ischemic brains. In addition, the dimeric form of BNIP3L was more prone to be degraded than its monomeric form. Carfilzomib, a drug for multiple myeloma therapy that inhibits proteasomes, reversed the BNIP3L degradation and restored mitophagy in ischemic brains. This treatment protected against either acute or chronic ischemic brain injury. Remarkably, these effects of carfilzomib were abolished in bnip3l-/- mice. Taken together, the present study linked BNIP3L degradation by proteasomes with mitophagy deficiency in cerebral ischemia. We propose carfilzomib as a novel therapy to rescue ischemic brain injury by preventing BNIP3L degradation.Abbreviations: 3-MA: 3-methyladenine; AAV: adeno-associated virus; ATG7: autophagy related 7; BCL2L13: BCL2-like 13 (apoptosis facilitator); BNIP3L/NIX: BCL2/adenovirus E1B interacting protein 3-like; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; CFZ: carfilzomib; COX4I1: cytochrome c oxidase subunit 4I1; CQ: chloroquine; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; I-R: ischemia-reperfusion; MAP1LC3A/LC3A: microtube-associated protein 1 light chain 3 alpha; MAP1LC3B/LC3B: microtube-associated protein 1 light chain 3 beta; O-R: oxygen and glucose deprivation-reperfusion; OGD: oxygen and glucose deprivation; PHB2: prohibitin 2; pMCAO: permanent middle cerebral artery occlusion; PRKN/PARK2: parkin RBR E3 ubiquitin protein ligase; PT: photothrombosis; SQSTM1: sequestosome 1; tMCAO: transient middle cerebral artery occlusion; TOMM20: translocase of outer mitochondrial membrane 20; TTC: 2,3,5-triphenyltetrazolium hydrochloride.


Subject(s)
Autophagy/drug effects , Ischemia/drug therapy , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/drug effects , Oligopeptides/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Ischemia/metabolism , Membrane Proteins/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Mitophagy/genetics , Reactive Oxygen Species/metabolism
9.
J Alzheimers Dis ; 79(1): 249-265, 2021.
Article in English | MEDLINE | ID: mdl-33252074

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß (Aß) species and deposition of senile plaques (SPs). Clinical trials with the anti-Aß antibody aducanumab have been completed recently. OBJECTIVE: To characterize the proteomic profile of SPs and surrounding tissue in a mouse model of AD in 10-month-old tgAPPPS1-21 mice after chronic treatment with aducanumab for four months with weekly dosing (10 mg/kg). METHODS: After observing significant reduction of SP numbers in hippocampi of aducanumab-treated mice, we applied a localized proteomic analysis by combining laser microdissection and liquid chromatography-tandem mass spectrometry (LC-MS/MS) of the remaining SPs in hippocampi. We microdissected three subregions, containing SPs, SP penumbra level 1, and an additional penumbra level 2 to follow the proteomic profile as gradient. RESULTS: In the aducanumab-treated mice, we identified 17 significantly regulated proteins that were associated with 1) mitochondria and metabolism (ACAT2, ATP5J, ETFA, EXOG, HK1, NDUFA4, NDUFS7, PLCB1, PPP2R4), 2) cytoskeleton and axons (ADD1, CAPZB, DPYSL3, MAG), 3) stress response (HIST1H1C/HIST1H1D, HSPA12A), and 4) AßPP trafficking/processing (CD81, GDI2). These pathways and some of the identified proteins are implicated in AD pathogenesis. Proteins associated with mitochondria and metabolism were mainly upregulated while proteins associated with AßPP trafficking/processing and stress response pathways were mainly downregulated, suggesting that aducanumab could lead to a beneficial proteomic profile around SPs in tgAPPPS1-21 mice. CONCLUSION: We identified novel proteomic patterns of SPs and surrounding tissue indicating that chronic treatment with aducanumab could inhibit Aß toxicity and increase phagocytosis and cell viability.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Amyloid beta-Protein Precursor/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Brain/drug effects , Plaque, Amyloid/metabolism , Proteome/drug effects , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Chromatography, Liquid , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Presenilin-1/genetics , Protein Transport/drug effects , Proteomics , Stress, Physiological/drug effects , Tandem Mass Spectrometry
10.
Mitochondrion ; 56: 62-72, 2021 01.
Article in English | MEDLINE | ID: mdl-33221353

ABSTRACT

Brain is a fully differentiated organ and is sensitive towards oxidative damage of various compounds including lipids, proteins, and DNA that occurs during process of normal aging and is mainly due to its high energy metabolism and reduced activity of anti-oxidative defense mechanism. Mitochondria are dynamic ATP-generating organelles which constitutes cellular functions such as regulation of intracellular calcium, bio-energetic processes, and reduction-oxidation of cells. Such functioning is negatively affected due to the presence of amyloid ß peptide (Aß) which is involved in pathogenesis of Alzheimer disease (AD). Aß interacts with mitochondria and leads to mitochondrial dysfunction. Mitochondrial dysfunction, abnormal interactions, oxidative stress, and mis-folding of synaptic proteins inside nervous system are explored and regarded as primary or initial features in insurgence of pathology (AD and other neurological disease). The major histopathological hallmarks of AD are characterized by presence of these hallmarks intracellularly, its further progression and exacerbation which leads to excessive accumulation of oligomeric as well as fibrillar-ß-amyloid peptides (present extracellularly) and accumulation of neurofibrillary tangles intracellularly. The current review will focus on alterations and variation in mitochondria/mitochondrial DNA (mtDNA) and the rationale for involvement of related abnormalities in pathogenesis of AD.


Subject(s)
Alzheimer Disease/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Disease Progression , Gene Expression Regulation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Proteins/drug effects , Molecular Targeted Therapy , Oxidative Stress/drug effects
11.
Environ Res ; 188: 109824, 2020 09.
Article in English | MEDLINE | ID: mdl-32593899

ABSTRACT

Exposure to arsenic is a risk factor for nonalcoholic steatohepatitis (NASH). Ferroptosis is a form of regulated cell death defined by the accumulation of lipid peroxidation. In the current study, we observed the occurrence of ferroptosis in arsenic-induced NASH by assessing ferroptosis related hallmarks. In vitro, we found that ferrostatin-1 effectively attenuated the executing of ferroptosis and NASH. Simultaneously, the expression of ACSL4 (acyl-CoA synthetase long-chain family member 4) was upregulated in rat's liver and L-02 cells exposed to arsenic. While, suppression of ACSL4 with rosiglitazone or ACSL4 siRNA remarkably alleviated arsenic-induced NASH and ferroptosis through diminishing 5-hydroxyeicosatetraenoic acid (5-HETE) content. Additionally, Mitofusin 2 (Mfn2), a physical tether between endoplasmic reticulum and mitochondria, has rarely been explored in the ferroptosis. Using Mfn2 siRNA or inositol-requiring enzyme 1 alpha (IRE1α) inhibitor, we found NASH and ferroptosis were obviously mitigated through reducing 5-HETE content. Importantly, Co-IP assay indicated that Mfn2 could interact with IRE1α and promoted the production of 5-HETE, ultimately led to ferroptosis and NASH. Collectively, our data showed that ferroptosis is involved in arsenic-induced NASH. These data provide insightful viewpoints into the mechanism of arsenic-induced NASH.


Subject(s)
Arsenic , Non-alcoholic Fatty Liver Disease , Animals , Arsenic/toxicity , Coenzyme A Ligases , Endoribonucleases/drug effects , Endoribonucleases/physiology , Ferroptosis , GTP Phosphohydrolases/drug effects , GTP Phosphohydrolases/physiology , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/physiology , Multienzyme Complexes/drug effects , Multienzyme Complexes/physiology , Non-alcoholic Fatty Liver Disease/chemically induced , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/physiology , Rats
12.
Cells ; 9(5)2020 04 29.
Article in English | MEDLINE | ID: mdl-32365537

ABSTRACT

Sirtuin (SIRT) is known to prevent nonalcoholic fatty liver disease (NAFLD); however, the role of SIRT4 in the progression of hepatic fibrosis remains unknown. We hypothesize that EX-527, a selective SIRT1 inhibitor, can inhibit the progression of high-fat diet (HFD)-induced hepatic fibrosis. We found that SIRT4 expression in the liver of NAFLD patients is significantly lower than that in normal subjects. In this study, EX-527 (5 µg/kg), administered to HFD rats twice a week for ten weeks, reduced the serum levels of triglyceride (TG), total cholesterol, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) and attenuated hepatic fibrosis evidenced by Masson's trichrome and hepatic fat by oil red-O staining. EX-527 upregulated SIRT2, SIRT3, and SIRT4 expression in the liver of HFD fed rats but downregulated transforming growth factor-ß1 (TGF-ß1) and α-smooth muscle actin (α-SMA) expression. It decreased proinflammatory cytokine production and hydroxyproline levels in the serum and SMAD4 expression and restored apoptotic protein (Bcl-2, Bax, and cleaved caspase-3) expression. These data propose a critical role for the SIRT4/SMAD4 axis in hepatic fibrogenesis. SIRT4 upregulation has the potential to counter HFD-induced lipid accumulation, inflammation, and fibrogenesis. We demonstrate that EX-527 is a promising candidate in inhibiting the progression of HFD-induced liver fibrosis.


Subject(s)
Carbazoles/pharmacology , Diet, High-Fat/adverse effects , Liver Cirrhosis/prevention & control , Non-alcoholic Fatty Liver Disease/drug therapy , Animal Feed , Animals , Aspartate Aminotransferases/drug effects , Disease Progression , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/drug therapy , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Rats, Zucker
13.
Sci Rep ; 10(1): 5843, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32246012

ABSTRACT

Mitochondrial dysfunction has been thought to play roles in the pathogenesis of diabetic nephropathy (DN). However, precise mechanisms underlying mitochondrial dysfunction in DN remained unclear. Herein, mitochondria were isolated from renal tubular cells after exposure to normal glucose (5.5 mM glucose), high glucose (25 mM glucose), or osmotic control (5.5 mM glucose + 19.5 mM mannitol) for 96 h. Comparative proteomic analysis revealed six differentially expressed proteins among groups that were subsequently identified by tandem mass spectrometry (nanoLC-ESI-ETD MS/MS) and confirmed by Western blotting. Several various types of post-translational modifications (PTMs) were identified in all of these identified proteins. Interestingly, phosphorylation and oxidation were most abundant in mitochondrial proteins whose levels were exclusively increased in high glucose condition. The high glucose-induced increases in phosphorylation and oxidation of mitochondrial proteins were successfully confirmed by various assays including MS/MS analyses. Moreover, high glucose also increased levels of phosphorylated ezrin, intracellular ATP and ROS, all of which could be abolished by a p38 MAPK inhibitor (SB239063), implicating a role of p38 MAPK-mediated phosphorylation in high glucose-induced mitochondrial dysfunction. These data indicate that phosphorylation and oxidation of mitochondrial proteins are, at least in part, involved in mitochondrial dysfunction in renal tubular cells during DN.


Subject(s)
Glucose/pharmacology , Kidney Tubules/drug effects , Mitochondrial Proteins/drug effects , Animals , Blotting, Western , Dogs , Kidney Tubules/metabolism , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/metabolism , Mass Spectrometry , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidation-Reduction/drug effects , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proteomics/methods
14.
Cells ; 9(4)2020 04 18.
Article in English | MEDLINE | ID: mdl-32325691

ABSTRACT

Aiming to promote cancer cell apoptosis is a mainstream strategy of cancer therapy. The second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis protein (IAP)-binding protein with low pI (DIABLO) protein is an essential and endogenous antagonist of inhibitor of apoptosis proteins (IAPs). SMAC mimetics (SMs) are a series of synthetically chemical compounds. Via database analysis and literature searching, we summarize the potential mechanisms of endogenous SMAC inefficiency, degradation, mutation, releasing blockage, and depression. We review the development of SMs, as well as preclinical and clinical outcomes of SMs in solid tumor treatment, and we analyze their strengths, weaknesses, opportunities, and threats from our point of view. We also highlight several questions in need of further investigation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Animals , Apoptosis Regulatory Proteins/drug effects , Caspases/drug effects , Caspases/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Neoplasms/metabolism
15.
Int J Neurosci ; 130(2): 161-169, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31516040

ABSTRACT

Purpose: Evidence has shown that propofol may cause widespread apoptotic neurodegeneration. Hypoxic preconditioning has been demonstrated to provide neuroprotection and brain recovery from both acute and chronic neurodegeneration in several cellular and animal models. However, the mechanism has not been well elucidated. Therefore, the present study was designed to investigate the expression of glucose transporters (GLUT1 and GLUT3) and mitochondrial division and fusion (Drp1 and Mfn2) proteins in rats exposed to hypoxic preconditioning to attenuate propofol neurotoxicity.Methods: Propofol (100 mg/kg) was given to 7-day-old Sprague-Dawley rats; in some rats, hypoxic preconditioning was administered before intraperitoneal propofol injection by subjecting rats to five cycles of 10 min of hypoxia (8% O2) and 10 min of normoxia (21% O2). Then, the rats were allowed to breathe room air for 2 h. Neuronal mitochondrial morphology was observed by transmission electron microscopy. ATP content was detected using an ATP assay kit. The expression levels of GLUT1, GLUT3, pDrp1, Drp1 and Mfn2 were detected by Western blot, and the expression levels of GLUT1 and GLUT3 were further examined by immunohistochemistry.Results: Propofol damaged mitochondria, and decreased ATP content and GLUT3 and pDrp1 protein expression. However, our results suggested that hypoxic preconditioning could attenuate propofol neurotoxicity by reducing mitochondrial damage and increasing ATP content and pDrp1, GLUT1 and GLUT3 protein expression.Conclusion: Hypoxic preconditioning reduced propofol-induced damage in the hippocampus of neonatal rats by attenuating the increase in mitochondrial division and decrease in GLUT3 expression.


Subject(s)
Dynamins , GTP Phosphohydrolases , Glucose Transporter Type 1 , Glucose Transporter Type 3 , Hippocampus , Hypnotics and Sedatives/toxicity , Hypoxia, Brain , Mitochondria , Mitochondrial Proteins , Neurons , Neurotoxicity Syndromes/prevention & control , Propofol/toxicity , Animals , Animals, Newborn , Disease Models, Animal , Dynamins/drug effects , Dynamins/metabolism , GTP Phosphohydrolases/drug effects , GTP Phosphohydrolases/metabolism , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/drug effects , Glucose Transporter Type 3/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypnotics and Sedatives/administration & dosage , Hypoxia, Brain/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Propofol/administration & dosage , Rats , Rats, Sprague-Dawley
16.
Toxicol Lett ; 319: 66-73, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31726083

ABSTRACT

Thallium ion (Tl+) and its neurotoxic products are widely known to cause severe neurological complications. However, the exact mechanism of action remains unknown, with limited therapeutic options available. This study aims to examine the toxic effects of Thallium (I) Nitrate (TlNO3) on primary hippocampal neurons of E17-E18 Wistar rat embryos, and the potential neuroprotective role of Nrf2- Keap1 signaling pathway against thallium-induced oxidative stress and mitochondrial dysfunction. TlNO3 induces a significant increase in reactive oxygen species levels and mitochondrial dysfunction in primary hippocampal neurons. Furthermore, the Nrf2-Keap1 signaling pathway played a protective role against TlNO3-induced hippocampal neuronal cytotoxicity. Moreover, mitochondrial fusion protein Mitofusin 2 (Mfn2) levels significantly decreased in hippocampal neurons when exposed to TlNO3, indicating that Mfn2 protein levels are linked to TlNO3-induced neurotoxicity. t-BHQ, a Nrf2 and phase II detoxification enzyme inducer, counteracted the oxidative damage in hippocampal neurons by activating the Nrf2-Keap1 signaling pathway after TlNO3 exposure; the activated Nrf2-Keap1 pathway could then maintain Mfn2 function by regulating Mfn2 protein expression. Thus, Nrf2-Keap1 pathway activation plays a protective role in Tl+-induced brain damage, and specific agonists have been identified to have great potential for treating thallium poisoning.


Subject(s)
Hippocampus/drug effects , Kelch-Like ECH-Associated Protein 1/drug effects , Mitochondria/drug effects , NF-E2-Related Factor 2/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Thallium/toxicity , Animals , Brain Diseases/chemically induced , Brain Diseases/pathology , Brain Diseases/prevention & control , Female , GTP Phosphohydrolases , Hippocampus/cytology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Primary Cell Culture , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
17.
Physiol Rep ; 7(19): e14266, 2019 10.
Article in English | MEDLINE | ID: mdl-31599131

ABSTRACT

Clenbuterol induces a slow-to-fast fiber type transition in skeletal muscle. This muscle fiber transition decreased mitochondrial oxidative capacity and respiratory function. We hypothesized that the clenbuterol-mediated reduction in oxidative capacity is associated with the alteration in mitochondrial morphology. To verify this hypothesis, we examined whether clenbuterol alters mitochondrial morphology and mitochondrial regulatory proteins in rat skeletal muscle. Clenbuterol was administered to rats via drinking water (30 mg/L) for 3 weeks. Myosin heavy chain (MHC) isoform composition, mitochondrial morphology, and fusion and fission regulatory protein levels in deep region and superficial region in tibialis anterior (TA) muscles were assessed. Clenbuterol induced the fiber type transition from slow to fast in both the regions of TA. The levels of optic atrophy protein 1, mitofusin 2, and mitochondrial fission 1, but not of dynamin-related protein 1, significantly decreased in deep and superficial muscles after clenbuterol administration (P < 0.01). Also, observation using the transmission electron microscopy showed a decrease in mitochondrial volume (P < 0.05) and an increase in proportion of continuous or interacting mitochondria across Z-lines (P < 0.05). We showed that clenbuterol administration induces a transition in the muscle fiber type composition toward fast phenotype and causes alterations in mitochondrial morphology with a concomitant decrease in mitochondrial fusion and fission regulatory protein levels. These mitochondrial morphological alterations may influence deleterious effects on skeletal muscle metabolism.


Subject(s)
Adrenergic beta-Agonists/toxicity , Clenbuterol/toxicity , Mitochondria/drug effects , Mitochondrial Proteins/drug effects , Muscle, Skeletal/drug effects , Animals , Male , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Rats , Rats, Wistar
18.
Toxicon ; 168: 141-146, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31356822

ABSTRACT

Ochratoxin A (OTA) is a mycotoxin ubiquitous in feeds and foodstuffs. The water-insoluble pentacyclic triterpene bioactive compound, ursolic acid (UA), is widespread in various cuticular waxes of edible fruits, food materials, and medicinal plants. Although studies have reported that oxidative stress was involved in both the nephrotoxicity of OTA and the renoprotective function of UA, the role of stress-responsive Lon protease 1 (Lonp1) in the renoprotection of UA against OTA is still unknown. In this study, cell viability, reactive oxygen species (ROS) production, and several proteins' expressions of human embryonic kidney 293T (HEK293T) cells in response to UA, OTA, and/or Lonp1 inhibitor CDDO-me treatment were detected to reveal the protective mechanism of UA against OTA-induced renal cytotoxicity. Results indicated that a 2 h-treatment of 1 µM UA could significantly alleviate the ROS production and cell death induced by a 24 h-treatment of 8 µM OTA in HEK293T cells (P < 0.05). Compared with the control, the protein expressions of Lonp1, Aco2 and Hsp75 were significantly inhibited after 8 µM OTA treating for 24 h (P < 0.05), which could be notably reversed by the pre-treatment and post-treatment of 1 µM UA (P < 0.05). The protein expressions of Lonp1, Aco2 and Hsp75 were inhibited by the addition of CDDO-me. The three protein expression trends were similar before and after the addition of CDDO-me. In conclusion, OTA could inhibit the expression of Lonp1, suppressing Aco2 and Hsp75 as a result, thereby activating ROS and inducing cell death in HEK293T cells, which could be alleviated by UA pre-treatment.


Subject(s)
Kidney/drug effects , Ochratoxins/toxicity , Triterpenes/pharmacology , ATP-Dependent Proteases/drug effects , Aconitate Hydratase/drug effects , Cell Survival/drug effects , HEK293 Cells , HSP90 Heat-Shock Proteins/drug effects , Humans , Kidney/metabolism , Mitochondrial Proteins/drug effects , Reactive Oxygen Species/metabolism , Ursolic Acid
20.
PLoS One ; 13(12): e0208973, 2018.
Article in English | MEDLINE | ID: mdl-30586434

ABSTRACT

Dietary macronutrient composition alters metabolism through several mechanisms, including post-translational modification (PTM) of proteins. To connect diet and molecular changes, here we performed short- and long-term feeding of mice with standard chow diet (SCD) and high-fat diet (HFD), with or without glucose or fructose supplementation, and quantified liver metabolites, 861 proteins, and 1,815 protein level-corrected mitochondrial acetylation and succinylation sites. Nearly half the acylation sites were altered by at least one diet; nutrient-specific changes in protein acylation sometimes encompass entire pathways. Although acetyl-CoA is an intermediate in both sugar and fat metabolism, acetyl-CoA had a dichotomous fate depending on its source; chronic feeding of dietary sugars induced protein hyperacetylation, whereas the same duration of HFD did not. Instead, HFD resulted in citrate accumulation, anaplerotic metabolism of amino acids, and protein hypo-succinylation. Together, our results demonstrate novel connections between dietary macronutrients, protein post-translational modifications, and regulation of fuel selection in liver.


Subject(s)
Fatty Liver/metabolism , Liver/metabolism , Mitochondria, Liver/drug effects , Mitochondrial Proteins/genetics , Acetylation/drug effects , Animals , Citric Acid/metabolism , Diet, High-Fat/adverse effects , Fatty Liver/genetics , Fatty Liver/pathology , Glucose/metabolism , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Proteins/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...