Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 760
1.
Circ Res ; 134(10): 1292-1305, 2024 May 10.
Article En | MEDLINE | ID: mdl-38618716

BACKGROUND: During myocardial ischemia/reperfusion (I/R) injury, high levels of matrix Ca2+ and reactive oxygen species (ROS) induce the opening of the mitochondrial permeability transition pore (mPTP), which causes mitochondrial dysfunction and ultimately necrotic death. However, the mechanisms of how these triggers individually or cooperatively open the pore have yet to be determined. METHODS: Here, we use a combination of isolated mitochondrial assays and in vivo I/R surgery in mice. We challenged isolated liver and heart mitochondria with Ca2+, ROS, and Fe2+ to induce mitochondrial swelling. Using inhibitors of the mPTP (cyclosporine A or ADP) lipid peroxidation (ferrostatin-1, MitoQ), we determined how the triggers elicit mitochondrial damage. Additionally, we used the combination of inhibitors during I/R injury in mice to determine if dual inhibition of these pathways is additivity protective. RESULTS: In the absence of Ca2+, we determined that ROS fails to trigger mPTP opening. Instead, high levels of ROS induce mitochondrial dysfunction and rupture independently of the mPTP through lipid peroxidation. As expected, Ca2+ in the absence of ROS induces mPTP-dependent mitochondrial swelling. Subtoxic levels of ROS and Ca2+ synergize to induce mPTP opening. Furthermore, this synergistic form of Ca2+- and ROS-induced mPTP opening persists in the absence of CypD (cyclophilin D), suggesting the existence of a CypD-independent mechanism for ROS sensitization of the mPTP. These ex vivo findings suggest that mitochondrial dysfunction may be achieved by multiple means during I/R injury. We determined that dual inhibition of the mPTP and lipid peroxidation is significantly more protective against I/R injury than individually targeting either pathway alone. CONCLUSIONS: In the present study, we have investigated the relationship between Ca2+ and ROS, and how they individually or synergistically induce mitochondrial swelling. Our findings suggest that Ca2+ mediates mitochondrial damage through the opening of the mPTP, although ROS mediates its damaging effects through lipid peroxidation. However, subtoxic levels both Ca2+ and ROS can induce mPTP-mediated mitochondrial damage. Targeting both of these triggers to preserve mitochondria viability unveils a highly effective therapeutic approach for mitigating I/R injury.


Lipid Peroxidation , Mice, Inbred C57BL , Mitochondria, Heart , Mitochondria, Liver , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury , Reactive Oxygen Species , Animals , Lipid Peroxidation/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Reactive Oxygen Species/metabolism , Mice , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondria, Liver/drug effects , Calcium/metabolism , Mitochondrial Swelling/drug effects
2.
Liver Int ; 43(11): 2365-2378, 2023 11.
Article En | MEDLINE | ID: mdl-37615254

This thematic review aims to provide an overview of the current state of knowledge about the occurrence of giant mitochondria or megamitochondria in liver parenchymal cells. Their presence and accumulation are considered to be a major pathological hallmark of the health and fate of liver parenchymal cells that leads to overall tissue deterioration and eventually results in organ failure. The first description on giant mitochondria dates back to the 1960s, coinciding with the availability of the first generation of electron microscopes in clinical diagnostic laboratories. Detailed accounts on their ultrastructure have mostly been described in patients suffering from alcoholic liver disease, chronic hepatitis, hepatocellular carcinoma and non-alcoholic fatty liver disease. Interestingly, from this extensive literature survey, it became apparent that giant mitochondria or megamitochondria present themselves with or without highly organised crystal-like intramitochondrial inclusions. The origin, formation and potential role of giant mitochondria remain to-date largely unanswered. Likewise, the biochemical composition of the well-organised crystal-like inclusions and their possible impact on mitochondrial function is unclear. Herein, concepts about the possible mechanism of their formation and three-dimensional architecture will be approached. We will furthermore discuss their importance in diagnostics, including future research outlooks and potential therapeutic interventions to cure liver disease where giant mitochondria are implemented.


Liver Diseases, Alcoholic , Non-alcoholic Fatty Liver Disease , Humans , Mitochondrial Swelling , Mitochondria, Liver/pathology , Liver Diseases, Alcoholic/pathology , Non-alcoholic Fatty Liver Disease/pathology , Hepatitis, Chronic/pathology , Liver/pathology
3.
J Biol Chem ; 298(6): 102008, 2022 06.
Article En | MEDLINE | ID: mdl-35513069

Mitochondrial ATPase ATAD3A is essential for cholesterol transport, mitochondrial structure, and cell survival. However, the relationship between ATAD3A and nonalcoholic fatty liver disease (NAFLD) is largely unknown. In this study, we found that ATAD3A was upregulated in the progression of NAFLD in livers from rats with diet-induced nonalcoholic steatohepatitis and in human livers from patients diagnosed with NAFLD. We used CRISPR-Cas9 to delete ATAD3A in Huh7 human hepatocellular carcinoma cells and used RNAi to silence ATAD3A expression in human hepatocytes isolated from humanized liver-chimeric mice to assess the influence of ATAD3A deletion on liver cells with free cholesterol (FC) overload induced by treatment with cholesterol plus 58035, an inhibitor of acetyl-CoA acetyltransferase. Our results showed that ATAD3A KO exacerbated FC accumulation under FC overload in Huh7 cells and also that triglyceride levels were significantly increased in ATAD3A KO Huh7 cells following inhibition of lipolysis mediated by upregulation of lipid droplet-binding protein perilipin-2. Moreover, loss of ATAD3A upregulated autophagosome-associated light chain 3-II protein and p62 in Huh7 cells and fresh human hepatocytes through blockage of autophagosome degradation. Finally, we show the mitophagy mediator, PTEN-induced kinase 1, was downregulated in ATAD3A KO Huh7 cells, suggesting that ATAD3A KO inhibits mitophagy. These results also showed that loss of ATAD3A impaired mitochondrial basal respiration and ATP production in Huh7 cells under FC overload, accompanied by downregulation of mitochondrial ATP synthase. Taken together, we conclude that loss of ATAD3A promotes the progression of NAFLD through the accumulation of FC, triglyceride, and damaged mitochondria in hepatocytes.


ATPases Associated with Diverse Cellular Activities , Non-alcoholic Fatty Liver Disease , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Hepatocytes/enzymology , Humans , Liver/enzymology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria, Liver/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/enzymology , Rats , Triglycerides/metabolism
4.
Biochim Biophys Acta Biomembr ; 1864(1): 183776, 2022 02 01.
Article En | MEDLINE | ID: mdl-34547253

Alpha-synuclein (α-syn) aggregation and mitochondrial dysfunction are considered as two of the main factors associated with Parkinson's disease (PD). In the present investigation, the effectiveness of the amyloid fibrils obtained from α-syn with those of hen egg white lysozyme (HEWL), as disease-related and-unrelated proteins, to damage rat brain and rat liver mitochondria have been investigated. This was extended by looking at SH-SY5Y human neuroblastoma cells and erythrocytes, thereby investigating the significance of structural characteristics of amyloid fibrils related to their interactions with biomembranes obtained from various sources. Results presented clearly demonstrate substantial differences in the response of tested biomembranes to toxicity induced by α-syn/HEWL amyloid fibrils, highlighting a structure-function relationship. We found that fibrillar aggregates of α-syn, but not HEWL, caused a significant increase in mitochondrial ROS, loss of membrane potential, and mitochondrial swelling, in a dose-dependent manner. Toxicity was found to be more pronounced in brain mitochondria, as compared to liver mitochondria. For SH-SY5Y cells and erythrocytes, however, both α-syn and HEWL amyloid fibrils showed the capacity to induce toxicity. Taken together, these results may suggest selective toxicity of α-syn amyloid fibrils to mitochondria mediated likely by their direct interaction with the outer mitochondrial membrane, indicating a correlation between specific structural characteristics of α-syn fibrils and an organelle strongly implicated in PD pathology.


Amyloid/chemistry , Brain/drug effects , Mitochondria, Liver/drug effects , alpha-Synuclein/chemistry , Amyloid/pharmacology , Animals , Brain/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Chickens , Egg White/chemistry , Erythrocytes/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/pathology , Muramidase/chemistry , Muramidase/pharmacology , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats , Structure-Activity Relationship , alpha-Synuclein/genetics
5.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166298, 2022 01 01.
Article En | MEDLINE | ID: mdl-34751152

In the diagnostic work-up of a newborn infant with a metabolic crisis, lethal multiorgan failure on day six of life, and increased excretion of 3-methylglutaconic acid, we found using whole genome sequencing a homozygous SERAC1 mutation indicating MEGDHEL syndrome (3-methylglutaconic aciduria with deafness-dystonia, hepatopathy, encephalopathy, and Leigh-like syndrome). The SERAC1 protein is located at the contact site between mitochondria and the endoplasmic reticulum (ER) and is crucial for cholesterol trafficking. Our aim was to investigate the effect of the homozygous truncating mutation on mitochondrial structure and function. In the patient fibroblasts, no SERAC1 protein was detected, the mitochondrial network was severely fragmented, and the cristae morphology was altered. Filipin staining showed uneven localization of unesterified cholesterol. The calcium buffer function between cytoplasm and mitochondria was deficient. In liver mitochondria, complexes I, III, and IV were clearly decreased. In transfected COS-1 cells the mutant protein with the a 45-amino acid C-terminal truncation was distributed throughout the cell, whereas wild-type SERAC1 partially colocalized with the mitochondrial marker MT-CO1. The structural and functional mitochondrial abnormalities, caused by the loss of SERAC1, suggest that the crucial disease mechanism is disrupted interplay between the ER and mitochondria leading to decreased influx of calcium to mitochondria and secondary respiratory chain deficiency.


Carboxylic Ester Hydrolases/genetics , Metabolism, Inborn Errors/genetics , Mitochondria, Liver/genetics , Mitochondrial Diseases/genetics , Calcium/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Female , Glutarates/metabolism , Humans , Infant, Newborn , Male , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Whole Genome Sequencing
6.
Int J Mol Sci ; 22(21)2021 Oct 30.
Article En | MEDLINE | ID: mdl-34769236

Galactic cosmic rays are primarily composed of protons (85%), helium (14%), and high charge/high energy ions (HZEs) such as 56Fe, 28Si, and 16O. HZE exposure is a major risk factor for astronauts during deep-space travel due to the possibility of HZE-induced cancer. A systems biology integrated omics approach encompassing transcriptomics, proteomics, lipidomics, and functional biochemical assays was used to identify microenvironmental changes induced by HZE exposure. C57BL/6 mice were placed into six treatment groups and received the following irradiation treatments: 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), 350 MeV/n 28Si (0.2 Gy), 137Cs (1.0 Gy) gamma rays, 137Cs (3.0 Gy) gamma rays, and sham irradiation. Left liver lobes were collected at 30, 60, 120, 270, and 360 days post-irradiation. Analysis of transcriptomic and proteomic data utilizing ingenuity pathway analysis identified multiple pathways involved in mitochondrial function that were altered after HZE irradiation. Lipids also exhibited changes that were linked to mitochondrial function. Molecular assays for mitochondrial Complex I activity showed significant decreases in activity after HZE exposure. HZE-induced mitochondrial dysfunction suggests an increased risk for deep space travel. Microenvironmental and pathway analysis as performed in this research identified possible targets for countermeasures to mitigate risk.


Cosmic Radiation/adverse effects , Electron Transport Complex I/metabolism , Gamma Rays/adverse effects , Liver/enzymology , Mitochondria, Liver/enzymology , Radiation Injuries, Experimental/enzymology , Animals , Dose-Response Relationship, Radiation , Liver/pathology , Male , Mice , Mitochondria, Liver/pathology , Proteomics , Radiation Injuries, Experimental/pathology , Space Flight
7.
Toxicol Appl Pharmacol ; 432: 115758, 2021 12 01.
Article En | MEDLINE | ID: mdl-34678374

Mitochondrial dysfunction is a major factor in nonalcoholic fatty liver disease (NAFLD), preceding insulin resistance and hepatic steatosis. Carnosol (CAR) is a kind of diterpenoid with antioxidant, anti-inflammatory and antitumor activities. Peroxiredoxin 3 (PRDX3), a mitochondrial H2O2-eliminating enzyme, undergoes overoxidation and subsequent inactivation under oxidative stress. The purpose of this study was to investigate the protective effect of the natural phenolic compound CAR on NAFLD via PRDX3. Mice fed a high-fat diet (HFD) and AML-12 cells treated with palmitic acid (PA) were used to detect the molecular mechanism of CAR in NAFLD. We found that pharmacological treatment with CAR notably moderated HFD- and PA- induced steatosis and liver injury, as shown by biochemical assays, Oil Red O and Nile Red staining. Further mechanistic investigations revealed that CAR exerted anti-NAFLD effects by inhibiting mitochondrial oxidative stress, perturbation of mitochondrial dynamics, and apoptosis in vivo and in vitro. The decreased protein and mRNA levels of PRDX3 were accompanied by intense oxidative stress after PA intervention. Interestingly, CAR specifically bound PRDX3, as shown by molecular docking assays, and increased the expression of PRDX3. However, the hepatoprotection of CAR in NAFLD was largely abolished by specific PRDX3 siRNA, which increased mitochondrial dysfunction and exacerbated apoptosis in vitro. In conclusion, CAR suppressed lipid accumulation, mitochondrial dysfunction and hepatocyte apoptosis by activating PRDX3, mitigating the progression of NAFLD, and thus, CAR may represent a promising candidate for clinical treatment of steatosis.


Abietanes/pharmacology , Apoptosis/drug effects , Enzyme Activators/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Peroxiredoxin III/metabolism , Animals , Antioxidants/pharmacology , Cell Line , Diet, High-Fat , Disease Models, Animal , Enzyme Activation , Hepatocytes/enzymology , Hepatocytes/pathology , Liver/enzymology , Liver/pathology , Male , Mice, Inbred C57BL , Mitochondria, Liver/enzymology , Mitochondria, Liver/pathology , Mitochondrial Dynamics/drug effects , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Palmitic Acid/toxicity , Peroxiredoxin III/genetics
8.
Immunol Lett ; 240: 24-30, 2021 12.
Article En | MEDLINE | ID: mdl-34525396

The mitochondria are the primary source of reactive oxygen species (ROS) under pathological condition, but the significance of mitochondrial ROS in the development of Lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced acute liver injury remains unclear. In the present study, the level of mitochondrial ROS in LPS/D-Gal has been determined by MitoSox staining and the potential roles of mitochondrial ROS in LPS/D-Gal-induced liver injury have been investigated by using the mitochondria-targeting antioxidant MitoQ. The results indicated that LPS/D-Gal exposure induced the generation of mitochondrial ROS while treatment with MitoQ reduced the level of mitochondrial ROS. Treatment with MitoQ ameliorated LPS/D-Gal-induced histopathologic abnormalities, suppressed the elevation of AST and ALT, and increased the survival rate of the experimental animals. Treatment with MitoQ also suppressed LPS/D-Gal-induced production of tumor necrosis factor α (TNF-α), inhibited the activities of caspase-3, caspase-8 and caspase-9, decreased the level of cleaved caspase-3 and reduced the counts of TUNEL positive cells. These results indicate that mitochondrial ROS is involved in the development of LPS-induced acute liver injury and the mitochondria-targeting antioxidant MitoQ might have potential value for the treatment of inflammation-based acute liver injury.


Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/metabolism , Galactosamine/toxicity , Lipopolysaccharides/toxicity , Mitochondria, Liver/metabolism , Organophosphorus Compounds/pharmacology , Ubiquinone/analogs & derivatives , Animals , Chemical and Drug Induced Liver Injury/pathology , Male , Mice , Mice, Inbred BALB C , Mitochondria, Liver/pathology , Ubiquinone/pharmacology
9.
Biomed Pharmacother ; 143: 112172, 2021 Nov.
Article En | MEDLINE | ID: mdl-34560548

BACKGROUND: Renal ischemia-reperfusion injury (IRI) causes acute kidney injury as well as liver injury. Renal IRI depletes hepatic antioxidants, promotes hepatic inflammation and dysfunction through Tlr9 upregulation. There is no treatment available for liver injury during renal IRI. This study examines the hepatoprotective role of treprostinil, a prostacyclin analog, during renal IRI. METHODS: Male Sprague-Dawley rats were divided into four groups: control, sham, IRI-placebo, or IRI-treprostinil and subjected to bilateral ischemia (45 min) followed by reperfusion (1-72 h). Placebo or treprostinil (100 ng/kg/min) was administered subcutaneously via an osmotic minipump. RESULTS: Treprostinil significantly reduced peak serum creatinine, BUN, ALT and AST levels vs. IRI-placebo. Treprostinil also restored hepatic levels of superoxide dismutase, glutathione, catalase, and Gclc expression to baseline, while reducing lipid peroxidation vs. IRI-placebo. Additionally, treprostinil significantly reduced elevated hepatic Tlr9, Il-1ß, Ccl2, Vcam1, and Serpine1 mRNA expression. Renal IRI increased hepatic apoptosis which was inhibited by treprostinil through reduced cytochrome c and cleaved caspase-3 protein expression. Treprostinil enhanced hepatic ATP concentrations and mitochondrial DNA copy number and improved mitochondrial dynamics by restoring Pgc-1α expression and significantly upregulating Mfn1, Mfn2, and Sirt3 levels, while reducing Drp-1 protein vs. IRI-placebo. Non-targeted semi-quantitative proteomics showed improved oxidative stress indices and ATP subunits in the IRI-treprostinil group. CONCLUSIONS: Treprostinil improved hepatic function and antioxidant levels, while suppressing the inflammatory response and alleviating Tlr9-mediated apoptotic injury during renal IRI. Our study provides evidence of treprostinil's hepatoprotective effect, which supports the therapeutic potential of treprostinil in reducing hepatic injury during renal IRI.


Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Epoprostenol/analogs & derivatives , Hepatitis/prevention & control , Liver/drug effects , Mitochondria, Liver/drug effects , Reperfusion Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Epoprostenol/pharmacology , Hepatitis/metabolism , Hepatitis/pathology , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial Dynamics/drug effects , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Toll-Like Receptor 9/metabolism
10.
Life Sci ; 284: 119906, 2021 Nov 01.
Article En | MEDLINE | ID: mdl-34478761

The present study was performed to investigate the effects of Cd exposure on lipid metabolism and mitochondrial dysfunction and to explore the role of mitophagy in Cd-induced dysregulation of lipid metabolism in chicken embryo liver tissues and hepatocytes. To this end, seven-day-old chicken embryos were exposed to different concentrations of Cd for 7 days, and primary chicken embryo hepatocytes were treated with Cd at four different concentrations for 6 h. Furthermore, the mitophagy inhibitor cyclosporine A (CsA) was used to investigate the role of mitophagy in Cd-induced disruption of lipid metabolism. Lipid accumulation, the expression levels of genes involved in lipid metabolism, mitochondrial dysfunction, and mitophagy were measured. The results demonstrated that Cd exposure increases hepatic triglyceride (TG) accumulation and the expression levels of lipogenic genes while decreasing those of lipolytic genes. Furthermore, Cd exposure was observed to alter mitochondrial morphology in terms of reduced size, excessive mitochondrial damage, and the formation of mitophagosomes. The co-localization of lysosome-associated membrane glycoprotein 2 and LC3 puncta was significantly increased in primary chicken embryo hepatocytes after Cd exposure. Moreover, Cd exposure increased LC3, PINK1, and Parkin protein expression levels. CsA effectively alleviated Cd-induced mitochondrial dysfunction, blocked mitochondrial membrane potential collapse, and suppressed PINK1/Parkin-mediated mitophagy. Furthermore, CsA treatment reversed the Cd-induced TG accumulation in liver tissues but further increased it in hepatocytes. Taken together, our findings demonstrate (for the first time) the importance of mitochondrial dysfunction and mitophagy via the PINK1/Parkin pathway in Cd-induced disruption of lipid metabolism.


Cadmium/toxicity , Lipid Metabolism , Liver/metabolism , Mitochondria, Liver/pathology , Mitophagy , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Chick Embryo , Cyclosporine/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Lipid Metabolism/drug effects , Liver/drug effects , Liver/embryology , Mitochondria, Liver/drug effects , Mitochondria, Liver/ultrastructure , Mitophagy/drug effects , Models, Biological
11.
Biomed Pharmacother ; 143: 112072, 2021 Nov.
Article En | MEDLINE | ID: mdl-34464747

BACKGROUND AND PURPOSE: It has been shown that the antidiabetic drug metformin protects hepatocytes against toxicity by various stressors. Chronic or excessive consumption of diclofenac (DF) - a pain-relieving drug, leads to drug-induced liver injury via a mechanism involving mitochondrial damage and ultimately apoptotic death of hepatocytes. However, whether metformin protects against DF-induced toxicity is unknown. Recently, it was also shown that cAMP elevation is protective against DF-induced apoptotic death in hepatocytes, a protective effect primarily involving the downstream cAMP effector EPAC and preservation of mitochondrial function. This study therefore aimed at investigating whether metformin protects against DF-induced toxicity via cAMP-EPACs. EXPERIMENTAL APPROACH: Primary rat hepatocytes were exposed to 400 µmol/L DF. CE3F4 or ESI-O5 were used as EPAC-1 or 2 inhibitors respectively. Apoptosis was measured by caspase-3 activity and necrosis by Sytox green staining. Seahorse X96 assay was used to determine mitochondrial function. Mitochondrial reactive oxygen species (ROS) production was measured using MitoSox, mitochondrial MnSOD expression was determined by immunostaining and mitochondrial morphology (fusion and fission ratio) by 3D refractive index imaging. KEY RESULTS: Metformin (1 mmol/L) was protective against DF-induced apoptosis in hepatocytes. This protective effect was EPAC-dependent (mainly EPAC-2). Metformin restored mitochondrial morphology in an EPAC-independent manner. DF-induced mitochondrial dysfunction which was demonstrated by decreased oxygen consumption rate, an increased ROS production and a reduced MnSOD level, were all reversed by metformin in an EPAC-dependent manner. CONCLUSION AND IMPLICATIONS: Metformin protects hepatocytes against DF-induced toxicity via cAMP-dependent EPAC-2.


Antioxidants/pharmacology , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Cyclooxygenase Inhibitors/toxicity , Diclofenac/toxicity , Guanine Nucleotide Exchange Factors/metabolism , Hepatocytes/drug effects , Metformin/pharmacology , Mitochondria, Liver/drug effects , Animals , Caspase 3/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cyclic AMP/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Oxidative Stress/drug effects , Primary Cell Culture , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction
12.
Cells ; 10(8)2021 07 23.
Article En | MEDLINE | ID: mdl-34440632

Ischemia/reperfusion (I/R) injury unavoidably occurs during hepatic resection and transplantation. Aged livers poorly tolerate I/R during surgical treatment. Although livers have a powerful endogenous inhibitor of calpains, calpastatin (CAST), I/R activates calpains, leading to impaired autophagy, mitochondrial dysfunction, and hepatocyte death. It is unknown how I/R in aged livers affects CAST. Human and mouse liver biopsies at different ages were collected during in vivo I/R. Hepatocytes were isolated from 3-month- (young) and 26-month-old (aged) mice, and challenged with short in vitro simulated I/R. Cell death, protein expression, autophagy, and mitochondrial permeability transition (MPT) between the two age groups were compared. Adenoviral vector was used to overexpress CAST. Significant cell death was observed only in reperfused aged hepatocytes. Before the commencement of ischemia, CAST expression in aged human and mouse livers and mouse hepatocytes was markedly greater than that in young counterparts. However, reperfusion substantially decreased CAST in aged human and mouse livers. In hepatocytes, reperfusion rapidly depleted aged cells of CAST, cleaved autophagy-related protein 5 (ATG5), and induced defective autophagy and MPT onset, all of which were blocked by CAST overexpression. Furthermore, mitochondrial morphology was shifted toward an elongated shape with CAST overexpression. In conclusion, CAST in aged livers is intrinsically short-lived and lost after short I/R. CAST depletion contributes to age-dependent liver injury after I/R.


Calcium-Binding Proteins/metabolism , Hepatocytes/metabolism , Liver Diseases/metabolism , Liver/metabolism , Reperfusion Injury/metabolism , Age Factors , Animals , Autophagy , Autophagy-Related Protein 5/metabolism , Calcium-Binding Proteins/genetics , Calpain/metabolism , Cell Death , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Hepatocytes/pathology , Humans , Liver/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Male , Mice, Inbred C57BL , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Time Factors
13.
Cells ; 10(8)2021 07 27.
Article En | MEDLINE | ID: mdl-34440674

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. HCC progression and metastasis are closely related to altered mitochondrial metabolism, including mitochondrial stress responses, metabolic reprogramming, and mitoribosomal defects. Mitochondrial oxidative phosphorylation (OXPHOS) defects and reactive oxygen species (ROS) production are attributed to mitochondrial dysfunction. In response to oxidative stress caused by increased ROS production, misfolded or unfolded proteins can accumulate in the mitochondrial matrix, leading to initiation of the mitochondrial unfolded protein response (UPRmt). The mitokines FGF21 and GDF15 are upregulated during UPRmt and their levels are positively correlated with liver cancer development, progression, and metastasis. In addition, mitoribosome biogenesis is important for the regulation of mitochondrial respiration, cell viability, and differentiation. Mitoribosomal defects cause OXPHOS impairment, mitochondrial dysfunction, and increased production of ROS, which are associated with HCC progression in mouse models and human HCC patients. In this paper, we focus on the role of mitochondrial metabolic signatures in the development and progression of HCC. Furthermore, we provide a comprehensive review of cell autonomous and cell non-autonomous mitochondrial stress responses during HCC progression and metastasis.


Carcinoma, Hepatocellular/metabolism , Energy Metabolism , Liver Neoplasms/metabolism , Metabolome , Mitochondria, Liver/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Disease Progression , Humans , Liver Neoplasms/pathology , Mitochondria, Liver/pathology , Mitochondrial Ribosomes/metabolism , Mitochondrial Ribosomes/pathology , Proteostasis , Reactive Oxygen Species/metabolism , Signal Transduction , Unfolded Protein Response
14.
J Biochem Mol Toxicol ; 35(9): e22846, 2021 Sep.
Article En | MEDLINE | ID: mdl-34250697

The liver is the primary organ affected by cholestasis. However, the brain, skeletal muscle, heart, and kidney are also severely influenced by cholestasis/cirrhosis. However, little is known about the molecular mechanisms of organ injury in cholestasis. The current study was designed to evaluate the mitochondrial glutathione redox state as a significant index in cell death. Moreover, tissue energy charge (EC) was calculated. Rats underwent bile duct ligation (BDL) and the brain, heart, liver, kidney, and skeletal muscle mitochondria were assessed at scheduled time intervals (3, 7, 14, and 28 days after BDL). A significant decrease in mitochondrial glutathione redox state and EC was detected in BDL animals. Moreover, disturbed mitochondrial indices were evident in different organs of BDL rats. These data could offer new insight into the mechanisms of organ injury and the source of oxidative stress during cholestasis and might provide novel therapeutic strategies against these complications.


Cholestasis/metabolism , Energy Metabolism , Mitochondria, Liver/metabolism , Mitochondria, Muscle/metabolism , Animals , Cholestasis/pathology , Disease Models, Animal , Male , Mitochondria, Liver/pathology , Mitochondria, Muscle/pathology , Organ Specificity , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
15.
Int J Mol Sci ; 22(11)2021 May 26.
Article En | MEDLINE | ID: mdl-34073582

Oxidative stress occurs in a variety of clinical liver diseases and causes cellular damage and mitochondrial dysfunction. The clearance of damaged mitochondria by mitophagy may facilitate mitochondrial biogenesis and enhance cell survival. Although the supplementation of docosahexaenoic acid (DHA) has been recognized to relieve the symptoms of various liver diseases, the antioxidant effect of DHA in liver disease is still unclear. The purpose of our research was to investigate the antioxidant effect of DHA in the liver and the possible role of mitophagy in this. In vitro, H2O2-induced injury was caused in AML12 cells. The results showed that DHA repressed the level of reactive oxygen species (ROS) induced by H2O2 and stimulated the cellular antioxidation response. Most notably, DHA restored oxidative stress-impaired autophagic flux and promoted protective autophagy. In addition, PINK/Parkin-mediated mitophagy was activated by DHA in AML12 cells and alleviated mitochondrial dysfunction. The ERK1/2 signaling pathway was inhibited during oxidative stress but reactivated by DHA treatment. It was proven that the expression of ERK1/2 was involved in the regulation of mitophagy by the ERK1/2 inhibitor. We further proved these results in vivo. DHA effectively alleviated the liver oxidative damage caused by CCl4 and enhanced antioxidation capacity; intriguingly, autophagy was also activated. In summary, our data demonstrated that DHA protected hepatocytes from oxidative damage through GPR120/ERK-mediated mitophagy.


Docosahexaenoic Acids/pharmacology , Hepatocytes/metabolism , MAP Kinase Signaling System/drug effects , Mitochondria, Liver/metabolism , Mitophagy/drug effects , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Hepatocytes/pathology , Hydrogen Peroxide/adverse effects , Hydrogen Peroxide/pharmacology , Male , Mice , Mitochondria, Liver/pathology , Oxidation-Reduction/drug effects
16.
Methods Mol Biol ; 2310: 201-246, 2021.
Article En | MEDLINE | ID: mdl-34096005

Nonalcoholic fatty liver disease (NAFLD) is one of the most frequent metabolic chronic liver diseases in developed countries and puts the populations at risk of progression to liver necro-inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma. Mitochondrial dysfunction is involved in the onset of NAFLD and contributes to the progression from NAFLD to nonalcoholic steatohepatitis (NASH). Thus, liver mitochondria could become the target for treatments for improving liver function in NAFLD patients. This chapter describes the most important steps used for potential therapeutic interventions in NAFLD patients, discusses current options gathered from both experimental and clinical evidence, and presents some novel options for potentially improving mitochondrial function in NAFLD.


Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Hypoglycemic Agents/therapeutic use , Liver Cirrhosis/drug therapy , Liver/drug effects , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Anti-Inflammatory Agents/adverse effects , Antioxidants/adverse effects , Apoptosis/drug effects , Humans , Hypoglycemic Agents/adverse effects , Inflammation Mediators/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Risk Reduction Behavior
17.
Methods Mol Biol ; 2277: 277-287, 2021.
Article En | MEDLINE | ID: mdl-34080157

Isolation of mitochondria is a crucial method for examining molecular details of this organelle's manifold functions. Historically, mitochondrial isolations required large amounts of sample material which impeded their isolation from cultured cells. We have therefore developed a method allowing for controlled and reproducible isolation of intact and functional mitochondria from diverse cell types in culture. Here we provide a methodological update of this approach together with a protocol for the subsequent analysis of such isolated mitochondria by electron microscopy. Combining the isolation procedure with this powerful imaging method can reveal ultrastructural mitochondrial peculiarities in disease settings that might not be evident in intact cells and allows for assessment of mitochondrial membrane integrity and sample purity.


Liver Neoplasms/pathology , Microscopy, Electron/methods , Mitochondria, Liver/pathology , Cell Line, Tumor , Cell Separation/instrumentation , Cell Separation/methods , Equipment Design , Hep G2 Cells , Humans , Image Processing, Computer-Assisted/methods
18.
Cell Death Dis ; 12(5): 442, 2021 05 04.
Article En | MEDLINE | ID: mdl-33947842

Despite N6-methyladenosine (m6A) is functionally important in various biological processes, its role and the underlying regulatory mechanism in the liver remain largely unexplored. In the present study, we showed that fat mass and obesity-associated protein (FTO, an m6A demethylase) was involved in mitochondrial function during hepatic ischemia-reperfusion injury (HIRI). We found that the expression of m6A demethylase FTO was decreased during HIRI. In contrast, the level of m6A methylated RNA was enhanced. Adeno-associated virus-mediated liver-specific overexpression of FTO (AAV8-TBG-FTO) ameliorated the HIRI, repressed the elevated level of m6A methylated RNA, and alleviated liver oxidative stress and mitochondrial fragmentation in vivo and in vitro. Moreover, dynamin-related protein 1 (Drp1) was a downstream target of FTO in the progression of HIRI. FTO contributed to the hepatic protective effect via demethylating the mRNA of Drp1 and impairing the Drp1-mediated mitochondrial fragmentation. Collectively, our findings demonstrated the functional importance of FTO-dependent hepatic m6A methylation during HIRI and provided valuable insights into the therapeutic mechanisms of FTO.


Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Dynamins/metabolism , Liver/blood supply , Mitochondria, Liver/metabolism , Reperfusion Injury/metabolism , Adenosine/metabolism , Animals , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/pathology , Reperfusion Injury/pathology
19.
J Biochem Mol Toxicol ; 35(7): e22780, 2021 Jul.
Article En | MEDLINE | ID: mdl-33957011

4-Nonylphenol (4-NP) is an emerging environmental pollutant widely diffused in waters and sediments. It mainly derives from the degradation of alkyl phenol ethoxylates, compounds commonly employed as industrial surfactants. 4-NP strongly contaminates foods and waters for human use; thus, it displays a wide range of toxic effects not only for aquatic organisms but also for mammals and humans. After ingestion through the diet, it tends to accumulate in body fluids and tissues. One of the main organs where 4-NP and its metabolites are concentrated is the liver, where it causes, even at low doses, oxidative stress and apoptosis. In the present study, we analyzed the effects of 4-NP on a human hepatic cell line (HepG2) to deepen the knowledge of its cytotoxic mechanism. We found that 4-NP, in a range of concentration from 50 to 100 µM, significantly reduced cell viability; it caused a partial block of proliferation and induced apoptosis with activation of caspase-3 and overexpression of p53. Moreover, 4-NP induced-apoptosis seemed to involve both an ER-stress response, with the appearance of high level of GRP78, CHOP and the spliced XBP1, and a dysregulation of mitochondrial physiology, characterized by an overexpression of main markers of mitochondrial dynamics. Our data support the idea that a daily consumption of 4-NP-contaminated foods may lead to local damages at the level of gastrointestinal system, including liver, with negative consequences for the organ physiology.


Apoptosis/drug effects , Cytotoxins/toxicity , Endoplasmic Reticulum Stress/drug effects , Liver/metabolism , Mitochondria, Liver/metabolism , Phenols/toxicity , Endoplasmic Reticulum Chaperone BiP , Hep G2 Cells , Humans , Liver/pathology , Mitochondria, Liver/pathology
20.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article En | MEDLINE | ID: mdl-33802177

Allograft ischemia during liver transplantation (LT) adversely affects the function of mitochondria, resulting in impairment of oxidative phosphorylation and compromised post-transplant recovery of the affected organ. Several preservation methods have been developed to improve donor organ quality; however, their effects on mitochondrial functions have not yet been compared. This study aimed to summarize the available data on mitochondrial effects of graft preservation methods in preclinical models of LT. Furthermore, a network meta-analysis was conducted to determine if any of these treatments provide a superior benefit, suggesting that they might be used on humans. A systematic search was conducted using electronic databases (EMBASE, MEDLINE (via PubMed), the Cochrane Central Register of Controlled Trials (CENTRAL) and Web of Science) for controlled animal studies using preservation methods for LT. The ATP content of the graft was the primary outcome, as this is an indicator overall mitochondrial function. Secondary outcomes were the respiratory activity of mitochondrial complexes, cytochrome c and aspartate aminotransferase (ALT) release. Both a random-effects model and the SYRCLE risk of bias analysis for animal studies were used. After a comprehensive search of the databases, 25 studies were enrolled in the analysis. Treatments that had the most significant protective effect on ATP content included hypothermic and subnormothermic machine perfusion (HMP and SNMP) (MD = -1.0, 95% CI: (-2.3, 0.3) and MD = -1.1, 95% CI: (-3.2, 1.02)), while the effects of warm ischemia (WI) without cold storage (WI) and normothermic machine perfusion (NMP) were less pronounced (MD = -1.8, 95% CI: (-2.9, -0.7) and MD = -2.1 MD; CI: (-4.6; 0.4)). The subgroup of static cold storage (SCS) with shorter preservation time (< 12 h) yielded better results than SCS ≥ 12 h, NMP and WI, in terms of ATP preservation and the respiratory capacity of complexes. HMP and SNMP stand out in terms of mitochondrial protection when compared to other treatments for LT in animals. The shorter storage time at lower temperatures, together with the dynamic preservation, provided superior protection for the grafts in terms of mitochondrial function. Additional clinical studies on human patients including marginal donors and longer ischemia times are needed to confirm any superiority of preservation methods with respect to mitochondrial function.


Delayed Graft Function , Liver Transplantation , Mitochondria, Liver , Organ Preservation , Warm Ischemia , Animals , Delayed Graft Function/metabolism , Delayed Graft Function/pathology , Delayed Graft Function/prevention & control , Humans , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology
...