Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 464
Filter
1.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 311-318, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710515

ABSTRACT

Objective To investigate the effects of mitochondrial transcription factor A (TFAM) on mitochondrial function, autophagy, proliferation, invasion, and migration in cervical cancer HeLa cells and osteosarcoma U2OS cells. Methods TFAM small-interfering RNA (si-TFAM) was transfected to HeLa and U2OS cells for downregulating TFAM expression. Mito-Tracker Red CMXRos staining combined with laser confocal microscopy was used to detect mitochondrial membrane potential (MMP). MitoSOXTM Red labeling was used to test mitochondrial reactive oxygen species (mtROS) levels. The expression of mitochondrial DNA (mtDNA) was detected by real-time quantitative PCR. Changes in the number of autophagosomes were detected by immunofluorescence cytochemistry. Western blot analysis was used to detect the expressions of TFAM, autophagy microtubule associated protein 1 light chain 3A/B (LC3A/B), autophagy associated protein 2A (ATG2A), ATG2B, ATG9A, zinc finger transcription factor Snail, matrix metalloproteinase 2 (MMP2) and MMP9. CCK-8 assay and plate clony formation assay were used to detect cell proliferation, while TranswellTM assay and scratch healing assay were used to detect changes in cell invasion and migration. Results The downregulation of TFAM expression resulted in a decrease in MMP and mtDNA copy number, but an increase in mtROS production. The protein content of LC3A/B decreased significantly compared to the control group and the number of autophagosomes in the cytoplasm decreased significantly. The expressions of ATG2B and ATG9A in the early stage of autophagy were significantly reduced. The expressions of Snail, MMP2 and MMP9 proteins in HeLa and U2OS cells were also decreased. The proliferation, invasion and migration ability of HeLa and U2OS cells were inhibited after being interfered with TFAM expression. Conclusion Downregulation of TFAM expression inhibits mitochondrial function, delays autophagy process and reduces the proliferation, invasion and migration ability of cervical cancer cells and osteosarcoma cells.


Subject(s)
Autophagy , Cell Movement , Cell Proliferation , DNA-Binding Proteins , Mitochondrial Proteins , Neoplasm Invasiveness , Osteosarcoma , Transcription Factors , Uterine Cervical Neoplasms , Humans , Cell Movement/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Cell Proliferation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Autophagy/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Female , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Membrane Potential, Mitochondrial/genetics , Reactive Oxygen Species/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Mitochondria/metabolism , Mitochondria/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , HeLa Cells , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics
2.
Pathol Res Pract ; 258: 155337, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735276

ABSTRACT

BACKGROUND: Inappropriate expressions of various miRNAs have reported in different human malignancies. Evidence suggested that miR-330 may play as both onco-miR and/or tumor suppressor-miR in different cancers. In the present study, we evaluated effects of miR-330 on proliferation and migration of pancreatic cancer (PC) cells as well as underlying molecular mechanisms. DESIGN: The expression of miR-330 was evaluated in clinical tissue samples of patients with PC. Transfection of the PC cells (PANC-1) by miR-330 was conducted by pCMV vector. The cancer-related genes expression was investigated in mRNA and protein level following transfection of the PC cells. Furthermore, the PC cells viability, invasion, migration, mitochondrial membrane potential, apoptosis, autophagy, and cell cycle profile were investigated after transfection by miR-330. RESULTS: The results indicated that expression of miR-330 downregulated in patients with PC. Stable increase of miR-330 expression after transfection in PC cells reduces viability, mitochondrial membrane potential, invasion, and migration. Further assessments demonstrated that upregulation of miR-330 increases apoptosis and autophagy percentage in the PC cells. Moreover, a cell cycle arrest was observed in G1, Sub-G1, and S phases following transfection of the PC cells. These findings can be explained by modified mRNA and protein expression of apoptosis- and metastasis-related genes. CONCLUSION: Our study suggested that miR-330 acts as a tumor suppressor in PC cells, and revealed that upregulation of miR-330 may provide an effective therapeutic approach for overcoming progression and metastasis in patients with PC.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs , Pancreatic Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/genetics , Cell Line, Tumor , Carcinogenesis/genetics , Autophagy/genetics , Male , Female , Middle Aged , Membrane Potential, Mitochondrial/genetics
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(2): 184-193, 2024 Apr 25.
Article in English, Chinese | MEDLINE | ID: mdl-38562030

ABSTRACT

OBJECTIVES: To investigate the role of m.4435A>G and YARS2 c.572G>T (p.G191V) mutations in the development of essential hypertension. METHODS: A hypertensive patient with m.4435A>G and YARS2 p.G191V mutations was identified from previously collected mitochondrial genome and exon sequencing data. Clinical data were collected, and a molecular genetic study was conducted in the proband and his family members. Peripheral venous blood was collected, and immortalized lymphocyte lines constructed. The mitochondrial transfer RNA (tRNA), mitochondrial protein, adenosine triphosphate (ATP), mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) in the constructed lymphocyte cell lines were measured. RESULTS: Mitochondrial genome sequencing showed that all maternal members carried a highly conserved m.4435A>G mutation. The m.4435A>G mutation might affect the secondary structure and folding free energy of mitochondrial tRNA and change its stability, which may influence the anticodon ring structure. Compared with the control group, the cell lines carrying m.4435A>G and YARS2 p.G191V mutations had decreased mitochondrial tRNA homeostasis, mitochondrial protein expression, ATP production and MMP levels, as well as increased ROS levels (all P<0.05). CONCLUSIONS: The YARS2 p.G191V mutation aggravates the changes in mitochondrial translation and mitochondrial function caused by m.4435A>G through affecting the steady-state level of mitochondrial tRNA and further leads to cell dysfunction, indicating that YARS2 p.G191V and m.4435A>G mutations have a synergistic effect in this family and jointly participate in the occurrence and development of essential hypertension.


Subject(s)
Essential Hypertension , Mutation , Humans , Essential Hypertension/genetics , Male , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , RNA, Transfer/genetics , RNA, Transfer, Met/genetics , Genome, Mitochondrial , Female
4.
J Cell Sci ; 136(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36763487

ABSTRACT

Mitochondria and peroxisomes are dynamic signaling organelles that constantly undergo fission, driven by the large GTPase dynamin-related protein 1 (DRP1; encoded by DNM1L). Patients with de novo heterozygous missense mutations in DNM1L present with encephalopathy due to defective mitochondrial and peroxisomal fission (EMPF1) - a devastating neurodevelopmental disease with no effective treatment. To interrogate the mechanisms by which DRP1 mutations cause cellular dysfunction, we used human-derived fibroblasts from patients who present with EMPF1. In addition to elongated mitochondrial morphology and lack of fission, patient cells display lower coupling efficiency, increased proton leak and upregulation of glycolysis. Mitochondrial hyperfusion also results in aberrant cristae structure and hyperpolarized mitochondrial membrane potential. Peroxisomes show a severely elongated morphology in patient cells, which is associated with reduced respiration when cells are reliant on fatty acid oxidation. Metabolomic analyses revealed impaired methionine cycle and synthesis of pyrimidine nucleotides. Our study provides insight into the role of mitochondrial dynamics in cristae maintenance and the metabolic capacity of the cell, as well as the disease mechanism underlying EMPF1.


Subject(s)
Brain Diseases , Dynamins , Humans , Membrane Potential, Mitochondrial/genetics , Dynamins/genetics , Dynamins/metabolism , Brain Diseases/genetics , Brain Diseases/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mutation/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
5.
J Biol Chem ; 298(4): 101829, 2022 04.
Article in English | MEDLINE | ID: mdl-35293314

ABSTRACT

The mitochondrial F1Fo ATP synthase of the parasite Trypanosoma brucei has been previously studied in detail. This unusual enzyme switches direction in functionality during the life cycle of the parasite, acting as an ATP synthase in the insect stages, and as an ATPase to generate mitochondrial membrane potential in the mammalian bloodstream stages. Whereas the trypanosome F1 moiety is relatively highly conserved in structure and composition, the Fo subcomplex and the peripheral stalk have been shown to be more variable. Interestingly, a core subunit of the latter, the normally conserved subunit b, has been resistant to identification by sequence alignment or biochemical methods. Here, we identified a 17 kDa mitochondrial protein of the inner membrane, Tb927.8.3070, that is essential for normal growth, efficient oxidative phosphorylation, and membrane potential maintenance. Pull-down experiments and native PAGE analysis indicated that the protein is both associated with the F1Fo ATP synthase and integral to its assembly. In addition, its knockdown reduced the levels of Fo subunits, but not those of F1, and disturbed the cell cycle. Finally, analysis of structural homology using the HHpred algorithm showed that this protein has structural similarities to Fo subunit b of other species, indicating that this subunit may be a highly diverged form of the elusive subunit b.


Subject(s)
Mitochondrial Proton-Translocating ATPases , Protozoan Proteins , Trypanosoma brucei brucei , Animals , Mammals/metabolism , Membrane Potential, Mitochondrial/genetics , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/chemistry , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics
6.
Nat Commun ; 13(1): 651, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115503

ABSTRACT

Sustained mitochondrial fitness relies on coordinated biogenesis and clearance. Both processes are regulated by constant targeting of proteins into the organelle. Thus, mitochondrial protein import sets the pace for mitochondrial abundance and function. However, our understanding of mitochondrial protein translocation as a regulator of longevity remains enigmatic. Here, we targeted the main protein import translocases and assessed their contribution to mitochondrial abundance and organismal physiology. We find that reduction in cellular mitochondrial load through mitochondrial protein import system suppression, referred to as MitoMISS, elicits a distinct longevity paradigm. We show that MitoMISS triggers the mitochondrial unfolded protein response, orchestrating an adaptive reprogramming of metabolism. Glycolysis and de novo serine biosynthesis are causatively linked to longevity, whilst mitochondrial chaperone induction is dispensable for lifespan extension. Our findings extent the pro-longevity role of UPRmt and provide insight, relevant to the metabolic alterations that promote or undermine survival and longevity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Serine/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Energy Metabolism/genetics , Longevity/genetics , Membrane Potential, Mitochondrial/genetics , Metabolomics/methods , Microscopy, Fluorescence , Mitochondria/genetics , Mitochondrial Precursor Protein Import Complex Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Mitochondrial Proteins/genetics , Protein Transport/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Serine/genetics , Survival Analysis
7.
MAbs ; 14(1): 2020081, 2022.
Article in English | MEDLINE | ID: mdl-35030984

ABSTRACT

With the aim of increasing protein productivity of Chinese hamster ovary (CHO) cells, we sought to generate new CHO hosts with favorable biomanufacturing phenotypes and improved functionality. Here, we present an innovative approach of enriching the CHO host cells with a high mitochondrial membrane potential (MMP). Stable transfectant pools and clonal cell lines expressing difficult-to-express bispecific molecules generated from the MMP-enriched host outperformed the parental host by displaying (1) improved fed-batch productivity; (2) enhanced long-term cell viability of pools; (3) more favorable lactate metabolism; and (4) improved cell cloning efficiency during monoclonal cell line generation. Proteomic analysis together with Western blot validation were used to investigate the underlying mechanisms by which high MMP influenced production performance. The MMP-enriched host exhibited multifaceted protection against mitochondrial dysfunction and endoplasmic reticulum stress. Our findings indicate that the MMP-enriched host achieved an overall "fitter" phenotype that contributes to the significant improvement in biomanufacturing capability.


Subject(s)
Membrane Potential, Mitochondrial/genetics , Metabolic Engineering , Mitochondria/genetics , Mitochondria/metabolism , Animals , CHO Cells , Cricetulus
8.
Reprod Biol Endocrinol ; 20(1): 5, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980136

ABSTRACT

BACKGROUND: Potassium channels are important for the structure and function of the spermatozoa. As a potassium transporter, the mSlo3 is essential for male fertility as Slo3 knockout male mice were infertile with the series of functional defects in sperm cells. However, no pathogenic variant has been detected in human SLO3 to date. Here we reported a human case with homozygous SLO3 mutation. The function of SLO3 in human sperm and the corresponding assisted reproductive strategy are also investigated. METHODS: We performed whole-exome sequencing analysis from a large cohort of 105 patients with asthenoteratozoospermia. The effects of the variant were investigated by quantitative RT-PCR, western blotting, and immunofluorescence assays using the patient spermatozoa. Sperm morphological and ultrastructural studies were conducted using haematoxylin and eosin staining, scanning and transmission electron microscopy. RESULTS: We identified a homozygous missense variant (c.1237A > T: p.Ile413Phe) in the sperm-specific SLO3 in one Chinese patient with male infertility. This SLO3 variant was rare in human control populations and predicted to be deleterious by multiple bioinformatic tools. Sperm from the individual harbouring the homozygous SLO3 variant exhibited severe morphological abnormalities, such as acrosome hypoplasia, disruption of the mitochondrial sheath, coiled tails, and motility defects. The levels of SLO3 mRNA and protein in spermatozoa from the affected individual were reduced. Furthermore, the acrosome reaction, mitochondrial membrane potential, and membrane potential during capacitation were also afflicted. The levels of acrosome marker glycoproteins and PLCζ1 as well as the mitochondrial sheath protein HSP60 and SLO3 auxiliary subunit LRRC52, were significantly reduced in the spermatozoa from the affected individual. The affected man was sterile due to acrosome and mitochondrial dysfunction; however, intra-cytoplasmic sperm injection successfully rescued this infertile condition. CONCLUSIONS: SLO3 deficiency seriously impact acrosome formation, mitochondrial sheath assembly, and the function of K+ channels. Our findings provided clinical implications for the genetic and reproductive counselling of affected families.


Subject(s)
Acrosome/pathology , Asthenozoospermia/genetics , Infertility, Male/genetics , Acrosome Reaction/genetics , Adult , Asthenozoospermia/pathology , China , Cohort Studies , Consanguinity , Family Characteristics , Female , Homozygote , Humans , Infertility, Male/pathology , Infertility, Male/therapy , Large-Conductance Calcium-Activated Potassium Channels , Male , Membrane Potential, Mitochondrial/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membranes/pathology , Mutation, Missense , Pedigree , Pregnancy , Sperm Injections, Intracytoplasmic , Spermatozoa/abnormalities , Spermatozoa/pathology
9.
Mitochondrion ; 62: 13-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34656796

ABSTRACT

Leigh syndrome (LS) is one of the most common mitochondrial diseases in children, for which at least 90 causative genes have been identified. However, many LS patients have no genetic diagnosis, indicating that more disease-related genes remain to be identified. In this study, we identified a novel variant, m.3955G > A, in mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (MT-ND1) in two unrelated LS patients, manifesting as infancy-onset frequent seizures, neurodegeneration, elevated lactate levels, and bilateral symmetrical lesions in the brainstem, basal ganglia, and thalamus. Transfer of the mutant mtDNA with m.3955G > A into cybrids disturbed the MT-ND1 expression and CI assembly, followed by remarkable mitochondrial dysfunction, reactive oxygen species production, and mitochondrial membrane potential reduction. Our findings demonstrated the pathogenicity of the novel m.3955G > A variant, and extend the spectrum of pathogenic mtDNA variants.


Subject(s)
Genetic Predisposition to Disease , Leigh Disease/genetics , Membrane Potential, Mitochondrial/physiology , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Electron Transport/genetics , Female , Humans , Infant , Male , Membrane Potential, Mitochondrial/genetics , Models, Molecular , Mutation , Oxygen Consumption/genetics , Pedigree , Protein Conformation , Reactive Oxygen Species
10.
FEBS J ; 289(1): 262-278, 2022 01.
Article in English | MEDLINE | ID: mdl-34310050

ABSTRACT

Mitochondria form a branched tubular network in many types of cells, depending on a balance between mitochondrial fusion and fission. How mitochondrial fusion and fission are involved in regulating mitochondrial function and cell proliferation is not well understood. Here, we dissected the roles of mitochondrial fusion and fission in mitochondrial function and cell proliferation in fission yeast. We examined mitochondrial membrane potential by staining cells with DiOC6 and assessed mitochondrial respiration by directly measuring oxygen consumption of cells with a dissolved oxygen respirometer. We found that defects in mitochondrial fission or fusion reduce mitochondrial membrane potential and compromise mitochondrial respiration while the absence of both mitochondrial fusion and fission restores wild type-like respiration, normal membrane potential, and tubular networks of mitochondria. Moreover, we found that the absence of either mitochondrial fission or fusion prolongs the cell cycle and that the absence of both mitochondrial fusion and fission significantly delays cell cycle progression after nitrogen replenishment. The prolonged/delayed cell cycle is likely due to the deregulation of Cdc2 activation. Hence, our work not only establishes an intimate link between mitochondrial morphology and function but also underscores the importance of mitochondrial dynamics in regulating the cell cycle.


Subject(s)
DNA Polymerase III/genetics , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Dynamics/genetics , Saccharomyces cerevisiae Proteins/genetics , Carbocyanines/pharmacology , Cell Cycle/genetics , Cell Division/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Fungal/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects , Saccharomyces cerevisiae/genetics
11.
PLoS One ; 16(10): e0247211, 2021.
Article in English | MEDLINE | ID: mdl-34695170

ABSTRACT

Gaucher disease (GD) is caused by deficiency of the lysosomal membrane enzyme glucocerebrosidase (GCase) and the subsequent accumulation of its substrate, glucosylceramide (GC). Mostly missense mutations of the glucocerebrosidase gene (GBA) cause GCase misfolding and inhibition of proper lysosomal trafficking. The accumulated GC leads to lysosomal dysfunction and impairs the autophagy pathway. GD types 2 and 3 (GD2-3), or the neuronopathic forms, affect not only the Central Nervous System (CNS) but also have severe systemic involvement and progressive bone disease. Enzyme replacement therapy (ERT) successfully treats the hematologic manifestations; however, due to the lack of equal distribution of the recombinant enzyme in different organs, it has no direct impact on the nervous system and has minimal effect on bone involvement. Small molecules have the potential for better tissue distribution. Ambroxol (AMB) is a pharmacologic chaperone that partially recovers the mutated GCase activity and crosses the blood-brain barrier. Eliglustat (EGT) works by inhibiting UDP-glucosylceramide synthase, an enzyme that catalyzes GC biosynthesis, reducing GC influx load into the lysosome. Substrate reduction therapy (SRT) using EGT is associated with improvement in GD bone marrow burden score and bone mineral density parallel with the improvement in hematological parameters. We assessed the effects of EGT and AMB on GCase activity and autophagy-lysosomal pathway (ALP) in primary cell lines derived from patients with GD2-3 and compared to cell lines from healthy controls. We found that EGT, same as AMB, enhanced GCase activity in control cells and that an individualized response, that varied with GBA mutations, was observed in cells from patients with GD2-3. EGT and AMB enhanced the formation of lysosomal/late endosomal compartments and improved autophagy, independent of GBA mutations. Both AMB and EGT increased mitochondrial mass and density in GD2-3 fibroblasts, suggesting enhancement of mitochondrial function by activating the mitochondrial membrane potential. These results demonstrate that EGT and AMB, with different molecular mechanisms of action, enhance GCase activity and improve autophagy-lysosome dynamics and mitochondrial functions.


Subject(s)
Gaucher Disease/genetics , Molecular Chaperones/genetics , Adolescent , Adult , Autophagy/genetics , Child , Child, Preschool , Endosomes/genetics , Female , Fibroblasts/pathology , Glucosylceramidase/genetics , Glucosylceramides/genetics , Humans , Infant , Lysosomes/genetics , Male , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , Mutation/genetics , Young Adult
12.
Cancer Sci ; 112(12): 5114-5126, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34628698

ABSTRACT

Ovarian carcinoma immunoreactive antigen domain 2 (OCIAD2) has been reported to show significantly higher expression in invasive lung adenocarcinoma than in lung adenocarcinoma in situ, and its abnormal expression is associated with poorer prognosis of the patients. However, the cellular function of OCIAD2 in this tumor remains poorly understood. In the present study, we first validated that OCIAD2 showed higher expression in human lung adenocarcinoma tissues or cell lines than in normal lung tissue or immortalized normal bronchial epithelial cells. OCIAD2 was localized predominantly at the mitochondrial membrane in lung adenocarcinoma cells. Interestingly, suppression of OCIAD2 led to loss of mitochondrial structure and a reduction in the number of mitochondria. Moreover, OCIAD2 suppression led to downregulation of cellular growth, proliferation, migration, and invasion, and upregulation of mitochondria-related apoptosis. We also showed that OCIAD2 suppression induced a decrease in mitochondrial membrane potential and release of cytochrome c. Transcriptional profiling using RNA sequencing revealed a total of 137 genes whose expression was commonly altered after OCIAD2 knockdown in three lung adenocarcinoma cell lines (A549, HCC827, and PC9). Pathway enrichment analysis of those genes demonstrated significant enrichment in apoptotic signaling or endoplasmic reticulum (ER) stress pathways. Our data suggest that OCIAD2 inhibits the mitochondria-initiated apoptosis and thus promotes the survival of lung cancer cells. Therefore, OCIAD2 may be an effective target for treatment of lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Mitochondria/genetics , Neoplasm Proteins/genetics , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Blotting, Western , Cell Line, Tumor , Gene Ontology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/genetics , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Neoplasm Proteins/metabolism , RNA-Seq/methods , Reverse Transcriptase Polymerase Chain Reaction
13.
Exp Cell Res ; 408(2): 112861, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34624325

ABSTRACT

Ischemia reperfusion-induced acute kidney injury (IR-induced AKI) is a life-threatening disease with many complications. Mitofusin 2 (Mfn2) ubiquitination is related to AKI. But the underlying molecular mechanisms remain unknown. This study aimed to probe the mechanism of Mfn2 ubiquitination in IR-induced AKI development. In IR-induced AKI mouse models, orbital blood and urine were collected for assessing kidney function. The kidney injury, ultrastructure of mitochondria, and histopathology in mice were evaluated after injection of G5, an ubiquitination inhibitor. Oxygen glucose deprivation/reoxygenation (OGD/R) models were established in HK-2 cells, and the mitochondria were extracted. Cell viability, apoptosis, oxidative stress, inflammatory reaction, mitochondrial membrane potential, and ATP production were measured. Mfn2 ubiquitination in mouse and cell models was evaluated. si-SIRT3 and pcDNA3.1-SIRT3 were transfected into cell models. Consequently, kidney function in mice was impaired by IR-induced AKI. Mfn2 ubiquitination and degradation promoted IR-induced AKI. OGD/R induced renal tubular epithelial cell injury and disrupted mitochondrial dynamics and functions through promoting Mfn2 ubiquitination. SIRT3 knockdown led to Mfn2 ubiquitination by binding to UBC; while its overexpression alleviated tubular epithelial cell injury. Briefly, SIRT3 mediates Mfn2 ubiquitination to relieve IR-induced AKI. This investigation may offer new insights for the treatment of IR-induced AKI injury.


Subject(s)
Acute Kidney Injury/genetics , GTP Phosphohydrolases/genetics , Reperfusion Injury/genetics , Sirtuin 3/genetics , Acute Kidney Injury/pathology , Adenosine Triphosphate/genetics , Animals , Apoptosis/genetics , Cell Survival/genetics , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Membrane Potential, Mitochondrial/genetics , Mice , Oxidative Stress/genetics , Proteolysis , Reperfusion Injury/pathology , Ubiquitination/genetics
14.
Neuroreport ; 32(17): 1395-1402, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34718247

ABSTRACT

OBJECTIVE: Hypoxic/ischemic brain damage (HIBD) results in increased neonatal mortality and serious neurologic morbidity. Long noncoding RNAs (lncRNAs) are shown as essential modulators of various neurological diseases. Here, we determined the mechanisms of lncRNA GAS5 in mitochondrial apoptosis in HIBD rats. METHODS: The HIBD neonatal rat model was established and treated with shRNA-GAS5 or antagomir miR-128-3p. The morphological changes and apoptosis rate were observed by histological staining. Expressions of GAS5, miR-128-3p, and Bax mRNA in brain tissues of HIBD neonatal rats were determined. The binding relationships between GAS5 and miR-128-3p, and miR-128-3p and Bax were confirmed by dual-luciferase assay. Subsequently, the mitochondrial membrane potential and apoptosis-related factors in brain tissues of HIBD neonatal rats were detected. Western blot analysis was performed to detect the expression of Akt/GSK3ß pathway-associated proteins. RESULTS: The neurons in the brain tissue of HIBD neonatal rats decreased with disordered arrangement, and showed vacuolization and nuclear pyknosis, obvious brain damage, increased neuronal apoptosis, and enhanced mitochondrial apoptotic pathway. Downregulated miR-128-3p and upregulated GAS5 and Bax mRNA were found in HIBD neonatal rats. There were binding relationships between GAS5 and miR-128-3p, and miR-128-3p and Bax mRNA. Inhibition of lncRNA GAS5 in HIBD neonatal rats suppressed mitochondrial apoptosis. miR-128-3p knockdown annulled the inhibitory effect of inhibiting lncRNA GAS5 on mitochondrial apoptosis. Silencing GAS5 increased the phosphorylation levels of Akt and GSK3ß. CONCLUSION: Downregulation of lncRNA GAS5 prevents mitochondrial apoptosis in neonatal HIBD rats by regulating the miR-128-3p/Bax/Akt/GSK-3ß axis.


Subject(s)
Brain/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Hypoxia-Ischemia, Brain/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Nucleolar/genetics , bcl-2-Associated X Protein/genetics , Animals , Animals, Newborn , Apoptosis/genetics , Blotting, Western , Down-Regulation , Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia-Ischemia, Brain/metabolism , Membrane Potential, Mitochondrial/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , Rats , bcl-2-Associated X Protein/metabolism
15.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502546

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by an accumulation of amyloid ß (Aß) peptides in the brain and mitochondrial dysfunction. Platelet activation is enhanced in AD and platelets contribute to AD pathology by their ability to facilitate soluble Aß to form Aß aggregates. Thus, anti-platelet therapy reduces the formation of cerebral amyloid angiopathy in AD transgenic mice. Platelet mitochondrial dysfunction plays a regulatory role in thrombotic response, but its significance in AD is unknown and explored herein. METHODS: The effects of Aß-mediated mitochondrial dysfunction in platelets were investigated in vitro. RESULTS: Aß40 stimulation of human platelets led to elevated reactive oxygen species (ROS) and superoxide production, while reduced mitochondrial membrane potential and oxygen consumption rate. Enhanced mitochondrial dysfunction triggered platelet-mediated Aß40 aggregate formation through GPVI-mediated ROS production, leading to enhanced integrin αIIbß3 activation during synergistic stimulation from ADP and Aß40. Aß40 aggregate formation of human and murine (APP23) platelets were comparable to controls and could be reduced by the antioxidant vitamin C. CONCLUSIONS: Mitochondrial dysfunction contributes to platelet-mediated Aß aggregate formation and might be a promising target to limit platelet activation exaggerated pathological manifestations in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Blood Platelets/metabolism , Mitochondria/metabolism , Protein Aggregation, Pathological/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , Cells, Cultured , Humans , Integrins/metabolism , Membrane Potential, Mitochondrial/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oxygen Consumption/drug effects , Platelet Activation/drug effects , Platelet Function Tests/methods , Reactive Oxygen Species/metabolism
16.
FASEB J ; 35(9): e21814, 2021 09.
Article in English | MEDLINE | ID: mdl-34369624

ABSTRACT

Alteration in glucose homeostasis during cancer metabolism is an important phenomenon. Though several important transcription factors have been well studied in the context of the regulation of metabolic gene expression, the role of epigenetic readers in this regard remains still elusive. Epigenetic reader protein transcription factor 19 (TCF19) has been recently identified as a novel glucose and insulin-responsive factor that modulates histone posttranslational modifications to regulate glucose homeostasis in hepatocytes. Here we report that TCF19 interacts with a non-histone, well-known tumor suppressor protein 53 (p53) and co-regulates a wide array of metabolic genes. Among these, the p53-responsive carbohydrate metabolic genes Tp53-induced glycolysis and apoptosis regulator (TIGAR) and Cytochrome C Oxidase assembly protein 2 (SCO2), which are the key regulators of glycolysis and oxidative phosphorylation respectively, are under direct regulation of TCF19. Remarkably, TCF19 can form different transcription activation/repression complexes which show substantial overlap with that of p53, depending on glucose-mediated variant stress situations as obtained from IP/MS studies. Interestingly, we observed that TCF19/p53 complexes either have CBP or HDAC1 to epigenetically program the expression of TIGAR and SCO2 genes depending on short-term high glucose or prolonged high glucose conditions. TCF19 or p53 knockdown significantly altered the cellular lactate production and led to increased extracellular acidification rate. Similarly, OCR and cellular ATP production were reduced and mitochondrial membrane potential was compromised upon depletion of TCF19 or p53. Subsequently, through RNA-Seq analysis from patients with hepatocellular carcinoma, we observed that TCF19/p53-mediated metabolic regulation is fundamental for sustenance of cancer cells. Together the study proposes that TCF19/p53 complexes can regulate metabolic gene expression programs responsible for mitochondrial energy homeostasis and stress adaptation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Mitochondria/genetics , Molecular Chaperones/genetics , Phosphoric Monoester Hydrolases/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Adaptation, Biological/genetics , Apoptosis/genetics , Cell Line, Tumor , Energy Metabolism/genetics , Glucose/genetics , Hep G2 Cells , Homeostasis/genetics , Humans , Membrane Potential, Mitochondrial/genetics , Stress, Physiological/genetics , Transcriptional Activation/genetics
17.
Nat Commun ; 12(1): 4900, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385433

ABSTRACT

Skeletal muscle subsarcolemmal mitochondria (SSM) and intermyofibrillar mitochondria subpopulations have distinct metabolic activity and sensitivity, though the mechanisms that localize SSM to peripheral areas of muscle fibers are poorly understood. A protein interaction study and complexome profiling identifies PERM1 interacts with the MICOS-MIB complex. Ablation of Perm1 in mice reduces muscle force, decreases mitochondrial membrane potential and complex I activity, and reduces the numbers of SSM in skeletal muscle. We demonstrate PERM1 interacts with the intracellular adaptor protein ankyrin B (ANKB) that connects the cytoskeleton to the plasma membrane. Moreover, we identify a C-terminal transmembrane helix that anchors PERM1 into the outer mitochondrial membrane. We conclude PERM1 functions in the MICOS-MIB complex and acts as an adapter to connect the mitochondria with the sarcolemma via ANKB.


Subject(s)
Ankyrins/metabolism , Mitochondria, Muscle/metabolism , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Sarcolemma/metabolism , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mice, Knockout , Mitochondrial Proteins/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology
18.
J Pharmacol Sci ; 147(2): 200-207, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34384568

ABSTRACT

Leber hereditary optic neuropathy (LHON) is caused by mitochondrial DNA mutations and is the most common inherited mitochondrial disease. It is responsible for central vision loss in young adulthood. However, the precise mechanisms of onset are unknown. This study aimed to elucidate the mechanisms underlying LHON pathology and to discover new therapeutic agents. First, we assessed whether rotenone, a mitochondrial complex Ⅰ inhibitor, induced retinal degeneration such as that in LHON in a mouse model. Rotenone decreased the thickness of the inner retina and increased the expression levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG) and immunoglobulin heavy-chain binding protein (BiP). Second, we assessed whether rotenone reproduces LHON pathologies on RGC-5, a neural progenitor cell derived from the retina. Rotenone increased the cell death rate, ROS production and the expression levels of ER stress markers. During chemical compounds screening, we used anti-oxidative compounds, ER stress inhibitors and anti-inflammatory compounds in a rotenone-induced in vitro model. We found that SUN N8075, an ER stress inhibitor, reduced mitochondrial ROS production and improved the mitochondrial membrane potential. Consequently, the ER stress response is strongly related to the pathologies of LHON, and ER stress inhibitors may have a protective effect against LHON.


Subject(s)
Aniline Compounds/pharmacology , Drug Discovery , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Optic Atrophy, Hereditary, Leber/drug therapy , Optic Atrophy, Hereditary, Leber/genetics , Piperazines/pharmacology , Rotenone/adverse effects , Animals , Cells, Cultured , DNA, Mitochondrial/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/genetics , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice, Inbred C57BL , Molecular Targeted Therapy , Mutation , Optic Atrophy, Hereditary, Leber/chemically induced , Optic Atrophy, Hereditary, Leber/pathology , Reactive Oxygen Species/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Degeneration/chemically induced , Retinal Degeneration/genetics , Retinal Degeneration/pathology
19.
FEBS J ; 288(23): 6828-6843, 2021 12.
Article in English | MEDLINE | ID: mdl-34258867

ABSTRACT

Adverse fetal environment, in particular a shortage or excess of nutrients, is associated with increased risks of metabolic diseases later in life. However, the molecular mechanisms underlying this developmental origin of adult diseases remain unclear. Here, we directly tested the role of mitochondrial stress in mediating fetal programming in mice by enzymatically depleting mtDNA in zygotes. mtDNA-targeted plasmid microinjection is used to reduce embryonic mtDNA copy number directly, followed by embryo transfer. Mice with reduced zygote mtDNA copy number were born morphologically normal and showed no accelerated body weight gain. However, at 5 months of age these mice showed markedly increased hepatic lipidosis and became glucose-intolerant. Hepatic mRNA and protein expressions of peroxisome proliferator-activated receptor α (Pparα), a key transcriptional regulator of lipid metabolism, were significantly decreased as a result of increased DNA methylation in its proximal regulatory region. These results indicate that perturbation of mitochondrial function around the periconceptional period causes hypermethylation and thus suppressed expression of PPARα in fetal liver, leading to impaired hepatic lipid metabolism. Our findings provide the first direct evidence that mitochondrial stress mediates epigenetic changes associated with fetal programming of adult diseases in a mammalian system.


Subject(s)
DNA Copy Number Variations , DNA, Mitochondrial/genetics , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Lipid Metabolism/genetics , Lipolysis/genetics , Liver/metabolism , Age Factors , Animals , DNA Methylation , Embryo, Mammalian/embryology , Epigenomics/methods , Female , Gene Expression Regulation, Developmental , Liver/embryology , Male , Membrane Potential, Mitochondrial/genetics , Mice, Inbred ICR , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/physiology , Oxygen Consumption/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , RNA-Seq/methods
20.
Sci Rep ; 11(1): 15510, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330933

ABSTRACT

Ischemia is a major cause of kidney damage. Proximal tubular epithelial cells (PTECs) are highly susceptible to ischemic insults that frequently cause acute kidney injury (AKI), a potentially life-threatening condition with high mortality. Accumulating evidence has identified altered mitochondrial function as a central pathologic feature of AKI. The mitochondrial NAD+-dependent enzyme sirtuin 5 (SIRT5) is a key regulator of mitochondrial form and function, but its role in ischemic renal injury (IRI) is unknown. SIRT5 expression was increased in murine PTECs after IRI in vivo and in human PTECs (hPTECs) exposed to an oxygen/nutrient deprivation (OND) model of IRI in vitro. SIRT5-depletion impaired ATP production, reduced mitochondrial membrane potential, and provoked mitochondrial fragmentation in hPTECs. Moreover, SIRT5 RNAi exacerbated OND-induced mitochondrial bioenergetic dysfunction and swelling, and increased degradation by mitophagy. These findings suggest SIRT5 is required for normal mitochondrial function in hPTECs and indicate a potentially important role for the enzyme in the regulation of mitochondrial biology in ischemia.


Subject(s)
Acute Kidney Injury/metabolism , Mitochondria/metabolism , Sirtuins/metabolism , Acute Kidney Injury/genetics , Animals , Blotting, Western , Cell Line , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria/genetics , Mitophagy/genetics , Mitophagy/physiology , Sirtuins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...