Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 795
Filter
1.
Bioengineered ; 13(5): 12489-12503, 2022 05.
Article in English | MEDLINE | ID: mdl-35579419

ABSTRACT

Genome-derived microRNAs (miRNAs or miRs) control post-transcriptional gene expression critical for various cellular processes. Recently, we have invented a novel platform technology to achieve high-yield production of fully humanized, bioengineered miRNA agents (hBERAs) for research and development. This study is aimed to produce and utilize a new biologic miR-34a-5p (or miR-34a) molecule, namely, hBERA/miR-34a, to delineate the role of miR-34a-5p in the regulation of mitochondrial functions in human carcinoma cells. Bioengineered hBERA/miR-34a was produced through in vivo fermentation production and purified by anion exchange fast protein liquid chromatography. hEBRA/miR-34a was processed to target miR-34a-5p in human osteosarcoma and lung cancer cells, as determined by selective stem-loop reverse transcription quantitative polymerase chain reaction analysis. The mitochondrial inner membrane protein MPV17 like 2 (MPV17L2) was validated as a direct target for miR-34a-5p by dual luciferase reporter assay. Western blot analysis revealed that bioengineered miR-34a-5p effectively reduced MPV17L2 protein outcomes, leading to much lower levels of respiratory chain Complex I activities and intracellular ATP that were determined with specific assay kits. Moreover, Seahorse Mito Stress Test assay was conducted, and the results showed that biologic miR-34a-5p sharply reduced cancer cell mitochondrial respiration capacity, accompanied by a remarkable increase of oxidative stress and elevated apoptotic cell death, which are manifested by greater levels of reactive oxygen species and selective apoptosis biomarkers, respectively. These results demonstrate the presence and involvement of the miR-34a-5p-MPV17L2 pathway in the control of mitochondrial functions in human carcinoma cells and support the utility of novel bioengineered miRNA molecules for functional studies.


Subject(s)
Biological Products , Bone Neoplasms , Carcinoma , Lung Neoplasms , Membrane Proteins , MicroRNAs , Mitochondria , Mitochondrial Proteins , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Proliferation/genetics , Humans , Lung Neoplasms/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics
2.
JCI Insight ; 7(4)2022 02 22.
Article in English | MEDLINE | ID: mdl-35015731

ABSTRACT

Mitophagy and mitochondrial integrated stress response (ISR) are 2 primary protective mechanisms to maintain functional mitochondria. Whether these 2 processes are coordinately regulated remains unclear. Here we show that mitochondrial fission 1 protein (Fis1), which is required for completion of mitophagy, serves as a signaling hub linking mitophagy and ISR. In mouse hepatocytes, high fat diet (HFD) feeding induces unresolved oxidative stress, defective mitophagy and enhanced type I interferon (IFN-I) response implicated in promoting metabolic inflammation. Adenoviral-mediated acute hepatic Fis1 overexpression is sufficient to reduce oxidative damage and improve glucose homeostasis in HFD-fed mice. RNA-Seq analysis reveals that Fis1 triggers a retrograde mitochondria-to-nucleus communication upregulating ISR genes encoding anti-oxidant defense, redox homeostasis, and proteostasis pathways. Fis1-mediated ISR also suppresses expression of IFN-I-stimulated genes through activating transcription factor 5 (Atf5), which inhibits the transactivation activity of interferon regulatory factor 3 (Irf3) known to control IFN-I production. Metabolite analysis demonstrates that Fis1 activation leads to accumulation of fumarate, a TCA cycle intermediate capable of increasing Atf5 activity. Consequently, hepatic Atf5 overexpression or monomethyl fumarate (MMF) treatment improves glucose homeostasis in HFD-fed mice. Collectively, these results support the potential use of small molecules targeting the Fis1-Atf5 axis, such as MMF, to treat metabolic diseases.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitophagy/genetics , Oxidative Stress , RNA/genetics , Animals , Homeostasis , Liver/cytology , Mice , Mitochondrial Proteins/biosynthesis , Models, Animal , Signal Transduction
3.
J Biol Chem ; 298(2): 101508, 2022 02.
Article in English | MEDLINE | ID: mdl-34942149

ABSTRACT

The mitochondrial solute carrier family 8 sodium/calcium/lithium exchanger, member B1 (NCLX) is an important mediator of calcium extrusion from mitochondria. In this study, we tested the hypothesis that physiological expression levels of NCLX are essential for maintaining neuronal resilience in the face of excitotoxic challenge. Using an shRNA-mediated approach, we showed that reduced NCLX expression exacerbates neuronal mitochondrial calcium dysregulation, mitochondrial membrane potential (ΔΨm) breakdown, and reactive oxygen species generation during excitotoxic stimulation of primary hippocampal cultures. Moreover, NCLX knockdown-which affected both neurons and glia-resulted not only in enhanced neurodegeneration following an excitotoxic insult but also in neuronal and astrocytic cell death under basal conditions. Our data also revealed that synaptic activity, which promotes neuroprotective signaling, can become lethal upon NCLX depletion; expression of NCLX-targeted shRNA impaired the clearance of mitochondrial calcium following action potential bursts, and was associated both with ΔΨm breakdown and substantial neurodegeneration in hippocampal cultures undergoing synaptic activity. Finally, we showed that NCLX knockdown within the hippocampal cornu ammonis 1 region in vivo causes substantial neurodegeneration and astrodegeneration. In summary, we demonstrated that dysregulated NCLX expression not only sensitizes neuroglial networks to excitotoxic stimuli but also notably renders otherwise neuroprotective synaptic activity toxic. These findings may explain the emergence of neurodegeneration and astrodegeneration in patients with disorders characterized by disrupted NCLX expression or function, and suggest that treatments aimed at enhancing or restoring NCLX function may prevent central nervous system damage in these disease states.


Subject(s)
Calcium , Mitochondrial Proteins , Nerve Net , Neuroglia , Sodium-Calcium Exchanger , Calcium/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nerve Net/metabolism , Neuroglia/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sodium-Calcium Exchanger/biosynthesis , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
4.
PLoS One ; 16(11): e0259837, 2021.
Article in English | MEDLINE | ID: mdl-34793488

ABSTRACT

Human protoporphyrinogen oxidase IX (hPPO) is an oxygen-dependent enzyme catalyzing the penultimate step in the heme biosynthesis pathway. Mutations in the enzyme are linked to variegate porphyria, an autosomal dominant metabolic disease. Here we investigated eukaryotic cells as alternative systems for heterologous expression of hPPO, as the use of a traditional bacterial-based system failed to produce several clinically relevant hPPO variants. Using bacterially-produced hPPO, we first analyzed the impact of N-terminal tags and various detergent on hPPO yield, and specific activity. Next, the established protocol was used to compare hPPO constructs heterologously expressed in mammalian HEK293T17 and insect Hi5 cells with prokaryotic overexpression. By attaching various fusion partners at the N- and C-termini of hPPO we also evaluated the influence of the size and positioning of fusion partners on expression levels, specific activity, and intracellular targeting of hPPO fusions in mammalian cells. Overall, our results suggest that while enzymatically active hPPO can be heterologously produced in eukaryotic systems, the limited availability of the intracellular FAD co-factor likely negatively influences yields of a correctly folded protein making thus the E.coli a system of choice for recombinant hPPO overproduction. At the same time, PPO overexpression in eukaryotic cells might be preferrable in cases when the effects of post-translational modifications (absent in bacteria) on target protein functions are studied.


Subject(s)
Flavoproteins/biosynthesis , Flavoproteins/isolation & purification , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/isolation & purification , Protoporphyrinogen Oxidase/biosynthesis , Protoporphyrinogen Oxidase/isolation & purification , Animals , Cell Line , Escherichia coli/genetics , Flavoproteins/genetics , HEK293 Cells , Humans , Mitochondrial Proteins/genetics , Protoporphyrinogen Oxidase/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Sf9 Cells
5.
Mol Cell ; 81(20): 4191-4208.e8, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34686314

ABSTRACT

To survive, mammalian cells must adapt to environmental challenges. While the cellular response to mild stress has been widely studied, how cells respond to severe stress remains unclear. We show here that under severe hyperosmotic stress, cells enter a transient hibernation-like state in anticipation of recovery. We demonstrate this adaptive pausing response (APR) is a coordinated cellular response that limits ATP supply and consumption through mitochondrial fragmentation and widespread pausing of mRNA translation. This pausing is accomplished by ribosome stalling at translation initiation codons, which keeps mRNAs poised to resume translation upon recovery. We further show that recovery from severe stress involves ISR (integrated stress response) signaling that permits cell cycle progression, resumption of growth, and reversal of mitochondria fragmentation. Our findings indicate that cells can respond to severe stress via a hibernation-like mechanism that preserves vital elements of cellular function under harsh environmental conditions.


Subject(s)
Cell Proliferation , Fibroblasts/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Osmotic Pressure , Protein Biosynthesis , Ribosomes/metabolism , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Codon, Initiator , Fibroblasts/pathology , HEK293 Cells , Humans , Kinetics , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Ribosomes/genetics , Signal Transduction
6.
Invest Ophthalmol Vis Sci ; 62(12): 8, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34505865

ABSTRACT

Purpose: Oxidative stress is a major factor underlying many neurodegenerative diseases. However, antioxidant therapy has had mixed results, possibly because of its indiscriminate activity. The purpose of our study was to determine if the human OXR1 (hOXR1) antioxidant regulatory gene could protect neurons from oxidative stress and delay photoreceptor cell death. Methods: The cone-like 661W cell line was transfected to stably express the hOXR1 gene. Oxidative stress was induced by the addition of hydrogen peroxide (H2O2). Intracellular levels of reactive oxygen species (ROS), caspase cleavage, and cellular resistance to oxidative stress were determined and compared between the control and hOXR1 cells. For in vivo analysis, AAV8-hOXR1 was injected subretinally into the rd1 mouse model of retinal degeneration. Functional and structural integrity of the photoreceptors were assessed using electroretinography (ERG), histology, and immunofluorescence analysis. Results: Expression of hOXR1 increased cellular resistance and reduced ROS levels and caspase cleavage in the 661W cell line after H2O2-induced oxidative stress. Subretinal injection of AAV8-hOXR1 in the rd1 mice improved their photoreceptor light response, expression and localization of photoreceptor-specific proteins, and delayed retinal degeneration. Conclusions: Our results suggest that OXR1 is a potential therapy candidate for retinal degeneration. Because OXR1 targets oxidative stress, a common feature of many retinal degenerative diseases, it should be of therapeutic value to multiple retinal degenerative diseases.


Subject(s)
Gene Expression Regulation , Genetic Therapy/methods , Mitochondrial Proteins/genetics , Oxidative Stress , RNA/genetics , Retina/pathology , Retinal Degeneration/therapy , Animals , Cell Death , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mitochondrial Proteins/biosynthesis , Photoreceptor Cells, Vertebrate , Reactive Oxygen Species/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism
8.
Biomed Res Int ; 2021: 5512624, 2021.
Article in English | MEDLINE | ID: mdl-34124242

ABSTRACT

Prostate cancer is currently associated with higher morbidity and mortality in men in the United States and Western Europe, so it is important to identify genes that regulate prostate cancer. The high-dimension gene expression profile impedes the discovery of biclusters which are of great significance to the identification of the basic cellular processes controlled by multiple genes and the identification of large-scale unknown effects hidden in the data. We applied the biclustering method MCbiclust to explore large biclusters in the TCGA cohort through a large number of iterations. Two biclusters were found with the highest silhouette coefficient value. The expression patterns of one bicluster are highly similar to those found by the gene expression profile of the known androgen-regulated genes. Further gene set enrichment revealed that mitochondrial function-related genes were negatively correlated with AR regulation-related genes. Then, we performed differential analysis, AR binding site analysis, and survival analysis on the core genes with high phenotypic contribution. Among the core genes, NDUFA10 showed a low expression value in cancer patients across different expression profiles, while NDUFV2 showed a high expression value in cancer patients. Survival analysis of NDUFA10 and NDUFV2 demonstrated that both genes were unfavorable prognostic markers.


Subject(s)
Biomarkers, Tumor , Databases, Nucleic Acid , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mitochondrial Proteins , NADH Dehydrogenase , Neoplasm Proteins , Prostatic Neoplasms , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Disease-Free Survival , Gene Expression Profiling , Humans , Male , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , NADH Dehydrogenase/biosynthesis , NADH Dehydrogenase/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Survival Rate
9.
J Ovarian Res ; 14(1): 75, 2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34078439

ABSTRACT

BACKGROUND: The BOLA gene family, comprising three members, is mainly involved in regulating intracellular iron homeostasis. Emerging evidence suggests that BolA family member 2 plays a vital role in tumorigenesis and hepatic cellular carcinoma progression. However, there was less known about its role in ovarian cancer. METHODS: In the present study, we investigated the expression profiles, prognostic roles, and genetic alterations of three BolA family members in patients with ovarian cancer through several public databases, containing Oncomine and Gene Expression Profiling Interactive Analysis, Human Protein Atlas, Kaplan-Meier plotter and cBioPortal. Then, we constructed the protein-protein interaction networks of BOLA proteins and their interactors by using the String database and Cytoscape software. In addition, we performed the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment by the Annotation, Visualization, and Integrated Discovery database. Finally, we explored the mechanisms underlying BolA family members' involvement in OC by using gene set enrichment analysis. RESULTS: The mRNA and protein expression levels of BOLA2 and BOLA3 were heavily higher in ovarian cancer tissues than in normal ovarian tissues. Dysregulated mRNA expressions of three BolA family members were significantly associated with prognosis in overall or subgroup analysis. Moreover, genetic alterations also occurred in three BolA family members in ovarian cancer. GO analysis indicated that BolA family members might regulate the function of metal ion binding and protein disulfide oxidoreductase activity. Gene set enrichment analysis indicated that BolA family members were mainly associated with oxidative phosphorylation, proteasome, protein export, and glutathione metabolism in ovarian cancer. CONCLUSION: In brief, our finding may contribute to increasing currently limited prognostic biomarkers and treatment options for ovarian cancer.


Subject(s)
Mitochondrial Proteins/biosynthesis , Ovarian Neoplasms/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Computational Biology , Female , Gene Expression Profiling , Humans , Mitochondrial Proteins/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Neurochem Int ; 148: 105083, 2021 09.
Article in English | MEDLINE | ID: mdl-34052298

ABSTRACT

Chronic stress is a risk factor for the development of psychiatric illnesses through impairment of the ability to appropriately regulate physiological and behavioral responses, but the molecular events that lead to damage of hippocampal neurons remain unclear. The medicinal herb Spilanthes acmella Murr. has been used as a traditional medicine for various diseases and its extracts exhibit antioxidant activity. The present study explored the molecular signals of mitochondrial dynamics and investigated the beneficial effects of S. acmella Murr. An ethyl acetate extract of this plant was used to assess mitochondrial dynamics in response to chronic restraint stress (CRS) in male Sprague-Dawley rats. The results demonstrated that the S. acmella Murr. extract reduced the expression of mitochondrial fission protein but induced HSP60, MnSOD and ATPsynthase in the hippocampus of the CRS rats. In addition, S. acmella Murr. extract reversed depressive symptoms in the forced swim test. Our findings suggested that S. acmella Murr. extract provides a potential treatment of chronic stress, and that the mechanism is associated with the alleviation of neuronal injury and maintenance of mitochondrial function.


Subject(s)
Asteraceae/chemistry , Mitochondria/drug effects , Plant Extracts/therapeutic use , Stress, Psychological/drug therapy , Animals , Antioxidants , Behavior, Animal/drug effects , Chaperonin 60/biosynthesis , Chaperonin 60/genetics , Chronic Disease , Cognition/drug effects , Depression/drug therapy , Depression/psychology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Plants, Medicinal , Rats , Rats, Sprague-Dawley , Restraint, Physical
11.
Sci Rep ; 11(1): 7320, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795807

ABSTRACT

DJ-1/PARK7 mutations are linked with familial forms of early-onset Parkinson's disease (PD). We have studied the degradation of untagged DJ-1 wild type (WT) and missense mutants in mouse embryonic fibroblasts obtained from DJ-1-null mice, an approach closer to the situation in patients carrying homozygous mutations. The results showed that the mutants L10P, M26I, A107P, P158Δ, L166P, E163K, and L172Q are unstable proteins, while A39S, E64D, R98Q, A104T, D149A, A171S, K175E, and A179T are as stable as DJ-1 WT. Inhibition of proteasomal and autophagic-lysosomal pathways had little effect on their degradation. Immunofluorescence and biochemical fractionation studies indicated that M26I, A107P, P158Δ, L166P, E163K, and L172Q mutants associate with mitochondria. Silencing of mitochondrial matrix protease LonP1 produced a strong reduction of the degradation of the mitochondrial-associated DJ-1 mutants A107P, P158Δ, L166P, E163K, and L172Q but not of mutant L10P. These results demonstrated a mitochondrial pathway of degradation of those DJ-1 missense mutants implicated in PD pathogenesis.


Subject(s)
ATP-Dependent Proteases/biosynthesis , Mitochondria/enzymology , Mitochondrial Proteins/biosynthesis , Mutation, Missense , Parkinson Disease/enzymology , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Animals , Fibroblasts/metabolism , Gene Silencing , Homozygote , Humans , Mice , Microscopy, Fluorescence , Proteasome Endopeptidase Complex , Subcellular Fractions
12.
Front Endocrinol (Lausanne) ; 12: 653602, 2021.
Article in English | MEDLINE | ID: mdl-33796081

ABSTRACT

The factors influencing Leydig cell maturity and the acquisition of functional capacity are incompletely defined. Here we analyzed the constant light (LL) influence on Leydig cells' endocrine function during reproductive maturation. Rats were exposed to LL from P21 to P90. Data were collected at juvenile (P35), peri/pubertal (P42, P49), and adult (P90) stages of life. The results proved the effect of LL on rats' physiology by changing of bimodal voluntary activity pattern into free-running. Additionally, the peripheral clock in Leydig cells changed in LL condition, indicating disturbed rhythm: the positive element (Bmal1) increased in pre-/pubertal but decreased in the adult period, while negative elements (Per2 and Reverba) were increased. The effects of LL were most prominent in puberty: pituitary genes encoding gonadotropic hormones (Cga, Lhb, Fshb) decreased; serum corticosterone increased, while serum androgens and mass of testicular and sex accessory organs reduced; markers of Leydig cells maturity/differentiation (Insl3, Lhcgr) and steroidogenesis-related genes (Scarb1, Star, Cyp11a1, Cyp17a1) decreased; the steroidogenic and energetic capacity of the Leydig cell mitochondria decreased; the mtDNA copy number reduced, and mitochondrial dynamics markers changed: fusion decreased (Opa1 and Mfn2), and mitophagy increased (Pink1). In adults, the negative effect of LL on mitochondrial function and steroidogenic capacity persists in adult Leydig cells while other parameters reached control values. Altogether, the results indicate that LL slows down Leydig cells' maturation by reducing the endocrine and energy capacity of cells leading to the delay of reproductive development.


Subject(s)
Corticosterone/blood , Endocrine System/physiology , Leydig Cells/metabolism , Light , Adenosine Triphosphate/metabolism , Androgens/pharmacology , Animals , Body Weight , Cell Differentiation , DNA, Mitochondrial/metabolism , GTP Phosphohydrolases/biosynthesis , Luteinizing Hormone/blood , Male , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Organ Size , Pituitary Gland/drug effects , Protein Kinases/biosynthesis , Rats , Rats, Wistar , Sexual Maturation , Steroids/metabolism , Testosterone/blood
13.
Int J Mol Sci ; 22(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803109

ABSTRACT

Mitochondrial misreading, conferred by mutation V338Y in mitoribosomal protein Mrps5, in-vivo is associated with a subtle neurological phenotype. Brain mitochondria of homozygous knock-in mutant Mrps5V338Y/V338Y mice show decreased oxygen consumption and reduced ATP levels. Using a combination of unbiased RNA-Seq with untargeted metabolomics, we here demonstrate a concerted response, which alleviates the impaired functionality of OXPHOS complexes in Mrps5 mutant mice. This concerted response mitigates the age-associated decline in mitochondrial gene expression and compensates for impaired respiration by transcriptional upregulation of OXPHOS components together with anaplerotic replenishment of the TCA cycle (pyruvate, 2-ketoglutarate).


Subject(s)
Aging/metabolism , Brain/metabolism , Gene Expression Regulation , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mutation, Missense , Protein Biosynthesis , Ribosomal Proteins/biosynthesis , Adenosine Triphosphate/metabolism , Aging/genetics , Aging/pathology , Animals , Brain/pathology , Citric Acid Cycle/genetics , Gene Knock-In Techniques , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Ribosomal Proteins/genetics
14.
PLoS Genet ; 17(4): e1009521, 2021 04.
Article in English | MEDLINE | ID: mdl-33857138

ABSTRACT

The RNA-binding protein Mrn1 in Saccharomyces cerevisiae targets over 300 messenger RNAs, including many involved in cell wall biogenesis. The impact of Mrn1 on these target transcripts is not known, however, nor is the cellular role for this regulation. We have shown that Mrn1 represses target mRNAs through the action of its disordered, asparagine-rich amino-terminus. Its endogenous targets include the paralogous SUN domain proteins Nca3 and Uth1, which affect mitochondrial and cell wall structure and function. While loss of MRN1 has no effect on fermentative growth, we found that mrn1Δ yeast adapt more quickly to respiratory conditions. These cells also have enlarged mitochondria in fermentative conditions, mediated in part by dysregulation of NCA3, and this may explain their faster switch to respiration. Our analyses indicated that Mrn1 acts as a hub for integrating cell wall integrity and mitochondrial biosynthesis in a carbon-source responsive manner.


Subject(s)
Cell Wall/genetics , Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Cell Wall/metabolism , Homeostasis/genetics , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Proteins/biosynthesis , Organelle Biogenesis , RNA, Messenger/genetics , Saccharomyces cerevisiae/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
15.
Can J Cardiol ; 37(10): 1593-1606, 2021 10.
Article in English | MEDLINE | ID: mdl-33838228

ABSTRACT

BACKGROUND: Downregulation of claudin-5 in the heart is associated with the end-stage heart failure. However, the underlying mechanism ofclaudin-5 is unclear. Here we investigated the molecular actions of claudin-5 in perspective of mitochondria in cardiomyocytes to better understand the role of claudin-5 in cardioprotection during ischemia. METHODS: Myocardial ischemia/reperfusion (I/R; 30 min/24 h) and hypoxia/reoxygenation (H/R; 24 h/4 h) were used in this study. Confocal microscopy and transmission electron microscope (TEM) were used to observe mitochondrial morphology. RESULTS: Claudin-5 was detected in murine heart tissue and neonatal rat cardiomyocytes (NRCM). Its protein level was severely decreased after myocardial I/R or H/R. Confocal microscopy showedclaudin-5 presented in the mitochondria of NRCM. H/R-induced claudin-5 downregulation was accompanied by mitochondrial fragmentation. The mitofusin 2 (Mfn2) expressionwas dramatically decreased while the dynamin-related protein (Drp) 1 expression was significantly increased after H/R. The TEM indicatedH/R-induced mitochondrial swelling and fission. Adenoviral claudin-5 overexpression reversed these structural disintegration of mitochondria. The mitochondria-centered intrinsic pathway of apoptosis triggered by H/R and indicated by the cytochrome c and cleaved caspase 3 in the cytoplasm of NRCMs was also reduced by overexpressing claudin-5. Claudin-5 overexpression in mouse heart also significantly decreased cleaved caspase 3 and the infarct size in ischemic heart with improved systolic function. CONCLUSION: We demonstrated for the first time the presence of claudin-5 in the mitochondria in cardiomyocytes and provided the firm evidence for the cardioprotective role of claudin-5 in the preservation of mitochondrial dynamics and cell fate against hypoxia- or ischemia-induced stress.


Subject(s)
Claudin-5/genetics , Hypoxia/prevention & control , Mitochondria, Heart/genetics , Mitochondrial Dynamics/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Apoptosis , Cells, Cultured , Claudin-5/biosynthesis , Dynamins/biosynthesis , Dynamins/genetics , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Hypoxia/genetics , Hypoxia/pathology , Membrane Proteins , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/ultrastructure , Rats , Rats, Sprague-Dawley
16.
Front Endocrinol (Lausanne) ; 12: 651211, 2021.
Article in English | MEDLINE | ID: mdl-33868178

ABSTRACT

Lipid overload of the mitochondria is linked to the development of insulin resistance in skeletal muscle which may be a contributing factor to the progression of type 2 diabetes during obesity. The targeted degradation of mitochondria through autophagy, termed mitophagy, contributes to the mitochondrial adaptive response to changes in dietary fat. Our previous work demonstrates long-term (2-4 months) consumption of a high-fat diet increases mitochondrial lipid oxidation capacity but does not alter markers of mitophagy in mice. The purpose of this study was to investigate initial stages of mitochondrial respiratory adaptations to high-fat diet and the activation of mitophagy. C57BL/6J mice consumed either a low-fat diet (LFD, 10% fat) or high-fat diet (HFD, 60% fat) for 3 or 7 days. We measured skeletal muscle mitochondrial respiration and protein markers of mitophagy in a mitochondrial-enriched fraction of skeletal muscle. After 3 days of HFD, mice had lower lipid-supported oxidative phosphorylation alongside greater electron leak compared with the LFD group. After 7 days, there were no differences in mitochondrial respiration between diet groups. HFD mice had greater autophagosome formation potential (Beclin-1) and greater activation of mitochondrial autophagy receptors (Bnip3, p62) in isolated mitochondria, but no difference in downstream autophagosome (LC3II) or lysosome (Lamp1) abundance after both 3 and 7 days compared with the LFD groups. In cultured myotubes, palmitate treatment decreased mitochondrial membrane potential and hydrogen peroxide treatment increased accumulation of upstream mitophagy markers. We conclude that several days of high-fat feeding stimulated upstream activation of skeletal muscle mitophagy, potentially through lipid-induced oxidative stress, without downstream changes in respiration.


Subject(s)
Lipids/chemistry , Mitochondria/pathology , Mitophagy/physiology , Muscle, Skeletal/physiology , Animals , Autophagy , Beclin-1/biosynthesis , Diabetes Mellitus, Type 2/genetics , Diet, Fat-Restricted , Diet, High-Fat , Fatty Acids/chemistry , Fatty Acids/metabolism , Hydrogen Peroxide/chemistry , Lipid Peroxidation , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Obesity/genetics , Oxidative Stress , Oxygen/chemistry , Phenotype , Reactive Oxygen Species , Time Factors
17.
Physiol Rep ; 9(6): e14797, 2021 03.
Article in English | MEDLINE | ID: mdl-33769716

ABSTRACT

AIM: Exercise is able to increase both muscle protein synthesis and mitochondrial biogenesis. However, acidosis, which can occur in pathological states as well as during high-intensity exercise, can decrease mitochondrial function, whilst its impact on muscle protein synthesis is disputed. Thus, the aim of this study was to determine the effect of a mild physiological decrease in pH, by administration of ammonium chloride, on myofibrillar and mitochondrial protein synthesis, as well as associated molecular signaling events. METHODS: Male Wistar rats were given either a placebo or ammonium chloride prior to a short interval training session. Rats were killed before exercise, immediately after exercise, or 3 h after exercise. RESULTS: Myofibrillar (p = 0.036) fractional protein synthesis rates was increased immediately after exercise in the soleus muscle of the placebo group, but this effect was absent in the ammonium chloride group. However, in the gastrocnemius muscle NH4 Cl increased myofibrillar (p = 0.044) and mitochondrial protein synthesis (0 h after exercise p = 0.01; 3 h after exercise p = 0.003). This was accompanied by some small differences in protein phosphorylation and mRNA expression. CONCLUSION: This study found ammonium chloride administration immediately prior to a single session of exercise in rats had differing effects on mitochondrial and myofibrillar protein synthesis rates in soleus (type I) and gastrocnemius (type II) muscle in rats.


Subject(s)
Acidosis/metabolism , Ammonium Chloride/administration & dosage , Mitochondrial Proteins/biosynthesis , Muscle Proteins/biosynthesis , Myofibrils/metabolism , Physical Conditioning, Animal , Animals , Male , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myofibrils/drug effects , Rats, Wistar
18.
Tissue Cell ; 71: 101516, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33744742

ABSTRACT

Diabetes mellitus (DM) is a worldwide health problem. The Micro- and macro-vascular complications are the major causes of morbidity and mortality of DM. Molecular regulation of mitochondrial fission/fusion cycles is being studied, but the results were not conclusive. The aim of this study is to investigate the possible functional role of lncRNA H19 and its relation to mitofusin-2 (Mfn-2) gene expression in diabetic rats with cardiac and renal complications. Streptozotocin-induced diabetic male, albino rats and a matched control group were investigated. Cardiac weights, blood pressure and ECG were recorded. Biochemical evaluation of cardiac and renal functions was performed. Molecular determination of lncRNA H19 and Mfn-2 gene expression and histological examination by light and electron microscopy for cardiac and renal tissues were performed. Diabetic rats showed a significant increase of left ventricle weight/whole body weight ratio, R wave voltage, and a significant decrease of blood pressure, heart rate, and P wave voltage. At the molecular level, lncRNA H19 and Mfn-2 mRNA showed altered expression with a statistically significant downregulation of Mfn-2 mRNA expression in renal tissues. In conclusion, the changes in lncRNA H19 and Mfn-2 mRNA expression may help better understanding of the pathogenesis of cardiac and renal dysfunctions associated with type 1 DM.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , GTP Phosphohydrolases/biosynthesis , Gene Expression Regulation , Mitochondria, Heart/metabolism , Mitochondrial Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Kidney/metabolism , Kidney/pathology , Male , Myocardium/metabolism , Myocardium/pathology , Rats
19.
Cell Death Dis ; 12(4): 325, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33771980

ABSTRACT

TEFM (transcription elongation factor of mitochondria) has been identified as a novel nuclear-encoded transcription elongation factor in the transcription of mitochondrial genome. Our bioinformatics analysis of TCGA data revealed an aberrant over-expression of TEFM in hepatocellular carcinoma (HCC). We analyzed its biological effects and clinical significance in this malignancy. TEFM expression was analyzed by quantitative real-time PCR, western blot, and immunohistochemistry analysis in HCC tissues and cell lines. The effects of TEFM on HCC cell growth and metastasis were determined by cell proliferation, colony formation, flow cytometric cell cycle and apoptosis, migration, and invasion assays. TEFM expression was significantly increased in HCC tissues mainly caused by down-regulation of miR-194-5p. Its increased expression is correlated with poor prognosis of HCC patients. TEFM promoted HCC growth and metastasis both in vitro and in vivo by promoting G1-S cell transition, epithelial-to-mesenchymal transition (EMT), and suppressing cell apoptosis. Mechanistically, TEFM exerts its tumor growth and metastasis promoting effects at least partly through increasing ROS production and subsequently by activation of ERK signaling. Our study suggests that TEFM functions as a vital oncogene in promoting growth and metastasis in HCC and may contribute to the targeted therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Mitochondrial Proteins/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Neoplasm Metastasis , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670365

ABSTRACT

MicroRNAs (miRNAs) are attractive therapeutic targets and promising candidates as molecular biomarkers for various therapy-resistant tumors. However, the association between miRNAs and drug resistance in melanoma remains to be elucidated. We used an integrative genomic analysis to comprehensively study the miRNA expression profiles of drug-resistant melanoma patients and cell lines. MicroRNA-181a and -181b (miR181a/b) were identified as the most significantly down-regulated miRNAs in resistant melanoma patients and cell lines. Re-establishment of miR-181a/b expression reverses the resistance of melanoma cells to the BRAF inhibitor dabrafenib. Introduction of miR-181 mimics markedly decreases the expression of TFAM in A375 melanoma cells resistant to BRAF inhibitors. Furthermore, melanoma growth was inhibited in A375 and M14 resistant melanoma cells transfected with miR-181a/b mimics, while miR-181a/b depletion enhanced resistance in sensitive cell lines. Collectively, our study demonstrated that miR-181a/b could reverse the resistance to BRAF inhibitors in dabrafenib resistant melanoma cell lines. In addition, miR-181a and -181b are strongly down-regulated in tumor samples from patients before and after the development of resistance to targeted therapies. Finally, melanoma tissues with high miR-181a and -181b expression presented favorable outcomes in terms of Progression Free Survival, suggesting that miR-181 is a clinically relevant candidate for therapeutic development or biomarker-based therapy selection.


Subject(s)
DNA-Binding Proteins/biosynthesis , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , MicroRNAs/biosynthesis , Mitochondrial Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/biosynthesis , Transcription Factors/biosynthesis , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Genomics , Humans , Male , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , Mitochondrial Proteins/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...