Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.553
Filter
1.
Nat Commun ; 15(1): 5446, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937458

ABSTRACT

Mitochondrial transcription factor A (TFAM) employs DNA bending to package mitochondrial DNA (mtDNA) into nucleoids and recruit mitochondrial RNA polymerase (POLRMT) at specific promoter sites, light strand promoter (LSP) and heavy strand promoter (HSP). Herein, we characterize the conformational dynamics of TFAM on promoter and non-promoter sequences using single-molecule fluorescence resonance energy transfer (smFRET) and single-molecule protein-induced fluorescence enhancement (smPIFE) methods. The DNA-TFAM complexes dynamically transition between partially and fully bent DNA conformational states. The bending/unbending transition rates and bending stability are DNA sequence-dependent-LSP forms the most stable fully bent complex and the non-specific sequence the least, which correlates with the lifetimes and affinities of TFAM with these DNA sequences. By quantifying the dynamic nature of the DNA-TFAM complexes, our study provides insights into how TFAM acts as a multifunctional protein through the DNA bending states to achieve sequence specificity and fidelity in mitochondrial transcription while performing mtDNA packaging.


Subject(s)
DNA Packaging , DNA, Mitochondrial , DNA-Binding Proteins , Fluorescence Resonance Energy Transfer , Mitochondrial Proteins , Nucleic Acid Conformation , Promoter Regions, Genetic , Transcription Factors , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/chemistry , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Transcription Initiation, Genetic , Mitochondria/metabolism , Mitochondria/genetics , Single Molecule Imaging , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Base Sequence , Protein Binding
2.
Biomolecules ; 14(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38927107

ABSTRACT

Alterations in mitochondrial function have been linked to a variety of cellular and organismal stress responses including apoptosis, aging, neurodegeneration and tumorigenesis. However, adaptation to mitochondrial dysfunction can occur through the activation of survival pathways, whose mechanisms are still poorly understood. The yeast Saccharomyces cerevisiae is an invaluable model organism for studying how mitochondrial dysfunction can affect stress response and adaptation processes. In this study, we analyzed and compared in the absence and in the presence of osmostress wild-type cells with two models of cells lacking mitochondrial DNA: ethidium bromide-treated cells (ρ0) and cells lacking the mitochondrial pyrimidine nucleotide transporter RIM2 (ΔRIM2). Our results revealed that the lack of mitochondrial DNA provides an advantage in the kinetics of stress response. Additionally, wild-type cells exhibited higher osmosensitivity in the presence of respiratory metabolism. Mitochondrial mutants showed increased glycerol levels, required in the short-term response of yeast osmoadaptation, and prolonged oxidative stress. The involvement of the mitochondrial retrograde signaling in osmoadaptation has been previously demonstrated. The expression of CIT2, encoding the peroxisomal isoform of citrate synthase and whose up-regulation is prototypical of RTG pathway activation, appeared to be increased in the mutants. Interestingly, selected TCA cycle genes, CIT1 and ACO1, whose expression depends on RTG signaling upon stress, showed a different regulation in ρ0 and ΔRIM2 cells. These data suggest that osmoadaptation can occur through different mechanisms in the presence of mitochondrial defects and will allow us to gain insight into the relationships among metabolism, mitochondria-mediated stress response, and cell adaptation.


Subject(s)
DNA, Mitochondrial , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Mitochondria/metabolism , Mitochondria/genetics , Adaptation, Physiological/genetics , Oxidative Stress/genetics , Glycerol/metabolism , Ethidium/metabolism
3.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928146

ABSTRACT

Mitochondrial quality control is essential in mitochondrial function. To examine the importance of Parkin-dependent mechanisms in mitochondrial quality control, we assessed the impact of modulating Parkin on proteome flux and mitochondrial function in a context of reduced mtDNA fidelity. To accomplish this, we crossed either the Parkin knockout mouse or ParkinW402A knock-in mouse lines to the Polg mitochondrial mutator line to generate homozygous double mutants. In vivo longitudinal isotopic metabolic labeling was followed by isolation of liver mitochondria and synaptic terminals from the brain, which are rich in mitochondria. Mass spectrometry and bioenergetics analysis were assessed. We demonstrate that slower mitochondrial protein turnover is associated with loss of mtDNA fidelity in liver mitochondria but not synaptic terminals, and bioenergetic function in both tissues is impaired. Pathway analysis revealed loss of mtDNA fidelity is associated with disturbances of key metabolic pathways, consistent with its association with metabolic disorders and neurodegeneration. Furthermore, we find that loss of Parkin leads to exacerbation of Polg-driven proteomic consequences, though it may be bioenergetically protective in tissues exhibiting rapid mitochondrial turnover. Finally, we provide evidence that, surprisingly, dis-autoinhibition of Parkin (ParkinW402A) functionally resembles Parkin knockout and fails to rescue deleterious Polg-driven effects. Our study accomplishes three main outcomes: (1) it supports recent studies suggesting that Parkin dependence is low in response to an increased mtDNA mutational load, (2) it provides evidence of a potential protective role of Parkin insufficiency, and (3) it draws into question the therapeutic attractiveness of enhancing Parkin function.


Subject(s)
DNA Polymerase gamma , DNA, Mitochondrial , Mice, Knockout , Mutation , Ubiquitin-Protein Ligases , Animals , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mice , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Proteomics/methods , Proteome/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics
4.
Cell Commun Signal ; 22(1): 328, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872145

ABSTRACT

BACKGROUND: Kawasaki disease (KD) is an immune vasculitis of unknown origin, characterized by transient inflammation. The activation of the cGAS-STING pathway, triggered by mitochondrial DNA (mtDNA) release, has been implicated in the onset of KD. However, its specific role in the progression of inflammation during KD's acute phase remains unclear. METHODS: We measured mtDNA and 2'3'-cGAMP expression in KD patient serum using RT-qPCR and ELISA. A murine model of KD was induced by injecting Lactobacillus casei cell wall extract (LCWE), after which cGAS-STING pathway activation and inflammatory markers were assessed via immunohistochemistry, western blot, and RT-qPCR. Human umbilical vein endothelial cells (HUVECs) were treated with KD serum and modulators of the cGAS-STING pathway for comparative analysis. Mitochondrial function was evaluated using Mitosox staining, mPTP opening was quantified by fluorescence microscopy, and mitochondrial membrane potential (MMP) was determined with JC-1 staining. RESULTS: KD patient serum exhibited increased mtDNA and 2'3'-cGAMP expression, with elevated levels of pathway-related proteins and inflammatory markers observed in both in vivo and in vitro models. TEM confirmed mitochondrial damage, and further studies demonstrated that inhibition of mPTP opening reduced mtDNA release, abrogated cGAS-STING pathway activation, and mitigated inflammation. CONCLUSION: These findings indicate that mtDNA released through the mPTP is a critical activator of the cGAS-STING pathway, contributing significantly to KD-associated inflammation. Targeting mtDNA release or the cGAS-STING pathway may offer novel therapeutic approaches for KD management.


Subject(s)
DNA, Mitochondrial , Inflammation , Membrane Proteins , Mitochondrial Permeability Transition Pore , Mucocutaneous Lymph Node Syndrome , Nucleotidyltransferases , Signal Transduction , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Mucocutaneous Lymph Node Syndrome/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Animals , Mitochondrial Permeability Transition Pore/metabolism , Male , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Female , Acute Disease , Mice, Inbred C57BL , Child, Preschool
5.
Biochem Soc Trans ; 52(3): 1243-1251, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38884788

ABSTRACT

Mitochondrial DNA replication is initiated by the transcription of mitochondrial RNA polymerase (mtRNAP), as mitochondria lack a dedicated primase. However, the mechanism determining the switch between continuous transcription and premature termination to generate RNA primers for mitochondrial DNA (mtDNA) replication remains unclear. The pentatricopeptide repeat domain of mtRNAP exhibits exoribonuclease activity, which is required for the initiation of mtDNA replication in Drosophila. In this review, we explain how this exonuclease activity contributes to primer synthesis in strand-coupled mtDNA replication, and discuss how its regulation might co-ordinate mtDNA replication and transcription in both Drosophila and mammals.


Subject(s)
DNA Replication , DNA, Mitochondrial , Mitochondria , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Animals , Mitochondria/metabolism , Mitochondria/genetics , Humans , DNA-Directed RNA Polymerases/metabolism , Transcription, Genetic , Drosophila/genetics , Drosophila/metabolism , Exoribonucleases/metabolism , Exoribonucleases/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
6.
J Exp Clin Cancer Res ; 43(1): 158, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38825680

ABSTRACT

BACKGROUND: Studies have shown that oxidative stress and its resistance plays important roles in the process of tumor metastasis, and mitochondrial dysfunction caused by mitochondrial DNA (mtDNA) damage is an important molecular event in oxidative stress. In lung cancer, the normal fibroblasts (NFs) are activated as cancer-associated fibroblasts (CAFs), and act in the realms of the tumor microenvironment (TME) with consequences for tumor growth and metastasis. However, its activation mechanism and whether it participates in tumor metastasis through antioxidative stress remain unclear. METHODS: The role and signaling pathways of tumor cell derived extracellular vesicles (EVs) activating NFs and the characteristic of induced CAFs (iCAFs) were measured by the transmission electron microscopy, nanoparticle tracking analysis, immunofluorescence, collagen contraction assay, quantitative PCR, immunoblotting, luciferase reporter assay and mitochondrial membrane potential detection. Mitochondrial genome and single nucleotide polymorphism sequencing were used to investigate the transport of mtDNA from iCAFs to ρ0 cells, which were tumor cells with mitochondrial dysfunction caused by depletion of mtDNA. Further, the effects of iCAFs on mitochondrial function, growth and metastasis of tumor cells were analysed in co-culture models both in vitro and in vivo, using succinate dehydrogenase, glutathione and oxygen consumption rate measurements, CCK-8 assay, transwell assay, xenotransplantation and metastasis experiments as well as in situ hybridization and immunohistochemistry. RESULTS: Our findings revealed that EVs derived from high-metastatic lung cancer cells packaged miR-1290 that directly targets MT1G, leading to activation of AKT signaling in NFs and inducing NFs conversion to CAFs. The iCAFs exhibit higher levels of autophagy and mitophagy and more mtDNA release, and reactive oxygen species (ROS) could further promote this process. After cocultured with the conditioned medium (CM) of iCAFs, the ρ0 cells may restore its mitochondrial function by acquisition of mtDNA from CAFs, and further promotes tumor metastasis. CONCLUSIONS: These results elucidate a novel mechanism that CAFs activated by tumor-derived EVs can promote metastasis by transferring mtDNA and restoring mitochondrial function of tumor cells which result in resistance of oxidative stress, and provide a new therapeutic target for lung cancer metastasis.


Subject(s)
Cancer-Associated Fibroblasts , DNA, Mitochondrial , Extracellular Vesicles , Lung Neoplasms , Mitophagy , Extracellular Vesicles/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Humans , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Mice , Animals , Neoplasm Metastasis , Cell Line, Tumor , Tumor Microenvironment
7.
Birth Defects Res ; 116(6): e2369, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38877673

ABSTRACT

BACKGROUND: Prenatal alcohol exposure (PAE) has been linked to congenital heart disease and fetal alcohol syndrome. The heart primarily relies on mitochondria to generate energy, so impaired mitochondrial function due to alcohol exposure can significantly affect cardiac development and function. Our study aimed to investigate the impact of PAE on myocardial and mitochondrial functions in offspring mice. METHODS: We administered 30% alcohol (3 g/kg) to pregnant C57BL/6 mice during the second trimester. We assessed cardiac function by transthoracic echocardiography, observed myocardial structure and fibrosis through staining tests and electron transmission microscopy, and detected cardiomyocyte apoptosis with dUTP nick end labeling assay and real-time quantitative PCR. Additionally, we measured the reactive oxygen species content, ATP level, and mitochondrial DNA copy number in myocardial mitochondria. Mitochondrial damage was evaluated by assessing the level of mitochondrial membrane potential and the opening degree of mitochondrial permeability transition pores. RESULTS: Our findings revealed that PAE caused cardiac systolic dysfunction, ventricular enlargement, thinned ventricular wall, cardiac fibrosis in the myocardium, scattered loss of cardiomyocytes, and disordered arrangement of myocardial myotomes in the offspring. Furthermore, we observed a significant increase in mitochondrial reactive oxygen species content, a decrease in mitochondrial membrane potential, ATP level, and mitochondrial DNA copy number, and sustained opening of mitochondrial permeability transition pores in the heart tissues of the offspring. CONCLUSIONS: These results indicated that PAE had adverse effects on the cardiac structure and function of the newborn mice and could trigger oxidative stress in their myocardia and contribute to mitochondrial dysfunction.


Subject(s)
Ethanol , Mice, Inbred C57BL , Myocytes, Cardiac , Prenatal Exposure Delayed Effects , Reactive Oxygen Species , Animals , Female , Pregnancy , Mice , Prenatal Exposure Delayed Effects/metabolism , Reactive Oxygen Species/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Ethanol/adverse effects , Ethanol/toxicity , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Apoptosis/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Membrane Potential, Mitochondrial/drug effects , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Echocardiography , Fibrosis
8.
FASEB J ; 38(13): e23695, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38885021

ABSTRACT

H. B. Suliman, K. E. Welty-Wolf, M. S. Carraway, D. A. Schwartz, J. W. Hollingsworth, C. A. Piantadosi, "Toll-like receptor 4 mediates mitochondrial DNA damage and biogenic responses after heat-inactivated E. coli," The FASEB Journal 19, no. 11 (2005): 1531-1533, https://doi.org/10.1096/fj.04-3500fje. This expression of concern is for the above article, published online on July 1, 2005, in Wiley Online Library (wileyonlinelibrary.com), and has been published by agreement between the journal's Editor-in-Chief, Loren E. Wold, Federation of American Societies for Experimental Biology, and Wiley Periodicals LLC. The expression of concern is being published due to concerns raised by a third party regarding suspected image manipulation. The authors have been contacted, and the journal has requested an investigation from the authors' institution, Duke University. The journal is issuing an expression of concern to alert the readers to these concerns while an investigation is taking place. A subsequent note will be published following the institutional investigation.


Subject(s)
DNA Damage , DNA, Mitochondrial , Escherichia coli , Toll-Like Receptor 4 , Escherichia coli/metabolism , Escherichia coli/genetics , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Humans , Toll-Like Receptor 4/metabolism , Hot Temperature , Animals
9.
Molecules ; 29(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38893565

ABSTRACT

L-theanine, a unique non-protein amino acid, is an important bioactive component of green tea. Previous studies have shown that L-theanine has many potent health benefits, such as anti-anxiety effects, regulation of the immune response, relaxing neural tension, and reducing oxidative damage. However, little is known concerning whether L-theanine can improve the clearance of mitochondrial DNA (mtDNA) damage in organisms. Here, we reported that L-theanine treatment increased ATP production and improved mitochondrial morphology to extend the lifespan of UVC-exposed nematodes. Mechanistic investigations showed that L-theanine treatment enhanced the removal of mtDNA damage and extended lifespan by activating autophagy, mitophagy, mitochondrial dynamics, and mitochondrial unfolded protein response (UPRmt) in UVC-exposed nematodes. In addition, L-theanine treatment also upregulated the expression of genes related to mitochondrial energy metabolism in UVC-exposed nematodes. Our study provides a theoretical basis for the possibility that tea drinking may prevent mitochondrial-related diseases.


Subject(s)
Caenorhabditis elegans , Glutamates , Longevity , Mitochondria , Ultraviolet Rays , Animals , Caenorhabditis elegans/drug effects , Glutamates/pharmacology , Ultraviolet Rays/adverse effects , Longevity/drug effects , Longevity/radiation effects , Mitochondria/metabolism , Mitochondria/drug effects , DNA, Mitochondrial/metabolism , Autophagy/drug effects , DNA Damage/drug effects , Mitophagy/drug effects , Unfolded Protein Response/drug effects , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/radiation effects , Adenosine Triphosphate/metabolism , Signal Transduction/drug effects , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics
10.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891898

ABSTRACT

The consequences of stroke include cognitive deficits and sensorimotor disturbances, which are largely related to mitochondrial impairments in the brain. In this work, we have shown that the mimetic of the ketogenic diet beta-hydroxybutyrate (ßHB) can improve neurological brain function in stroke. At 3 weeks after photothrombotic stroke, mice receiving ßHB with drinking water before and after surgery recovered faster in terms of sensorimotor functions assessed by the string test and static rods and cognitive functions assessed by the Morris water maze. At the same time, the ßHB-treated mice had lower expression of some markers of astrocyte activation and inflammation (Gfap, Il-1b, Tnf). We hypothesize that long-term administration of ßHB promotes the activation of the nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf2/ARE) pathway, which leads to increased expression of antioxidant genes targeting mitochondria and genes involved in signaling pathways necessary for the maintenance of synaptic plasticity. ßHB partially maintained mitochondrial DNA (mtDNA) integrity during the first days after photothrombosis. However, in the following three weeks, the number of mtDNA damages increased in all experimental groups, which coincided with a decrease in Ogg1 expression, which plays an important role in mtDNA repair. Thus, we can assume that ßHB is not only an important metabolite that provides additional energy to brain tissue during recovery from stroke under conditions of mitochondrial damage but also an important signaling molecule that supports neuronal plasticity and reduces neuroinflammation.


Subject(s)
3-Hydroxybutyric Acid , Cognitive Dysfunction , Ischemic Stroke , Animals , Mice , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/drug therapy , Ischemic Stroke/metabolism , Ischemic Stroke/complications , Male , Disease Models, Animal , NF-E2-Related Factor 2/metabolism , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Thrombosis/metabolism , Thrombosis/etiology , Brain/metabolism , Brain/drug effects , Brain/pathology , Mice, Inbred C57BL
11.
Sci Rep ; 14(1): 13015, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844752

ABSTRACT

Calcium hydroxide (Ca(OH)2NPs), calcium titanate (CaTiO3NPs) and yttrium oxide (Y2O3NPs) nanoparticles are prevalent in many industries, including food and medicine, but their small size raises concerns about potential cellular damage and genotoxic effects. However, there are very limited studies available on their genotoxic effects. Hence, this was done to investigate the effects of multiple administration of Ca(OH)2NPs, CaTiO3NPs or/and Y2O3NPs on genomic DNA stability, mitochondrial membrane potential integrity and inflammation induction in mouse brain tissues. Mice were orally administered Ca(OH)2NPs, CaTiO3NPs or/and Y2O3NPs at a dose level of 50 mg/kg b.w three times a week for 2 weeks. Genomic DNA integrity was studied using Comet assay and the level of reactive oxygen species (ROS) within brain cells was analyzed using 2,7 dichlorofluorescein diacetate dye. The expression level of Presenilin-1, tumor necrosis factor-alpha (TNF-α) and Interleukin-6 (IL-6) genes and the integrity of the mitochondrial membrane potential were also detected. Oral administration of Ca(OH)2NPs caused the highest damage to genomic DNA and mitochondrial membrane potential, less genomic DNA and mitochondrial damage was induced by CaTiO3NPs administration while administration of Y2O3NPs did not cause any remarkable change in the integrity of genomic DNA and mitochondrial membrane potential. Highest ROS generation and upregulation of presenilin-1, TNF-α and IL-6 genes were also observed within the brain cells of mice administrated Ca(OH)2NPs but Y2O3NPs administration almost caused no changes in ROS generation and genes expression compared to the negative control. Administration of CaTiO3NPs alone slightly increased ROS generation and the expression level of TNF-α and IL-6 genes. Moreover, no remarkable changes in the integrity of genomic DNA and mitochondrial DNA potential, ROS level and the expression level of presenilin-1, TNF-α and IL-6 genes were noticed after simultaneous coadministration of Y2O3NPs with Ca(OH)2NPs and CaTiO3NPs. Coadministration of Y2O3NPs with Ca(OH)2NPs and CaTiO3NPs mitigated Ca(OH)2NPs and CaTiO3NPs induced ROS generation, genomic DNA damage and inflammation along with restoring the integrity of mitochondrial membrane potential through Y2O3NPs scavenging free radicals ability. Therefore, further studies are recommended to study the possibility of using Y2O3NPs to alleviate Ca(OH)2NPs and CaTiO3NPs induced genotoxic effects.


Subject(s)
Calcium Hydroxide , DNA Damage , Inflammation , Membrane Potential, Mitochondrial , Nanoparticles , Reactive Oxygen Species , Titanium , Yttrium , Animals , Reactive Oxygen Species/metabolism , Mice , DNA Damage/drug effects , Calcium Hydroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Titanium/chemistry , Titanium/toxicity , Inflammation/metabolism , Inflammation/pathology , Yttrium/chemistry , Nanoparticles/chemistry , Mitochondria/metabolism , Mitochondria/drug effects , Male , Brain/metabolism , Brain/drug effects , Brain/pathology , DNA, Mitochondrial/metabolism
12.
Int J Biol Sci ; 20(8): 2860-2880, 2024.
Article in English | MEDLINE | ID: mdl-38904024

ABSTRACT

Mitochondrial diseases are associated with neuronal death and mtDNA depletion. Astrocytes respond to injury or stimuli and damage to the central nervous system. Neurodegeneration can cause astrocytes to activate and acquire toxic functions that induce neuronal death. However, astrocyte activation and its impact on neuronal homeostasis in mitochondrial disease remain to be explored. Using patient cells carrying POLG mutations, we generated iPSCs and then differentiated these into astrocytes. POLG astrocytes exhibited mitochondrial dysfunction including loss of mitochondrial membrane potential, energy failure, loss of complex I and IV, disturbed NAD+/NADH metabolism, and mtDNA depletion. Further, POLG derived astrocytes presented an A1-like reactive phenotype with increased proliferation, invasion, upregulation of pathways involved in response to stimulus, immune system process, cell proliferation and cell killing. Under direct and indirect co-culture with neurons, POLG astrocytes manifested a toxic effect leading to the death of neurons. We demonstrate that mitochondrial dysfunction caused by POLG mutations leads not only to intrinsic defects in energy metabolism affecting both neurons and astrocytes, but also to neurotoxic damage driven by astrocytes. These findings reveal a novel role for dysfunctional astrocytes that contribute to the pathogenesis of POLG diseases.


Subject(s)
Astrocytes , DNA Polymerase gamma , DNA-Directed DNA Polymerase , Mitochondria , Mutation , Astrocytes/metabolism , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , Humans , Mitochondria/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Neurons/metabolism , Membrane Potential, Mitochondrial , Induced Pluripotent Stem Cells/metabolism , Cells, Cultured , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Coculture Techniques
13.
Cell Death Dis ; 15(6): 405, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858390

ABSTRACT

Genetic mutations causing primary mitochondrial disease (i.e those compromising oxidative phosphorylation [OxPhos]) resulting in reduced bioenergetic output display great variability in their clinical features, but the reason for this is unknown. We hypothesized that disruption of the communication between endoplasmic reticulum (ER) and mitochondria at mitochondria-associated ER membranes (MAM) might play a role in this variability. To test this, we assayed MAM function and ER-mitochondrial communication in OxPhos-deficient cells, including cybrids from patients with selected pathogenic mtDNA mutations. Our results show that each of the various mutations studied indeed altered MAM functions, but notably, each disorder presented with a different MAM "signature". We also found that mitochondrial membrane potential is a key driver of ER-mitochondrial connectivity. Moreover, our findings demonstrate that disruption in ER-mitochondrial communication has consequences for cell survivability that go well beyond that of reduced ATP output. The findings of a "MAM-OxPhos" axis, the role of mitochondrial membrane potential in controlling this process, and the contribution of MAM dysfunction to cell death, reveal a new relationship between mitochondria and the rest of the cell, as well as providing new insights into the diagnosis and treatment of these devastating disorders.


Subject(s)
Endoplasmic Reticulum , Membrane Potential, Mitochondrial , Mitochondria , Mitochondrial Diseases , Oxidative Phosphorylation , Humans , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mutation/genetics , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics
14.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892349

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease affecting mostly women of child-bearing age. Immune dysfunction in SLE results from disrupted apoptosis which lead to an unregulated interferon (IFN) stimulation and the production of autoantibodies, leading to immune complex formation, complement activation, and organ damage. Lupus nephritis (LN) is a common and severe complication of SLE, impacting approximately 30% to 40% of SLE patients. Recent studies have demonstrated an alteration in mitochondrial homeostasis in SLE patients. Mitochondrial dysfunction contributes significantly to SLE pathogenesis by enhancing type 1 IFN production through various pathways involving neutrophils, platelets, and T cells. Defective mitophagy, the process of clearing damaged mitochondria, exacerbates this cycle, leading to increased immune dysregulation. In this review, we aim to detail the physiopathological link between mitochondrial dysfunction and disease activity in SLE. Additionally, we will explore the potential role of mitochondria as biomarkers and therapeutic targets in SLE, with a specific focus on LN. In LN, mitochondrial abnormalities are observed in renal cells, correlating with disease progression and renal fibrosis. Studies exploring cell-free mitochondrial DNA as a biomarker in SLE and LN have shown promising but preliminary results, necessitating further validation and standardization. Therapeutically targeting mitochondrial dysfunction in SLE, using drugs like metformin or mTOR inhibitors, shows potential in modulating immune responses and improving clinical outcomes. The interplay between mitochondria, immune dysregulation, and renal involvement in SLE and LN underscores the need for comprehensive research and innovative therapeutic strategies. Understanding mitochondrial dynamics and their impact on immune responses offers promising avenues for developing personalized treatments and non-invasive biomarkers, ultimately improving outcomes for LN patients.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Mitochondria , Humans , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/etiology , Mitochondria/metabolism , Mitochondria/pathology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/immunology , DNA, Mitochondrial/metabolism , Animals , Biomarkers , Mitophagy
15.
Front Biosci (Landmark Ed) ; 29(6): 218, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38940042

ABSTRACT

Mitochondrial DNA (mtDNA) is located in the mitochondrial matrix, in close proximity to major sources of reactive oxygen species (ROS) in the cell. This makes mtDNA one of the most susceptible components to damage in the cell. The nuclear factor E2-related factor 2/antioxidant response element (Nrf2/ARE) signaling pathway is an important cytoprotective mechanism. It is well-studied and described that Nrf2 can regulate the expression of mitochondrial-targeted antioxidant systems in the cell, indirectly protecting mtDNA from damage. However, the Nrf2/ARE pathway can also directly impact on the mtDNA repair processes. In this review, we summarize the existing data on the impact of Nrf2 on mtDNA repair, primarily base excision repair (BER), as it is considered the main repair pathway for the mitochondrial genome. We explore the crosstalk between Nrf2/ARE, BRCA1, and p53 signaling pathways in their involvement in maintaining mtDNA integrity. The role of other repair mechanisms in correcting mismatched bases and double-strand breaks is discussed. Additionally, the review addresses the role of Nrf2 in the repair of noncanonical bases, which contribute to an increased number of mutations in mtDNA and can contaminate the nucleotide pool.


Subject(s)
Antioxidant Response Elements , DNA Repair , DNA, Mitochondrial , NF-E2-Related Factor 2 , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Antioxidant Response Elements/genetics , Animals , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , DNA Damage
16.
Nat Cell Biol ; 26(6): 878-891, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783142

ABSTRACT

When cells are stressed, DNA from energy-producing mitochondria can leak out and drive inflammatory immune responses if not cleared. Cells employ a quality control system called autophagy to specifically degrade damaged components. We discovered that mitochondrial transcription factor A (TFAM)-a protein that binds mitochondrial DNA (mtDNA)-helps to eliminate leaked mtDNA by interacting with the autophagy protein LC3 through an autolysosomal pathway (we term this nucleoid-phagy). TFAM contains a molecular zip code called the LC3 interacting region (LIR) motif that enables this binding. Although mutating TFAM's LIR motif did not affect its normal mitochondrial functions, more mtDNA accumulated in the cell cytoplasm, activating inflammatory signalling pathways. Thus, TFAM mediates autophagic removal of leaked mtDNA to restrict inflammation. Identifying this mechanism advances understanding of how cells exploit autophagy machinery to selectively target and degrade inflammatory mtDNA. These findings could inform research on diseases involving mitochondrial damage and inflammation.


Subject(s)
Autophagy , DNA, Mitochondrial , DNA-Binding Proteins , Inflammation , Mitochondria , Mitochondrial Proteins , Transcription Factors , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Animals , Humans , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondria/metabolism , Mitochondria/genetics , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Protein Binding , Cytoplasm/metabolism , Lysosomes/metabolism , Signal Transduction , HEK293 Cells , Mice, Inbred C57BL , High Mobility Group Proteins
18.
Cell Metab ; 36(7): 1586-1597.e7, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38703762

ABSTRACT

The mitochondrial genome transcribes 13 mRNAs coding for well-known proteins essential for oxidative phosphorylation. We demonstrate here that cytochrome b (CYTB), the only mitochondrial-DNA-encoded transcript among complex III, also encodes an unrecognized 187-amino-acid-long protein, CYTB-187AA, using the standard genetic code of cytosolic ribosomes rather than the mitochondrial genetic code. After validating the existence of this mtDNA-encoded protein arising from cytosolic translation (mPACT) using mass spectrometry and antibodies, we show that CYTB-187AA is mainly localized in the mitochondrial matrix and promotes the pluripotent state in primed-to-naive transition by interacting with solute carrier family 25 member 3 (SLC25A3) to modulate ATP production. We further generated a transgenic knockin mouse model of CYTB-187AA silencing and found that reduction of CYTB-187AA impairs females' fertility by decreasing the number of ovarian follicles. For the first time, we uncovered the novel mPACT pattern of a mitochondrial mRNA and demonstrated the physiological function of this 14th protein encoded by mtDNA.


Subject(s)
Cytochromes b , Animals , Cytochromes b/genetics , Cytochromes b/metabolism , Mice , Female , Mice, Transgenic , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Humans , Mice, Inbred C57BL , Genes, Mitochondrial , RNA, Messenger/metabolism , RNA, Messenger/genetics , Male
19.
Int Immunopharmacol ; 134: 112185, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38701540

ABSTRACT

Chronic ethanol consumption is a prevalent condition in contemporary society and exacerbates anxiety symptoms in healthy individuals. The activation of microglia, leading to neuroinflammatory responses, may serve as a significant precipitating factor; however, the precise molecular mechanisms underlying this phenomenon remain elusive. In this study, we initially confirmed that chronic ethanol exposure (CEE) induces anxiety-like behaviors in mice through open field test and elevated plus maze test. The cGAS/STING signaling pathway has been confirmed to exhibits a significant association with inflammatory signaling responses in both peripheral and central systems. Western blot analysis confirmed alterations in the cGAS/STING signaling pathway during CEE, including the upregulation of p-TBK1 and p-IRF3 proteins. Moreover, we observed microglial activation in the prefrontal cortex (PFC) of CEE mice, characterized by significant alterations in branching morphology and an increase in cell body size. Additionally, we observed that administration of CEE resulted in mitochondrial dysfunction within the PFC of mice, accompanied by a significant elevation in cytosolic mitochondrial DNA (mtDNA) levels. Furthermore, our findings revealed that the inhibition of STING by H-151 effectively alleviated anxiety-like behavior and suppressed microglial activation induced by CEE. Our study unveiled a significant association between anxiety-like behavior, microglial activation, inflammation, and mitochondria dysfunction during CEE.


Subject(s)
Anxiety , Ethanol , Membrane Proteins , Mice, Inbred C57BL , Microglia , Nucleotidyltransferases , Prefrontal Cortex , Signal Transduction , Animals , Microglia/drug effects , Microglia/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Anxiety/chemically induced , Membrane Proteins/metabolism , Membrane Proteins/genetics , Ethanol/toxicity , Signal Transduction/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Male , Mice , Behavior, Animal/drug effects , DNA, Mitochondrial/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Disease Models, Animal , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Protein Serine-Threonine Kinases
20.
Free Radic Biol Med ; 220: 312-323, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38740101

ABSTRACT

Podocytes are crucial for regulating glomerular permeability. They have foot processes that are integral to the renal filtration barrier. Understanding their energy metabolism could shed light on the pathogenesis of filtration barrier injury. Lactate has been increasingly recognized as more than a waste product and has emerged as a significant metabolic fuel and reserve. The recent identification of lactate transporters in podocytes, the expression of which is modulated by glucose levels and lactate, highlights lactate's relevance. The present study investigated the impact of lactate on podocyte respiratory efficiency and mitochondrial dynamics. We confirmed lactate oxidation in podocytes, suggesting its role in cellular energy production. Under conditions of glucose deprivation or lactate supplementation, a significant shift was seen toward oxidative phosphorylation, reflected by an increase in the oxygen consumption rate/extracellular acidification rate ratio. Notably, lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB) isoforms, which are involved in lactate conversion to pyruvate, were detected in podocytes for the first time. The presence of lactate led to higher intracellular pyruvate levels, greater LDH activity, and higher LDHB expression. Furthermore, lactate exposure increased mitochondrial DNA-to-nuclear DNA ratios and resulted in upregulation of the mitochondrial biogenesis markers peroxisome proliferator-activated receptor coactivator-1α and transcription factor A mitochondrial, regardless of glucose availability. Changes in mitochondrial size and shape were observed in lactate-exposed podocytes. These findings suggest that lactate is a pivotal energy source for podocytes, especially during energy fluctuations. Understanding lactate's role in podocyte metabolism could offer insights into renal function and pathologies that involve podocyte injury.


Subject(s)
L-Lactate Dehydrogenase , Lactic Acid , Mitochondrial Dynamics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Podocytes , Podocytes/metabolism , Podocytes/pathology , Animals , Rats , Lactic Acid/metabolism , L-Lactate Dehydrogenase/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Mitochondria/metabolism , Mitochondria/pathology , Glucose/metabolism , Energy Metabolism , Lactate Dehydrogenase 5/metabolism , Oxidative Phosphorylation/drug effects , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Oxygen Consumption , Cells, Cultured , Pyruvic Acid/metabolism , Isoenzymes
SELECTION OF CITATIONS
SEARCH DETAIL
...