Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.622
Filter
1.
Orphanet J Rare Dis ; 19(1): 287, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095827

ABSTRACT

BACKGROUND: This study aimed to explore the demographic characteristics, diagnostic challenges, treatment patterns, and caregiver burden of mitochondrial diseases. METHODS: This retrospective cross-sectional study enrolled patients diagnosed with mitochondrial diseases from the Department of Neurology at Peking University First Hospital between January 2010 and December 2021. A questionnaire covering demographic characteristics, diagnostic dilemma, treatment, economic aspects, and caregiver stress was administered, and disability was assessed using the modified Rankin Scale (mRS). RESULTS: A total of 183 patients (mean age: 16 (IQR: 12-25), 49.72% males) were enrolled, including 124 pediatric patients and 59 adult patients. MELAS (106. 57.92%) and Leigh syndrome (37, 20.22%) were predominant among the mitochondrial disease subtypes. Among them, 132 (72.13%) patients were initially misdiagnosed with other diseases, 58 (31.69%) patients visited 2 hospitals before confirmed as mitochondrial disease, and 39 (21.31%) patients visited 3 hospitals before confirmed as mitochondrial disease. Metabolic modifiers were the most common type of drugs used, including several dietary supplements such as L-carnitine (117, 63.93%), Coenzyme Q10 (102, 55.74%), idebenone (82, 44.81%), and vitamins (99, 54.10%) for proper mitochondrial function. Mothers are the primary caregivers for both children (36.29%) and adults (38.98%). The mRS score ranged from 0 to 5, 92.35% of the patients had different degrees of disability due to mitochondrial disease. The average monthly treatment cost was 3000 RMB for children and 3100 RMB for adults. CONCLUSIONS: This study provided valuable insights into the characteristics and challenges of mitochondrial diseases, which underscores the need for improved awareness, diagnostic efficiency, and comprehensive support for patients and caregivers.


Subject(s)
Mitochondrial Diseases , Humans , Cross-Sectional Studies , Retrospective Studies , Mitochondrial Diseases/diagnosis , Male , Female , Adult , Child , Adolescent , Young Adult , Caregivers , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use , Caregiver Burden , Surveys and Questionnaires
3.
J Nanobiotechnology ; 22(1): 487, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143493

ABSTRACT

Mitochondria are crucial organelles responsible for energy generation in eukaryotic cells. Oxidative stress, calcium disorders, and mitochondrial DNA abnormalities can all cause mitochondrial dysfunction. It is now well documented that mitochondrial dysfunction significantly contributes to the pathogenesis of numerous illnesses. Hence, it is vital to investigate innovative treatment methods targeting mitochondrial dysfunction. Extracellular vesicles (EVs) are cell-derived nanovesicles that serve as intercellular messengers and are classified into small EVs (sEVs, < 200 nm) and large EVs (lEVs, > 200 nm) based on their sizes. It is worth noting that certain subtypes of EVs are rich in mitochondrial components (even structurally intact mitochondria) and possess the ability to transfer them or other contents including proteins and nucleic acids to recipient cells to modulate their mitochondrial function. Specifically, EVs can modulate target cell mitochondrial homeostasis as well as mitochondria-controlled apoptosis and ROS generation by delivering relevant substances. In addition, the artificial modification of EVs as delivery carriers for therapeutic goods targeting mitochondria is also a current research hotspot. In this article, we will focus on the ability of EVs to modulate the mitochondrial function of target cells, aiming to offer novel perspectives on therapeutic approaches for diverse conditions linked to mitochondrial dysfunction.


Subject(s)
Extracellular Vesicles , Mitochondria , Extracellular Vesicles/metabolism , Humans , Mitochondria/metabolism , Animals , Reactive Oxygen Species/metabolism , Oxidative Stress , Apoptosis , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/therapy , Drug Delivery Systems/methods
4.
Biomed Pharmacother ; 178: 117084, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39088967

ABSTRACT

The accumulation of excess reactive oxygen species (ROS) can lead to oxidative stress (OS), which can induce gene mutations, protein denaturation, and lipid peroxidation directly or indirectly. The expression is reduced ATP level in cells, increased cytoplasmic Ca2+, inflammation, and so on. Consequently, ROS are recognized as significant risk factors for human aging and various diseases, including diabetes, cardiovascular diseases, and neurodegenerative diseases. Mitochondria are involved in the production of ROS through the respiratory chain. Abnormal mitochondrial characteristics, including mitochondrial OS, mitochondrial fission, mitochondrial fusion, and mitophagy, play an important role in various tissues. However, previous excellent reviews focused on OS-induced diseases. In this review, we focus on the latest progress of OS-induced mitochondrial dynamics, discuss OS-induced mitochondrial damage-related diseases, and summarize the OS-induced mitochondrial dynamics-related signaling pathways. Additionally, it elaborates on potential therapeutic methods aimed at preventing oxidative stress from further exacerbating mitochondrial disorders.


Subject(s)
Mitochondria , Mitochondrial Dynamics , Oxidative Stress , Reactive Oxygen Species , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Mitochondria/metabolism , Signal Transduction , Mitochondrial Diseases/metabolism , Mitophagy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
6.
J Neurogenet ; 38(2): 27-34, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38975939

ABSTRACT

Tropical ataxic neuropathy (TAN) is characterised by ataxic polyneuropathy, degeneration of the posterior columns and pyramidal tracts, optic atrophy, and sensorineural hearing loss. It has been attributed to nutritional/toxic etiologies, but evidence for the same has been equivocal. TAN shares common clinical features with inherited neuropathies and mitochondrial disorders, it may be hypothesised that genetic abnormalities may underlie the pathophysiology of TAN. This study aimed to establish evidence for mitochondrial dysfunction by adopting an integrated biochemical and multipronged genetic analysis. Patients (n = 65) with chronic progressive ataxic neuropathy with involvement of visual and/or auditory pathways underwent deep phenotyping, genetic studies including mitochondrial DNA (mtDNA) deletion analysis, mtDNA and clinical exome sequencing (CES), and respiratory chain complex (RCC) assay. The phenotypic characteristics included dysfunction of visual (n = 14), auditory (n = 12) and visual + auditory pathways (n = 29). Reduced RCC activity was present in 13 patients. Mitochondrial DNA deletions were noted in five patients. Sequencing of mtDNA (n = 45) identified a homoplasmic variant (MT-ND6) and a heteroplasmic variant (MT-COI) in one patient each. CES (n = 45) revealed 55 variants in nuclear genes that are associated with neuropathy (n = 27), deafness (n = 7), ataxia (n = 4), and mitochondrial phenotypes (n = 5) in 36 patients. This study provides preliminary evidence that TAN is associated with a spectrum of genetic abnormalities, including those associated with mitochondrial dysfunction, which is in contradistinction from the prevailing hypothesis that TAN is related to dietary toxins. Analysing the functional relevance of these genetic variants may improve the understanding of the pathogenesis of TAN.


Subject(s)
Ataxia , DNA, Mitochondrial , Humans , Male , Female , DNA, Mitochondrial/genetics , Adult , Middle Aged , Ataxia/genetics , Adolescent , Mitochondrial Diseases/genetics , Young Adult , Mitochondria/genetics , Child , Aged , Exome Sequencing , Phenotype
7.
Future Cardiol ; 20(4): 179-182, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-39049772

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a well-known manifestation of inherited mitochondrial disease. Still, currently available gene panels do not include mitochondrial genome sequencing. Mitochondrial dysfunction plays a very important role in the pathogenesis of HCM, whether tested positive or negative by the currently available gene panels for HCM. Mitochondrial DNA variations may act as modifiers of disease manifestation in genotype-positive individuals. In genotype-negative individuals, it may be the primary driver of pathogenesis. A recent study has demonstrated that mitochondrial dysfunction is correlated with septal hypertrophy in genotype-negative HCM, which can be amenable to mitochondria-targeted therapy. It is important to consider mitochondrial genome sequencing as part of the genetic evaluation of HCM.


Hypertrophic cardiomyopathy or 'thick heart' is a common heart problem that can lead to abnormal heart rhythm and even heart failure. In older adults, it is often due to high blood pressure that causes the heart to pump against high resistance and hence becoming thick. However, it can occur without high blood pressure, often in young individuals with underlying heart muscle disease. Sometimes, there are many individuals in a family with thick hearts. In these instances, it is likely genetic. The individual may have a faulty gene related to heart muscle function causing the heart to become thick as an adaptation to inefficient heart muscle function. Mitochondria are tiny organelles inside our cells that make energy. When there is mitochondrial damage, heart muscles cannot generate energy efficiently. This can lead to a thick heart as well. Hence, it is important to test mitochondrial genes along with the heart muscle genes to find the cause of thick heart when it is unexplained, or a genetic cause is suspected.


Subject(s)
Cardiomyopathy, Hypertrophic , DNA, Mitochondrial , Humans , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/diagnosis , DNA, Mitochondrial/genetics , Mitochondria , Mitochondrial Diseases/genetics , Mitochondrial Diseases/diagnosis , Mutation
8.
Genes (Basel) ; 15(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39062673

ABSTRACT

Aminoacyl-tRNA synthetases are essential enzymes for the accurate translation of genetic information. IARS1 and IARS2 are isoleucyl-tRNA synthetases functioning in the cytoplasm and mitochondria, respectively, with genetic mutations in these enzymes causing diverse clinical phenotypes in specific organs and tissues. Mutations in IARS1 and IARS2 have recently been linked to mitochondrial diseases. This review aims to explore the relationship between IARS1 and IARS2 and these diseases, providing a comprehensive overview of their association with mitochondrial diseases. Mutations in IARS1 cause weak calf syndrome in cattle and mitochondrial diseases in humans, leading to growth retardation and liver dysfunction. Mutations in IARS2 are associated with Leigh syndrome, craniosynostosis and abnormal genitalia syndrome. Future research is expected to involve genetic analysis of a larger number of patients, identifying new mutations in IARS1 and IARS2, and elucidating their impact on mitochondrial function. Additionally, genetically modified mice and the corresponding phenotypic analysis will serve as powerful tools for understanding the functions of these gene products and unraveling disease mechanisms. This will likely promote the development of new therapies and preventive measures.


Subject(s)
Isoleucine-tRNA Ligase , Mitochondrial Diseases , Mutation , Humans , Animals , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Isoleucine-tRNA Ligase/genetics , Cattle , Mitochondria/genetics , Mice
9.
Drug Discov Today ; 29(8): 104086, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960132

ABSTRACT

Circulating cell-free mitochondrial DNA (ccf-mtDNA) has emerged as a promising biomarker, with potential implications for disease diagnosis. Changes in mtDNA, such as deletions, mutations or variations in the number of copies, have been associated with mitochondrial disorders, heart diseases, cancer and age-related non-communicable diseases. Previous methods, such as polymerase chain reaction-based approaches, next-generation sequencing and imaging-based techniques, have shown improved accuracy in identifying rare mtDNA variants or mutations, but they have limitations. This article explains the basic principles and benefits of using planar optical waveguide-based detection devices, which represent an advanced approach in the field of sensing.


Subject(s)
Cell-Free Nucleic Acids , DNA, Mitochondrial , Precision Medicine , Humans , DNA, Mitochondrial/genetics , Precision Medicine/methods , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , Nanotechnology/methods , Mutation , Animals , Mitochondrial Diseases/genetics , Mitochondrial Diseases/diagnosis
10.
Anal Chem ; 96(29): 11915-11922, 2024 07 23.
Article in English | MEDLINE | ID: mdl-39007441

ABSTRACT

G-quadruplex structures within the nuclear genome (nG4) is an important regulatory factor, while the function of G4 in the mitochondrial genome (mtG4) still needs to be explored, especially in human sperms. To gain a better understanding of the relationship between mtG4 and mitochondrial function, it is crucial to develop excellent probes that can selectively visualize and track mtG4 in both somatic cells and sperms. Herein, based on our previous research on purine frameworks, we attempted for the first time to extend the conjugated structure from the C-8 site of purine skeleton and discovered that the purine derivative modified by the C-8 aldehyde group is an ideal platform for constructing near-infrared probes with extremely large Stokes shift (>220 nm). Compared with the compound substituted with methylpyridine (PAP), the molecule substituted with methylthiazole orange (PATO) showed better G4 recognition ability, including longer emission (∼720 nm), more significant fluorescent enhancement (∼67-fold), lower background, and excellent photostability. PATO exhibited a sensitive response to mtG4 variation in both somatic cells and human sperms. Most importantly, PATO helped us to discover that mtG4 was significantly increased in cells with mitochondrial respiratory chain damage caused by complex I inhibitors (6-OHDA and rotenone), as well as in human sperms that suffer from oxidative stress. Altogether, our study not only provides a novel ideal molecular platform for constructing high-performance probes but also develops an effective tool for studying the relationship between mtG4 and mitochondrial function in both somatic cells and human sperms.


Subject(s)
Fluorescent Dyes , Purines , Humans , Purines/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Mitochondrial Diseases/metabolism , Up-Regulation , Genome, Mitochondrial , G-Quadruplexes , Mitochondria/metabolism , Infrared Rays , HeLa Cells
11.
Physiol Res ; 73(Suppl 1): S243-S278, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39016153

ABSTRACT

Disorders of ATP synthase, the key enzyme in mitochondrial energy supply, belong to the most severe metabolic diseases, manifesting as early-onset mitochondrial encephalo-cardiomyopathies. Since ATP synthase subunits are encoded by both mitochondrial and nuclear DNA, pathogenic variants can be found in either genome. In addition, the biogenesis of ATP synthase requires several assembly factors, some of which are also hotspots for pathogenic variants. While variants of MT-ATP6 and TMEM70 represent the most common cases of mitochondrial and nuclear DNA mutations respectively, the advent of next-generation sequencing has revealed new pathogenic variants in a number of structural genes and TMEM70, sometimes with truly peculiar genetics. Here we present a systematic review of the reported cases and discuss biochemical mechanisms, through which they are affecting ATP synthase. We explore how the knowledge of pathophysiology can improve our understanding of enzyme biogenesis and function. Keywords: Mitochondrial diseases o ATP synthase o Nuclear DNA o Mitochondrial DNA o TMEM70.


Subject(s)
Mitochondrial Proton-Translocating ATPases , Phenotype , Humans , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Mutation , Mitochondrial Diseases/genetics , Mitochondrial Diseases/enzymology , DNA, Mitochondrial/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Animals , Mitochondria/enzymology , Mitochondria/genetics
12.
Nat Metab ; 6(8): 1479-1491, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048800

ABSTRACT

Primary mitochondrial diseases (PMDs) are associated with pediatric neurological disorders and are traditionally related to oxidative phosphorylation system (OXPHOS) defects in neurons. Interestingly, both PMD mouse models and patients with PMD show gliosis, and pharmacological depletion of microglia, the innate immune cells of the brain, ameliorates multiple symptoms in a mouse model. Given that microglia activation correlates with the expression of OXPHOS genes, we studied whether OXPHOS deficits in microglia may contribute to PMDs. We first observed that the metabolic rewiring associated with microglia stimulation in vitro (via IL-33 or TAU treatment) was partially changed by complex I (CI) inhibition (via rotenone treatment). In vivo, we generated a mouse model deficient for CI activity in microglia (MGcCI). MGcCI microglia showed metabolic rewiring and gradual transcriptional activation, which led to hypertrophy and dysfunction in juvenile (1-month-old) and adult (3-month-old) stages, respectively. MGcCI mice presented widespread reactive astrocytes, a decrease of synaptic markers accompanied by an increased number of parvalbumin neurons, a behavioral deficit characterized by prolonged periods of immobility, loss of weight and premature death that was partially rescued by pharmacologic depletion of microglia. Our data demonstrate that microglia development depends on mitochondrial CI and suggest a direct microglial contribution to PMDs.


Subject(s)
Electron Transport Complex I , Microglia , Mitochondrial Diseases , Animals , Microglia/metabolism , Mice , Electron Transport Complex I/metabolism , Electron Transport Complex I/deficiency , Electron Transport Complex I/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/etiology , Oxidative Phosphorylation , Mitochondria/metabolism , Neurons/metabolism , Neuroglia/metabolism , Disease Models, Animal , Astrocytes/metabolism , Gliosis/metabolism , Gliosis/pathology , Brain/metabolism , Brain/pathology
13.
Ann Afr Med ; 23(3): 512-513, 2024 Jul 01.
Article in French, English | MEDLINE | ID: mdl-39034583

ABSTRACT

Hyperkinesias in a patient with complex-I deficiency due to the variant m.10191T>C in MT-ND3 have not been previously reported. The patient is a 32 years-old female with multisystem mitochondrial disease due to variant m.10191T>C in MT-ND3, who has been experiencing episodic, spontaneous or induced abnormal movements since age 23. The abnormal movements started as right hemi-athetosis, bilateral dystonia of the legs, or unilateral dystonia of the right arm and leg. They often progressed to severe ballism, involving the trunk, and limbs. The arms were more dystonic than the legs. In conclusion, complex-I deficiency due to the variant m.10191T>C in MT-ND3 may manifest as multisystem disease including hyperkinesias. Neurologists should be aware of hyperkinesias as a manifestation of complex-I deficiency.


RésuméL'hyperkinésie d'une patiente atteinte d'un déficit en complexe I dû à la variante m.10191T>C du gène MT-ND3 n'a jamais été rapportée auparavant. La patiente est une femme de 32 ans atteinte d'une maladie mitochondriale multisystémique due à la variante m.10191T>C du gène MT-ND3, qui présente des mouvements anormaux épisodiques, spontanés ou provoqués depuis l'âge de 18 ans. mouvements anormaux épisodiques, spontanés ou provoqués depuis l'âge de 23 ans. Les mouvements anormaux ont commencé par une hémiathétose droite, dystonie bilatérale des jambes ou dystonie unilatérale du bras et de la jambe droite. Ils ont souvent évolué vers un ballisme sévère, impliquant le tronc et les membres. le tronc et les membres. Les bras étaient plus dystoniques que les jambes. En conclusion, le déficit en complexe I dû à la variante m.10191T>C du gène MT-ND3 peut se manifester par une maladie multisystémique comprenant des hyperkinésies. Les neurologues doivent être conscients que l'hyperkinésie est une manifestation du déficit en complexe-I. de la déficience en complexe I.


Subject(s)
Leigh Disease , Humans , Female , Adult , Leigh Disease/complications , Leigh Disease/diagnosis , Hyperkinesis/etiology , Electron Transport Complex I/deficiency , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mutation
14.
J Diabetes Complications ; 38(8): 108798, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991492

ABSTRACT

AIMS: Type 1 diabetes has been associated with mitochondrial dysfunction. However, the mechanism of this dysfunction in adults remains unclear. METHODS: A secondary analysis was conducted using data from several clinical trials measuring in-vivo and ex-vivo mitochondrial function in adults with type 1 diabetes (n = 34, age 38.8 ± 14.6 years) and similarly aged controls (n = 59, age 44.6 ± 13.9 years). In-vivo mitochondrial function was assessed before, during, and after isometric exercise with 31phosphorous magnetic resonance spectroscopy. High resolution respirometry of vastus lateralis muscle tissue was used to assess ex-vivo measures. RESULTS: In-vivo data showed higher rates of anaerobic glycolysis (p = 0.013), and a lower maximal mitochondrial oxidative capacity (p = 0.012) and mitochondrial efficiency (p = 0.024) in adults with type 1 diabetes. After adjustment for age and percent body fat maximal mitochondrial capacity (p = 0.014) continued to be lower and anaerobic glycolysis higher (p = 0.040) in adults with type 1 diabetes. Ex-vivo data did not demonstrate significant differences between the two groups. CONCLUSIONS: The in-vivo analysis demonstrates that adults with type 1 diabetes have mitochondrial dysfunction. This builds on previous research showing in-vivo mitochondrial dysfunction in youths with type 1 diabetes and suggests that defects in substrate or oxygen delivery may play a role in in-vivo dysfunction.


Subject(s)
Diabetes Mellitus, Type 1 , Mitochondria, Muscle , Humans , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Adult , Male , Female , Middle Aged , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Glycolysis/physiology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Mitochondrial Diseases/complications , Case-Control Studies , Magnetic Resonance Spectroscopy , Young Adult , Exercise/physiology
15.
Sci Rep ; 14(1): 13789, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877095

ABSTRACT

Mitochondrial function is critical to continued cellular vitality and is an important contributor to a growing number of human diseases. Mitochondrial dysfunction is typically heterogeneous, mediated through the clonal expansion of mitochondrial DNA (mtDNA) variants in a subset of cells in a given tissue. To date, our understanding of the dynamics of clonal expansion of mtDNA variants has been technically limited to the single cell-level. Here, we report the use of nanobiopsy for subcellular sampling from human tissues, combined with next-generation sequencing to assess subcellular mtDNA mutation load in human tissue from mitochondrial disease patients. The ability to map mitochondrial mutation loads within individual cells of diseased tissue samples will further our understanding of mitochondrial genetic diseases.


Subject(s)
DNA, Mitochondrial , Heteroplasmy , High-Throughput Nucleotide Sequencing , Mutation , Humans , DNA, Mitochondrial/genetics , Heteroplasmy/genetics , High-Throughput Nucleotide Sequencing/methods , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Diseases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
18.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928331

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder currently affecting the ageing population. Although the aetiology of PD has yet to be fully elucidated, environmental factors such as exposure to the naturally occurring neurotoxin rotenone has been associated with an increased risk of developing PD. Rotenone inhibits mitochondrial respiratory chain (MRC) complex I activity as well as induces dopaminergic neuronal death. The aim of the present study was to investigate the underlying mechanisms of rotenone-induced mitochondrial dysfunction and oxidative stress in an in vitro SH-SY5Y neuronal cell model of PD and to assess the ability of pre-treatment with Coenzyme Q10 (CoQ10) to ameliorate oxidative stress in this model. Spectrophotometric determination of the mitochondrial enzyme activities and fluorescence probe studies of reactive oxygen species (ROS) production was assessed. Significant inhibition of MRC complex I and II-III activities was observed, together with a significant loss of neuronal viability, CoQ10 status, and ATP synthesis. Additionally, significant increases were observed in intracellular and mitochondrial ROS production. Remarkably, CoQ10 supplementation was found to reduce ROS formation. These results have indicated mitochondrial dysfunction and increased oxidative stress in a rotenone-induced neuronal cell model of PD that was ameliorated by CoQ10 supplementation.


Subject(s)
Mitochondria , Neurons , Oxidative Stress , Rotenone , Ubiquinone , Humans , Ataxia , Cell Line, Tumor , Cell Survival/drug effects , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Diseases , Muscle Weakness/metabolism , Muscle Weakness/chemically induced , Muscle Weakness/pathology , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/etiology , Reactive Oxygen Species/metabolism , Rotenone/toxicity , Rotenone/adverse effects , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/deficiency
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL