Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.280
Filter
2.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732047

ABSTRACT

Mitochondrial dysfunction plays a major role in physiological aging and in many pathological conditions. Yet, no study has explored the consequence of primary mitochondrial deficiency on the blood-brain barrier (BBB) structure and function. Addressing this question has major implications for pharmacological and genetic strategies aimed at ameliorating the neurological symptoms that are often predominant in patients suffering from these conditions. In this study, we examined the permeability of the BBB in the Ndufs4-/- mouse model of Leigh syndrome (LS). Our results indicated that the structural and functional integrity of the BBB was preserved in this severe model of mitochondrial disease. Our findings suggests that pharmacological or gene therapy strategies targeting the central nervous system in this mouse model and possibly other models of mitochondrial dysfunction require the use of specific tools to bypass the BBB. In addition, they raise the need for testing the integrity of the BBB in complementary in vivo models.


Subject(s)
Blood-Brain Barrier , Disease Models, Animal , Electron Transport Complex I , Leigh Disease , Animals , Mice , Blood-Brain Barrier/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/deficiency , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Mice, Knockout , Mitochondria/metabolism , Mitochondria/genetics
3.
J Hazard Mater ; 472: 134598, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38743975

ABSTRACT

N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) is an emerging pollutant transformed from 6-PPD. However, the effect of 6-PPDQ exposure on mitochondrion and underlying mechanism remains largely unclear. Using Caenorhabditis elegans as animal model, exposed to 6-PPDQ at 0.1-10 µg/L was performed form L1 larvae to adult day-1. Exposure to 6-PPDQ (1 and 10 µg/L) could increase oxygen consumption rate and decease adenosine 5'-triphosphate (ATP) content, suggesting induction of mitochondrial dysfunction. Activities of NADH dehydrogenase (complex I) and succinate dehydrogenase (complex II) were inhibited, accompanied by a decrease in expressions of gas-1, nuo-1, and mev-1. RNAi of gas-1 and mev-1 enhanced mitochondrial dysfunction and reduced lifespan of 6-PPDQ exposed nematodes. GAS-1 and MEV-1 functioned in parallel to regulate 6-PPDQ toxicity to reduce the lifespan. Insulin peptides and the insulin signaling pathway acted downstream of GAS-1 and MEV-1 to control the 6-PPDQ toxicity on longevity. Moreover, RNAi of sod-2 and sod-3, targeted genes of daf-16, caused susceptibility to 6-PPDQ toxicity in reducing lifespan and in causing reactive oxygen species (ROS) production. Therefore, 6-PPDQ at environmentally relevant concentrations (ERCs) potentially caused mitochondrial dysfunction by affecting mitochondrial complexes I and II, which was associated with lifespan reduction by affecting insulin signaling in organisms.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Electron Transport Complex I , Longevity , Mitochondria , Animals , Caenorhabditis elegans/drug effects , Longevity/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex II/metabolism , Electron Transport Complex II/genetics , Insulin/metabolism , Adenosine Triphosphate/metabolism , Reactive Oxygen Species/metabolism , NADH Dehydrogenase , Cytochromes b
4.
Proc Natl Acad Sci U S A ; 121(22): e2405123121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781208

ABSTRACT

Mitochondria play a central role in muscle metabolism and function. A unique family of iron-sulfur proteins, termed CDGSH Iron Sulfur Domain-containing (CISD/NEET) proteins, support mitochondrial function in skeletal muscles. The abundance of these proteins declines during aging leading to muscle degeneration. Although the function of the outer mitochondrial CISD/NEET proteins, CISD1/mitoNEET and CISD2/NAF-1, has been defined in skeletal muscle cells, the role of the inner mitochondrial CISD protein, CISD3/MiNT, is currently unknown. Here, we show that CISD3 deficiency in mice results in muscle atrophy that shares proteomic features with Duchenne muscular dystrophy. We further reveal that CISD3 deficiency impairs the function and structure of skeletal muscles, as well as their mitochondria, and that CISD3 interacts with, and donates its [2Fe-2S] clusters to, complex I respiratory chain subunit NADH Ubiquinone Oxidoreductase Core Subunit V2 (NDUFV2). Using coevolutionary and structural computational tools, we model a CISD3-NDUFV2 complex with proximal coevolving residue interactions conducive of [2Fe-2S] cluster transfer reactions, placing the clusters of the two proteins 10 to 16 Å apart. Taken together, our findings reveal that CISD3/MiNT is important for supporting the biogenesis and function of complex I, essential for muscle maintenance and function. Interventions that target CISD3 could therefore impact different muscle degeneration syndromes, aging, and related conditions.


Subject(s)
Electron Transport Complex I , Mitochondrial Proteins , Muscle, Skeletal , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondria/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Mice, Knockout , Mitochondria, Muscle/metabolism , Humans , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/genetics
5.
Mol Cell ; 84(10): 1964-1979.e6, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759628

ABSTRACT

The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.


Subject(s)
AMP-Activated Protein Kinases , Electron Transport Complex I , Ferroptosis , Phospholipid Hydroperoxide Glutathione Peroxidase , Protein Serine-Threonine Kinases , Ferroptosis/genetics , Ferroptosis/drug effects , Animals , Humans , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Cell Line, Tumor , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy , AMP-Activated Protein Kinase Kinases/genetics , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/drug effects , Xenograft Model Antitumor Assays , Signal Transduction , Female
6.
Nat Commun ; 15(1): 3631, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684731

ABSTRACT

Idiopathic Parkinson's disease (iPD) is believed to have a heterogeneous pathophysiology, but molecular disease subtypes have not been identified. Here, we show that iPD can be stratified according to the severity of neuronal respiratory complex I (CI) deficiency, and identify two emerging disease subtypes with distinct molecular and clinical profiles. The CI deficient (CI-PD) subtype accounts for approximately a fourth of all cases, and is characterized by anatomically widespread neuronal CI deficiency, a distinct cell type-specific gene expression profile, increased load of neuronal mtDNA deletions, and a predilection for non-tremor dominant motor phenotypes. In contrast, the non-CI deficient (nCI-PD) subtype exhibits no evidence of mitochondrial impairment outside the dopaminergic substantia nigra and has a predilection for a tremor dominant phenotype. These findings constitute a step towards resolving the biological heterogeneity of iPD with implications for both mechanistic understanding and treatment strategies.


Subject(s)
DNA, Mitochondrial , Electron Transport Complex I , Electron Transport Complex I/deficiency , Mitochondria , Mitochondrial Diseases , Parkinson Disease , Parkinson Disease/genetics , Parkinson Disease/metabolism , Humans , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Male , DNA, Mitochondrial/genetics , Female , Mitochondria/metabolism , Mitochondria/genetics , Aged , Substantia Nigra/metabolism , Substantia Nigra/pathology , Middle Aged , Phenotype , Neurons/metabolism
7.
Rinsho Shinkeigaku ; 64(5): 326-332, 2024 May 24.
Article in Japanese | MEDLINE | ID: mdl-38644210

ABSTRACT

Leber's hereditary optic atrophy (LHON) is a genetic optic neuropathy that is more prevalent in young males but can occur from childhood to old age. The primary cause is mitochondrial genetic mutations, which are associated with dysfunction of mitochondrial electron transport chain complex I. It manifests as acute to subacute visual impairment, often starting unilaterally but progressing to involve both eyes within weeks to months. Visual loss is severe, with many patients having corrected visual acuity below 0.1. The differential diagnosis of optic neuritis is essential, and assessments such as pupillary light reflex, fluorescein fundus angiography, and magnetic resonance imaging can be useful for differentiation. LHON should be considered as one of the differential diagnoses for optic neuritis, and collaboration between neurologists and ophthalmologists is crucial for accurate diagnosis and appropriate treatment.


Subject(s)
Magnetic Resonance Imaging , Optic Atrophy, Hereditary, Leber , Optic Atrophy, Hereditary, Leber/diagnosis , Optic Atrophy, Hereditary, Leber/genetics , Humans , Diagnosis, Differential , Male , Mutation , Optic Neuritis/diagnosis , Optic Neuritis/etiology , Optic Neuritis/diagnostic imaging , Fluorescein Angiography , Female , Electron Transport Complex I/genetics , Adult , Mitochondria/genetics , Child
8.
Int J Biochem Cell Biol ; 171: 106583, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657899

ABSTRACT

Protein crotonylation plays a role in regulating cellular metabolism, gene expression, and other biological processes. NDUFA9 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9) is closely associated with the activity and function of mitochondrial respiratory chain complex I. Mitochondrial function and respiratory chain are closely related to browning of white adipocytes, it's speculated that NDUFA9 and its crotonylation are associated with browning of white adipocytes. Firstly, the effect of NDUFA9 on white adipose tissue was verified in white fat browning model mice, and it was found that NDUFA9 promoted mitochondrial respiration, thermogenesis, and browning of white adipose tissue. Secondly, in cellular studies, it was discovered that NDUFA9 facilitated browning of white adipocytes by enhancing mitochondrial function, mitochondrial complex I activity, ATP synthesis, and mitochondrial respiration. Again, the level of NDUFA9 crotonylation was increased by treating cells with vorinostat (SAHA)+sodium crotonate (NaCr) and overexpressing NDUFA9, it was found that NDUFA9 crotonylation promoted browning of white adipocytes. Meanwhile, the acetylation level of NDUFA9 was increased by treating cells with SAHA+sodium acetate (NaAc) and overexpressing NDUFA9, the assay revealed that NDUFA9 acetylation inhibited white adipocytes browning. Finally, combined with the competitive relationship between acetylation and crotonylation, it was also demonstrated that NDUFA9 crotonylation promoted browning of white adipocytes. Above results indicate that NDUFA9 and its crotonylation modification promote mitochondrial function, which in turn promotes browning of white adipocytes. This study establishes a theoretical foundation for the management and intervention of obesity, which is crucial in addressing obesity and related medical conditions in the future.


Subject(s)
Adipocytes, White , Mitochondria , Animals , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Adipocytes, White/metabolism , Adipocytes, White/drug effects , Adipocytes, White/cytology , Male , Mice, Inbred C57BL , Thermogenesis/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , 3T3-L1 Cells , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Acetylation/drug effects
9.
Neuropathol Appl Neurobiol ; 50(3): e12977, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38680020

ABSTRACT

AIM: Leigh syndrome (LS), the most common paediatric presentation of genetic mitochondrial dysfunction, is a multi-system disorder characterised by severe neurologic and metabolic abnormalities. Symmetric, bilateral, progressive necrotizing lesions in the brainstem are defining features of the disease. Patients are often symptom free in early life but typically develop symptoms by about 2 years of age. The mechanisms underlying disease onset and progression in LS remain obscure. Recent studies have shown that the immune system causally drives disease in the Ndufs4(-/-) mouse model of LS: treatment of Ndufs4(-/-) mice with the macrophage-depleting Csf1r inhibitor pexidartinib prevents disease. While the precise mechanisms leading to immune activation and immune factors involved in disease progression have not yet been determined, interferon-gamma (IFNγ) and interferon gamma-induced protein 10 (IP10) were found to be significantly elevated in Ndufs4(-/-) brainstem, implicating these factors in disease. Here, we aimed to explore the role of IFNγ and IP10 in LS. METHODS: To establish the role of IFNγ and IP10 in LS, we generated IFNγ and IP10 deficient Ndufs4(-/-)/Ifng(-/-) and Ndufs4(-/-)/IP10(-/-) double knockout animals, as well as IFNγ and IP10 heterozygous, Ndufs4(-/-)/Ifng(+/-) and Ndufs4(-/-)/IP10(+/-), animals. We monitored disease onset and progression to define the impact of heterozygous or homozygous loss of IFNγ and IP10 in LS. RESULTS: Loss of IP10 does not significantly impact the onset or progression of disease in the Ndufs4(-/-) model. IFNγ loss significantly extends survival and delays disease progression in a gene dosage-dependent manner, though the benefits are modest compared to Csf1r inhibition. CONCLUSIONS: IFNγ contributes to disease onset and progression in LS. Our findings suggest that IFNγ targeting therapies may provide some benefits in genetic mitochondrial disease, but targeting IFNγ alone would likely yield only modest benefits in LS.


Subject(s)
Disease Progression , Electron Transport Complex I , Interferon-gamma , Leigh Disease , Animals , Mice , Brain Stem/pathology , Brain Stem/metabolism , Disease Models, Animal , Electron Transport Complex I/genetics , Electron Transport Complex I/deficiency , Interferon-gamma/metabolism , Leigh Disease/pathology , Leigh Disease/genetics , Mice, Inbred C57BL , Mice, Knockout
10.
Biochim Biophys Acta Bioenerg ; 1865(3): 149046, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38642871

ABSTRACT

The respiratory chain alternative enzymes (AEs) NDX and AOX from the tunicate Ciona intestinalis (Ascidiacea) have been xenotopically expressed and characterized in human cells in culture and in the model organisms Drosophila melanogaster and mouse, with the purpose of developing bypass therapies to combat mitochondrial diseases in human patients with defective complexes I and III/IV, respectively. The fact that the genes coding for NDX and AOX have been lost from genomes of evolutionarily successful animal groups, such as vertebrates and insects, led us to investigate if the composition of the respiratory chain of Ciona and other tunicates differs significantly from that of humans and Drosophila, to accommodate the natural presence of AEs. We have failed to identify in tunicate genomes fifteen orthologous genes that code for subunits of the respiratory chain complexes; all of these putatively missing subunits are peripheral to complexes I, III and IV in mammals, and many are important for complex-complex interaction in supercomplexes (SCs), such as NDUFA11, UQCR11 and COX7A. Modeling of all respiratory chain subunit polypeptides of Ciona indicates significant structural divergence that is consistent with the lack of these fifteen clear orthologous subunits. We also provide evidence using Ciona AOX expressed in Drosophila that this AE cannot access the coenzyme Q pool reduced by complex I, but it is readily available to oxidize coenzyme Q molecules reduced by glycerophosphate oxidase, a mitochondrial inner membrane-bound dehydrogenase that is not involved in SCs. Altogether, our results suggest that Ciona AEs might have evolved in a mitochondrial inner membrane environment much different from that of mammals and insects, possibly without SCs; this correlates with the preferential functional interaction between these AEs and non-SC dehydrogenases in heterologous mammalian and insect systems. We discuss the implications of these findings for the applicability of Ciona AEs in human bypass therapies and for our understanding of the evolution of animal respiratory chain.


Subject(s)
Ciona intestinalis , Mitochondrial Proteins , Oxidative Phosphorylation , Animals , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Ciona intestinalis/genetics , Ciona intestinalis/enzymology , Humans , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Subunits/metabolism , Protein Subunits/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/enzymology , Urochordata/genetics , Urochordata/enzymology , Electron Transport , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Phylogeny , Plant Proteins
11.
Adv Sci (Weinh) ; 11(21): e2306871, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38569495

ABSTRACT

RNA splicing dysregulation and the involvement of specific splicing factors are emerging as common factors in both obesity and metabolic disorders. The study provides compelling evidence that the absence of the splicing factor SRSF1 in mature adipocytes results in whitening of brown adipocyte tissue (BAT) and impaired thermogenesis, along with the inhibition of white adipose tissue browning in mice. Combining single-nucleus RNA sequencing with transmission electron microscopy, it is observed that the transformation of BAT cell types is associated with dysfunctional mitochondria, and SRSF1 deficiency leads to degenerated and fragmented mitochondria within BAT. The results demonstrate that SRSF1 effectively binds to constitutive exon 6 of Ndufs3 pre-mRNA and promotes its inclusion. Conversely, the deficiency of SRSF1 results in impaired splicing of Ndufs3, leading to reduced levels of functional proteins that are essential for mitochondrial complex I assembly and activity. Consequently, this deficiency disrupts mitochondrial integrity, ultimately compromising the thermogenic capacity of BAT. These findings illuminate a novel role for SRSF1 in influencing mitochondrial function and BAT thermogenesis through its regulation of Ndufs3 splicing within BAT.


Subject(s)
Adipocytes, Brown , Homeostasis , Mitochondria , Serine-Arginine Splicing Factors , Thermogenesis , Animals , Thermogenesis/genetics , Thermogenesis/physiology , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Mice , Adipocytes, Brown/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Homeostasis/genetics , Homeostasis/physiology , RNA Splicing/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Male
12.
Pediatr Neurol ; 155: 91-103, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626668

ABSTRACT

BACKGROUND: Pathogenic variants in the NDUFV1 gene disrupt mitochondrial complex I, leading to neuroregression with leukoencephalopathy and basal ganglia involvement on neuroimaging. This study aims to provide a concise review on NDUFV1-related disorders while adding the largest cohort from a single center to the existing literature. METHODS: We retrospectively collected genetically proven cases of NDUFV1 pathogenic variants from our center over the last decade and explored reported instances in existing literature. Magnetic resonance imaging (MRI) patterns observed in these patients were split into three types-Leigh (putamen, basal ganglia, thalamus, and brainstem involvement), mitochondrial leukodystrophy (ML) (cerebral white matter involvement with cystic cavitations), and mixed (both). RESULTS: Analysis included 44 children (seven from our center and 37 from literature). The most prevalent comorbidities were hypertonia, ocular abnormalities, feeding issues, and hypotonia at onset. Children with the Leigh-type MRI pattern exhibited significantly higher rates of breathing difficulties, whereas those with a mixed phenotype had a higher prevalence of dystonia. The c.1156C>T variant in exon 8 of the NDUFV1 gene was the most common variant among individuals of Asian ethnicity and is predominantly associated with irritability and dystonia. Seizures and Leigh pattern of MRI of the brain was found to be less commonly associated with this variant. Higher rate of mortality was observed in children with Leigh-type pattern on brain MRI and those who did not receive mitochondrial cocktail. CONCLUSIONS: MRI phenotyping might help predict outcome. Appropriate and timely treatment with mitochondrial cocktail may reduce the probability of death and may positively impact the long-term outcomes, regardless of the genetic variant or age of onset.


Subject(s)
Electron Transport Complex I , Mitochondrial Diseases , NADH Dehydrogenase , Humans , Retrospective Studies , Male , Electron Transport Complex I/genetics , Female , Child, Preschool , Infant , Child , NADH Dehydrogenase/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/diagnostic imaging , Magnetic Resonance Imaging , Leigh Disease/genetics , Leigh Disease/diagnostic imaging , Adolescent
13.
J Cell Mol Med ; 28(8): e18276, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38546629

ABSTRACT

Histidine triad nucleotide-binding protein 2 (HINT2) is an enzyme found in mitochondria that functions as a nucleotide hydrolase and transferase. Prior studies have demonstrated that HINT2 plays a crucial role in ischemic heart disease, but its importance in cardiac remodelling remains unknown. Therefore, the current study intends to determine the role of HINT2 in cardiac remodelling. HINT2 expression levels were found to be lower in failing hearts and hypertrophy cardiomyocytes. The mice that overexpressed HINT2 exhibited reduced myocyte hypertrophy and cardiac dysfunction in response to stress. In contrast, the deficiency of HINT2 in the heart of mice resulted in a worsening hypertrophic phenotype. Further analysis indicated that upregulated genes were predominantly associated with the oxidative phosphorylation and mitochondrial complex I pathways in HINT2-overexpressed mice after aortic banding (AB) treatment. This suggests that HINT2 increases the expression of NADH dehydrogenase (ubiquinone) flavoprotein (NDUF) genes. In cellular studies, rotenone was used to disrupt mitochondrial complex I, and the protective effect of HINT2 overexpression was nullified. Lastly, we predicted that thyroid hormone receptor beta might regulate HINT2 transcriptional activity. To conclusion, the current study showcased that HINT2 alleviates pressure overload-induced cardiac remodelling by influencing the activity and assembly of mitochondrial complex I. Thus, targeting HINT2 could be a novel therapeutic strategy for reducing cardiac remodelling.


Subject(s)
Heart , Ventricular Remodeling , Animals , Mice , Ventricular Remodeling/genetics , Mitochondria , Hypertrophy , Electron Transport Complex I/genetics , Nucleotides , Hydrolases , Mitochondrial Proteins/genetics
14.
Int J Mol Sci ; 25(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38542518

ABSTRACT

Mitochondria are essential organelles that generate energy via oxidative phosphorylation. Plant mitochondrial genome encodes some of the respiratory complex subunits, and these transcripts require accurate processing, including C-to-U RNA editing and intron splicing. Pentatricopeptide repeats (PPR) proteins are involved in various organellar RNA processing events. PPR596, a P-type PPR protein, was previously identified to function in the C-to-U editing of mitochondrial rps3 transcripts in Arabidopsis. Here, we demonstrate that PPR596 functions in the cis-splicing of nad2 intron 3 in mitochondria. Loss of the PPR596 function affects the editing at rps3eU1344SS, impairs nad2 intron 3 splicing and reduces the mitochondrial complex I's assembly and activity, while inducing alternative oxidase (AOX) gene expression. This defect in nad2 intron splicing provides a plausible explanation for the slow growth of the ppr595 mutants. Although a few P-type PPR proteins are involved in RNA C-to-U editing, our results suggest that the primary function of PPR596 is intron splicing.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Gene Expression Regulation, Plant , Introns/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Plant Proteins/genetics , RNA Splicing
15.
Nat Commun ; 15(1): 1965, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38438382

ABSTRACT

The mitochondrial electron transport chain (ETC) is a highly adaptive process to meet metabolic demands of the cell, and its dysregulation has been associated with diverse clinical pathologies. However, the role and nature of impaired ETC in kidney diseases remains poorly understood. Here, we generate diabetic mice with podocyte-specific overexpression of Ndufs4, an accessory subunit of mitochondrial complex I, as a model investigate the role of ETC integrity in diabetic kidney disease (DKD). We find that conditional male mice with genetic overexpression of Ndufs4 exhibit significant improvements in cristae morphology, mitochondrial dynamics, and albuminuria. By coupling proximity labeling with super-resolution imaging, we also identify the role of cristae shaping protein STOML2 in linking NDUFS4 with improved cristae morphology. Together, we provide the evidence on the central role of NDUFS4 as a regulator of cristae remodeling and mitochondrial function in kidney podocytes. We propose that targeting NDUFS4 represents a promising approach to slow the progression of DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Male , Animals , Mice , Diabetic Nephropathies/genetics , Diabetes Mellitus, Experimental/genetics , Mitochondrial Membranes , Kidney , Mitochondria , Electron Transport Complex I/genetics
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167131, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521420

ABSTRACT

Mitochondrial DNA (mtDNA) deletions which clonally expand in skeletal muscle of patients with mtDNA maintenance disorders, impair mitochondrial oxidative phosphorylation dysfunction. Previously we have shown that these mtDNA deletions arise and accumulate in perinuclear mitochondria causing localised mitochondrial dysfunction before spreading through the muscle fibre. We believe that mito-nuclear signalling is a key contributor in the accumulation and spread of mtDNA deletions, and that knowledge of how muscle fibres respond to mitochondrial dysfunction is key to our understanding of disease mechanisms. To understand the contribution of mito-nuclear signalling to the spread of mitochondrial dysfunction, we use imaging mass cytometry. We characterise the levels of mitochondrial Oxidative Phosphorylation proteins alongside a mitochondrial mass marker, in a cohort of patients with mtDNA maintenance disorders. Our expanded panel included protein markers of key signalling pathways, allowing us to investigate cellular responses to different combinations of oxidative phosphorylation dysfunction and ragged red fibres. We find combined Complex I and IV deficiency to be most common. Interestingly, in fibres deficient for one or more complexes, the remaining complexes are often upregulated beyond the increase of mitochondrial mass typically observed in ragged red fibres. We further find that oxidative phosphorylation deficient fibres exhibit an increase in the abundance of proteins involved in proteostasis, e.g. HSP60 and LONP1, and regulation of mitochondrial metabolism (including oxidative phosphorylation and proteolysis, e.g. PHB1). Our analysis suggests that the cellular response to mitochondrial dysfunction changes depending on the combination of deficient oxidative phosphorylation complexes in each fibre.


Subject(s)
DNA, Mitochondrial , Mitochondrial Diseases , Oxidative Phosphorylation , Prohibitins , Humans , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Male , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Diseases/genetics , Female , Adult , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Signal Transduction , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics
17.
Mitochondrion ; 76: 101858, 2024 May.
Article in English | MEDLINE | ID: mdl-38437941

ABSTRACT

Mitochondrial diseases are caused by nuclear, or mitochondrial DNA (mtDNA) variants and related co-factors. Here, we report a novel m.10197G > C variant in MT-ND3 in a patient, and two other patients with m.10191 T > C. MT-ND3 variants are known to cause Leigh syndrome or mitochondrial complex I deficiency. We performed the functional analyses of the novel m.10197G > C variant that significantly lowered MT-ND3 protein levels, causing complex I assembly and activity deficiency, and reduction of ATP synthesis. We adapted a previously described re-engineering technique of delivering mitochondrial genes into mitochondria through codon optimization for nuclear expression and translation by cytoplasmic ribosomes to rescue defects arising from the MT-ND3 variants. We constructed mitochondrial targeting sequences along with the codon-optimized MT-ND3 and imported them into the mitochondria. To achieve the goal, we imported codon-optimized MT-ND3 into mitochondria in three patients with m.10197G > C and m.10191 T > C missense variants in the MT-ND3. Nuclear expression of the MT-ND3 gene partially restored protein levels, complex I deficiency, and significant improvement of ATP production indicating a functional rescue of the mutant phenotype. The codon-optimized nuclear expression of mitochondrial protein and import inside the mitochondria can supplement the requirements for ATP in energy-deficient mitochondrial disease patients.


Subject(s)
Electron Transport Complex I , Mitochondria , Mitochondrial Diseases , Humans , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/deficiency , Male , Female , Leigh Disease/genetics , Leigh Disease/metabolism , Mutation, Missense , Adenosine Triphosphate/metabolism
18.
Genet Med ; 26(6): 101117, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459834

ABSTRACT

PURPOSE: We describe 3 families with Charcot-Marie-Tooth neuropathy (CMT), harboring a homozygous NDUFS6 NM_004553.6:c.309+5G>A variant previously linked to fatal Leigh syndrome. We aimed to characterize clinically and molecularly the newly identified patients and understand the mechanism underlying their milder phenotype. METHODS: The patients underwent extensive clinical examinations. Exome sequencing was done in 4 affected individuals. The functional effect of the c.309+5G>A variant was investigated in patient-derived EBV-transformed lymphoblasts at the complementary DNA, protein, and mitochondrial level. Alternative splicing was evaluated using complementary DNA long-read sequencing. RESULTS: All patients presented with early-onset, slowly progressive axonal CMT, and nystagmus; some exhibited additional central nervous system symptoms. The c.309+5G>A substitution caused the expression of aberrantly spliced transcripts and negligible levels of the canonical transcript. Immunoblotting showed reduced levels of mutant isoforms. No detectable defects in mitochondrial complex stability or bioenergetics were found. CONCLUSION: We expand the clinical spectrum of NDUFS6-related mitochondrial disorders to include axonal CMT, emphasizing the clinical and pathophysiologic overlap between these 2 clinical entities. This work demonstrates the critical role that alternative splicing may play in modulating the severity of a genetic disorder, emphasizing the need for careful consideration when interpreting splice variants and their implications on disease prognosis.


Subject(s)
Alternative Splicing , Charcot-Marie-Tooth Disease , Mitochondrial Diseases , Humans , Alternative Splicing/genetics , Male , Female , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Child , NADH Dehydrogenase/genetics , Pedigree , Mutation/genetics , Phenotype , Exome Sequencing , Leigh Disease/genetics , Leigh Disease/pathology , Mitochondria/genetics , Mitochondria/pathology , Electron Transport Complex I/genetics , Adult , Child, Preschool , Adolescent
20.
Brain ; 147(6): 1967-1974, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38478578

ABSTRACT

Leigh syndrome spectrum (LSS) is a primary mitochondrial disorder defined neuropathologically by a subacute necrotizing encephalomyelopathy and characterized by bilateral basal ganglia and/or brainstem lesions. LSS is associated with variants in several mitochondrial DNA genes and more than 100 nuclear genes, most often related to mitochondrial complex I (CI) dysfunction. Rarely, LSS has been reported in association with primary Leber hereditary optic neuropathy (LHON) variants of the mitochondrial DNA, coding for CI subunits (m.3460G>A in MT-ND1, m.11778G>A in MT-ND4 and m.14484T>C in MT-ND6). The underlying mechanism by which these variants manifest as LSS, a severe neurodegenerative disease, as opposed to the LHON phenotype of isolated optic neuropathy, remains an open question. Here, we analyse the exome sequencing of six probands with LSS carrying primary LHON variants, and report digenic co-occurrence of the m.11778G > A variant with damaging heterozygous variants in nuclear disease genes encoding CI subunits as a plausible explanation. Our findings suggest a digenic mechanism of disease for m.11778G>A-associated LSS, consistent with recent reports of digenic disease in individuals manifesting with LSS due to biallelic variants in the recessive LHON-associated disease gene DNAJC30 in combination with heterozygous variants in CI subunits.


Subject(s)
Leigh Disease , Optic Atrophy, Hereditary, Leber , Humans , Leigh Disease/genetics , Optic Atrophy, Hereditary, Leber/genetics , Male , Female , Adult , DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Child , Adolescent , NADH Dehydrogenase/genetics , Mutation , Young Adult , Exome Sequencing , Child, Preschool
SELECTION OF CITATIONS
SEARCH DETAIL
...