Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.263
1.
Genes (Basel) ; 15(4)2024 Apr 17.
Article En | MEDLINE | ID: mdl-38674434

Oxidative phosphorylation involves a complex multi-enzymatic mitochondrial machinery critical for proper functioning of the cell, and defects herein cause a wide range of diseases called "primary mitochondrial disorders" (PMDs). Mutations in about 400 nuclear and 37 mitochondrial genes have been documented to cause PMDs, which have an estimated birth prevalence of 1:5000. Here, we describe a 4-year-old female presenting from early childhood with psychomotor delay and white matter signal changes affecting several brain regions, including the brainstem, in addition to lactic and phytanic acidosis, compatible with Leigh syndrome, a genetically heterogeneous subgroup of PMDs. Whole genome sequencing of the family trio identified a homozygous 12.9 Kb deletion, entirely overlapping the NDUFA4 gene. Sanger sequencing of the breakpoints revealed that the genomic rearrangement was likely triggered by Alu elements flanking the gene. NDUFA4 encodes for a subunit of the respiratory chain Complex IV, whose activity was significantly reduced in the patient's fibroblasts. In one family, dysfunction of NDUFA4 was previously documented as causing mitochondrial Complex IV deficiency nuclear type 21 (MC4DN21, OMIM 619065), a relatively mild form of Leigh syndrome. Our finding confirms the loss of NDUFA4 function as an ultra-rare cause of Complex IV defect, clinically presenting as Leigh syndrome.


Electron Transport Complex I , Leigh Disease , Humans , Leigh Disease/genetics , Leigh Disease/pathology , Female , Child, Preschool , Electron Transport Complex IV/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Pedigree , Sequence Deletion
2.
DNA Cell Biol ; 43(4): 158-174, 2024 Apr.
Article En | MEDLINE | ID: mdl-38588493

Polycystic ovary syndrome (PCOS) refers to an endocrine disorder syndrome that are correlated with multiple organs and systems. PCOS has an effect on women at all stages of their lives, and it has an incidence nearly ranging from 6% to 20% worldwide. Mitochondrial dysfunctions (e.g., oxidative stress, dynamic imbalance, and abnormal quality control system) have been identified in patients and animal models of PCOS, and the above processes may play a certain role in the development of PCOS and its associated complications. However, their specific pathogenic roles should be investigated in depth. In this review, recent studies on the mechanisms of action of mitochondrial dysfunction in PCOS and its associated clinical manifestations are summarized from the perspective of tissues and organs, and some studies on the treatment of the disease by improving mitochondrial function are reviewed to highlight key role of mitochondrial dysfunction in this syndrome.


Mitochondrial Diseases , Polycystic Ovary Syndrome , Animals , Female , Humans , Polycystic Ovary Syndrome/pathology , Mitochondria/metabolism , Oxidative Stress , Mitochondrial Diseases/pathology
3.
Free Radic Biol Med ; 218: 105-119, 2024 Jun.
Article En | MEDLINE | ID: mdl-38565400

Beyond their crucial role in energy production, mitochondria harbor a distinct genome subject to epigenetic regulation akin to that of nuclear DNA. This paper delves into the nascent but rapidly evolving fields of mitoepigenetics and mitoepigenomics, exploring the sophisticated regulatory mechanisms governing mitochondrial DNA (mtDNA). These mechanisms encompass mtDNA methylation, the influence of non-coding RNAs (ncRNAs), and post-translational modifications of mitochondrial proteins. Together, these epigenetic modifications meticulously coordinate mitochondrial gene transcription, replication, and metabolism, thereby calibrating mitochondrial function in response to the dynamic interplay of intracellular needs and environmental stimuli. Notably, the dysregulation of mitoepigenetic pathways is increasingly implicated in mitochondrial dysfunction and a spectrum of human pathologies, including neurodegenerative diseases, cancer, metabolic disorders, and cardiovascular conditions. This comprehensive review synthesizes the current state of knowledge, emphasizing recent breakthroughs and innovations in the field. It discusses the potential of high-resolution mitochondrial epigenome mapping, the diagnostic and prognostic utility of blood or tissue mtDNA epigenetic markers, and the promising horizon of mitochondrial epigenetic drugs. Furthermore, it explores the transformative potential of mitoepigenetics and mitoepigenomics in precision medicine. Exploiting a theragnostic approach to maintaining mitochondrial allostasis, this paper underscores the pivotal role of mitochondrial epigenetics in charting new frontiers in medical science.


DNA Methylation , DNA, Mitochondrial , Epigenesis, Genetic , Mitochondria , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Protein Processing, Post-Translational/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
4.
Adv Sci (Weinh) ; 11(18): e2307136, 2024 May.
Article En | MEDLINE | ID: mdl-38445970

In this research, a 3D brain organoid model is developed to study POLG-related encephalopathy, a mitochondrial disease stemming from POLG mutations. Induced pluripotent stem cells (iPSCs) derived from patients with these mutations is utilized to generate cortical organoids, which exhibited typical features of the diseases with POLG mutations, such as altered morphology, neuronal loss, and mitochondiral DNA (mtDNA) depletion. Significant dysregulation is also identified in pathways crucial for neuronal development and function, alongside upregulated NOTCH and JAK-STAT signaling pathways. Metformin treatment ameliorated many of these abnormalities, except for the persistent affliction of inhibitory dopamine-glutamate (DA GLU) neurons. This novel model effectively mirrors both the molecular and pathological attributes of diseases with POLG mutations, providing a valuable tool for mechanistic understanding and therapeutic screening for POLG-related disorders and other conditions characterized by compromised neuronal mtDNA maintenance and complex I deficiency.


DNA Polymerase gamma , Induced Pluripotent Stem Cells , Mitochondrial Diseases , Organoids , Organoids/metabolism , Organoids/pathology , Humans , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Brain/pathology , Brain/metabolism
5.
J Cell Mol Med ; 28(7): e18204, 2024 Apr.
Article En | MEDLINE | ID: mdl-38506068

Podocyte apoptosis exerts a crucial role in the pathogenesis of DN. Recently, long noncoding RNAs (lncRNAs) have been gradually identified to be functional in a variety of different mechanisms associated with podocyte apoptosis. This study aimed to investigate whether lncRNA Glis2 could regulate podocyte apoptosis in DN and uncover the underlying mechanism. The apoptosis rate was detected by flow cytometry. Mitochondrial membrane potential (ΔΨM) was measured using JC-1 staining. Mitochondrial morphology was detected by MitoTracker Deep Red staining. Then, the histopathological and ultrastructure changes of renal tissues in diabetic mice were observed using periodic acid-Schiff (PAS) staining and transmission electron microscopy. We found that lncRNA Glis2 was significantly downregulated in high-glucose cultured podocytes and renal tissues of db/db mice. LncRNA Glis2 overexpression was found to alleviate podocyte mitochondrial dysfunction and apoptosis. The direct interaction between lncRNA Glis2 and miR-328-5p was confirmed by dual luciferase reporter assay. Furthermore, lncRNA Glis2 overexpression alleviated podocyte apoptosis in diabetic mice. Taken together, this study demonstrated that lncRNA Glis2, acting as a competing endogenous RNA (ceRNA) of miRNA-328-5p, regulated Sirt1-mediated mitochondrial dysfunction and podocyte apoptosis in DN.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , MicroRNAs , Mitochondrial Diseases , Podocytes , RNA, Long Noncoding , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , RNA, Long Noncoding/genetics , MicroRNAs/genetics , Podocytes/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Transcription Factors , Apoptosis/genetics , Mitochondrial Diseases/pathology , Glucose
6.
Stem Cell Res ; 76: 103379, 2024 Apr.
Article En | MEDLINE | ID: mdl-38458030

Leigh syndrome is a rare autosomal recessive disorder showcasing a diverse range of neurological symptoms. Classical Leigh syndrome is associated with mitochondrial complex I deficiency, primarily resulting from biallelic mutations in the NDUFAF5 gene, encoding the NADH:ubiquinone oxidoreductase complex assembly factor 5. Using the Sendai virus delivery system, we generated an induced pluripotent stem cell line from peripheral blood mononuclear cells of a 47-years-old female patient who carried a homozygous NDUFAF5 c.836 T > G (p.Met279Arg) mutation. This cellular model serves as a tool for investigating the underlying pathogenic mechanisms and for the development of potential treatments for Leigh syndrome.


Induced Pluripotent Stem Cells , Leigh Disease , Mitochondrial Diseases , Humans , Female , Middle Aged , Leigh Disease/genetics , Mutation, Missense , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mutation , Methyltransferases/genetics , Mitochondrial Proteins/genetics
7.
Front Biosci (Schol Ed) ; 16(1): 7, 2024 Mar 14.
Article En | MEDLINE | ID: mdl-38538347

Disorders of mitochondrial function are responsible for many inherited neuromuscular and metabolic diseases. Their combination of high mortality, multi-systemic involvement, and economic burden cause devastating effects on patients and their families. Molecular diagnostic tools are becoming increasingly important in providing earlier diagnoses and guiding more precise therapeutic treatments for patients suffering from mitochondrial disorders. This review addresses fundamental molecular concepts relating to the pathogenesis of mitochondrial dysfunction and disorders. A series of short cases highlights the various clinical presentations, inheritance patterns, and pathogenic mutations in nuclear and mitochondrial genes that cause mitochondrial diseases. Graphical and tabular representations of the results are presented to guide the understanding of the important concepts related to mitochondrial molecular genetics and pathology. Emerging technology is incorporating preimplantation genetic testing for mtDNA disorders, while mitochondrial replacement shows promise in significantly decreasing the transfer of diseased mitochondrial DNA (mtDNA) to embryos. Medical professionals must maintain an in-depth understanding of the gene mutations and molecular mechanisms underlying mitochondrial disorders. Continued diagnostic advances and comprehensive management of patients with mitochondrial disorders are essential to achieve robust clinical impacts from comprehensive genomic testing. This is especially true when supported by non-genetic tests such as biochemical analysis, histochemical stains, and imaging studies. Such a multi-pronged investigation should improve the management of mitochondrial disorders by providing accurate and timely diagnoses to reduce disease burden and improve the lives of patients and their families.


Mitochondrial Diseases , Humans , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Genes, Mitochondrial
8.
Front Biosci (Schol Ed) ; 16(1): 5, 2024 Mar 08.
Article En | MEDLINE | ID: mdl-38538341

The pathogenesis of type 2 diabetes mellitus (T2DM) is based on the development of insulin resistance, which is a disruption to the ability of the tissues to bind to insulin, leading to a general metabolic disorder. Mitochondria are the main participants in cellular energy metabolism, meaning their dysfunction is associated with the development of insulin resistance in T2DM. Mitochondrial function is affected by insulin resistance in various tissues, including skeletal muscle and the liver, which greatly influence glucose homeostasis throughout the body. This review studies mitochondrial dysfunction in T2DM and its impact on disease progression. In addition, it considers the causes underlying the development of mitochondrial dysfunction in T2DM, including mutations in the mitochondrial genome, mitochondrial DNA methylation, and other epigenetic influences, as well as the impact of impaired mitochondrial membrane potential. New therapeutic strategies for diabetes that have been developed to target the mitochondria will also be presented.


Diabetes Mellitus, Type 2 , Insulin Resistance , Metabolic Diseases , Mitochondrial Diseases , Humans , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Mitochondria/metabolism , Mitochondria/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Energy Metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology
9.
Cell Stress Chaperones ; 29(2): 349-357, 2024 Apr.
Article En | MEDLINE | ID: mdl-38485043

This comprehensive review delves into the pivotal role of mitochondria in doxorubicin-induced cardiotoxicity, a significant complication limiting the clinical use of this potent anthracycline chemotherapeutic agent. Doxorubicin, while effective against various malignancies, is associated with dose-dependent cardiotoxicity, potentially leading to irreversible cardiac damage. The review meticulously dissects the molecular mechanisms underpinning this cardiotoxicity, particularly focusing on mitochondrial dysfunction, a central player in this adverse effect. Central to the discussion is the concept of mitochondrial quality control, including mitochondrial dynamics (fusion/fission balance) and mitophagy. The review presents evidence linking aberrations in these processes to cardiotoxicity in doxorubicin-treated patients. It elucidates how doxorubicin disrupts mitochondrial dynamics, leading to an imbalance between mitochondrial fission and fusion, and impairs mitophagy, culminating in the accumulation of dysfunctional mitochondria and subsequent cardiac cell damage. Furthermore, the review explores emerging therapeutic strategies targeting mitochondrial dysfunction. It highlights the potential of modulating mitochondrial dynamics and enhancing mitophagy to mitigate doxorubicin-induced cardiac damage. These strategies include pharmacological interventions with mitochondrial fission inhibitors, fusion promoters, and agents that modulate mitophagy. The review underscores the promising results from preclinical studies while advocating for more extensive clinical trials to validate these approaches in human patients. In conclusion, this review offers valuable insights into the intricate relationship between mitochondrial dysfunction and doxorubicin-mediated cardiotoxicity. It underscores the need for continued research into targeted mitochondrial therapies as a means to improve the cardiac safety profile of doxorubicin, thereby enhancing the overall treatment outcomes for cancer patients.


Cardiotoxicity , Mitochondrial Diseases , Humans , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Doxorubicin/adverse effects , Mitochondria , Antibiotics, Antineoplastic/adverse effects , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Myocytes, Cardiac
10.
Wiad Lek ; 77(1): 153-159, 2024.
Article En | MEDLINE | ID: mdl-38431820

OBJECTIVE: Aim: To determine the role of mitochondrial dysfunction in the pathogenesis of atherosclerosis based on the analysis of research data and statistics from the MEDLINE, Scopus and Web of Science Core Collection electronic databases for 2007-2023. PATIENTS AND METHODS: Materials and Methods: A comprehensive review of literature sources from the MEDLINE, Scopus and Web of Science Core Collection electronic databases was conducted to critically analyse the data and determine the role of mitochondrial dysfunction in the pathogenesis of atherosclerosis. CONCLUSION: Conclusions: In this review, we have summarized the latest literature data on the association between mitochondrial dysfunction and the development of atherosclerosis. Mitochondria have been recognized as a novel therapeutic target in the development of atherosclerosis. However, the presence of current gaps in therapeutic strategies for mitochondrial dysfunction control still hinders clinical success in the prevention and treatment of atherosclerosis. Both antioxidants and gene therapy are appealing approaches to treating atherosclerosis. Nevertheless, further research is needed to determine the proper therapeutic strategy to reduce the impact of mitochondrial dysfunction on the progression of atherosclerosis.


Atherosclerosis , Mitochondrial Diseases , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism , Atherosclerosis/etiology , Atherosclerosis/pathology , Mitochondria , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology
11.
Int J Med Sci ; 21(4): 714-724, 2024.
Article En | MEDLINE | ID: mdl-38464839

Sepsis-induced cardiomyopathy (SIC) represents a severe complication of systemic infection, characterized by significant cardiac dysfunction. This study examines the role of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Inverted Formin 2 (INF2) in the pathogenesis of SIC, focusing on their impact on mitochondrial homeostasis and dynamics. Our research demonstrates that silencing DNA-PKcs alleviates lipopolysaccharide (LPS)-induced cardiomyocyte death and dysfunction. Using HL-1 cardiomyocytes treated with LPS, we observed that DNA-PKcs knockdown notably reverses LPS-induced cytotoxicity, indicating a protective role against cellular damage. This effect is further substantiated by the reduction in caspase-3 and caspase-9 activation, key markers of apoptosis, upon DNA-PKcs knockdown. Besides, our data further reveal that DNA-PKcs knockdown attenuates LPS-induced mitochondrial dysfunction, evidenced by improved ATP production, enhanced activities of mitochondrial respiratory complexes, and preserved mitochondrial membrane potential. Moreover, DNA-PKcs deletion counteracts LPS-induced shifts towards mitochondrial fission, indicating its regulatory influence on mitochondrial dynamics. Conclusively, our research elucidates the intricate interplay between DNA-PKcs and INF2 in the modulation of mitochondrial function and dynamics during sepsis-induced cardiomyopathy. These findings offer new insights into the molecular mechanisms underpinning SIC and suggest potential therapeutic targets for mitigating mitochondrial dysfunction in this critical condition.


Cardiomyopathies , Mitochondrial Diseases , Sepsis , Humans , DNA-Activated Protein Kinase/metabolism , Mitochondrial Dynamics , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Catalytic Domain , Cardiomyopathies/genetics , Myocytes, Cardiac , Sepsis/complications , Sepsis/genetics , Mitochondrial Diseases/pathology , DNA/adverse effects , DNA/metabolism
12.
Cancer Lett ; 588: 216802, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38467180

Multiple myeloma (MM) is a hematological malignancy that remains incurable, primarily due to the high likelihood of relapse or development of resistance to current treatments. To explore and discover new medications capable of overcoming drug resistance in MM, we conducted cell viability inhibition screens of 1504 FDA-approved drugs. Lomitapide, a cholesterol-lowering agent, was found to exhibit effective inhibition on bortezomib-resistant MM cells in vitro and in vivo. Our data also indicated that lomitapide decreases the permeability of the mitochondrial outer membrane and induces mitochondrial dysfunction in MM cells. Next, lomitapide treatment upregulated DRP1 and PINK1 expression levels, coupled with the mitochondrial translocation of Parkin, leading to MM cell mitophagy. Excessive mitophagy caused mitochondrial damage and dysfunction induced by lomitapide. Meanwhile, PARP14 was identified as a direct target of lomitapide by SPR-HPLC-MS, and we showed that DRP1-induced mitophagy was crucial in the anti-MM activity mediated by PARP14. Furthermore, PARP14 is overexpressed in MM patients, implying that it is a novel therapeutic target in MM. Collectively, our results demonstrate that DRP1-mediated mitophagy induced by PARP14 may be the cause for mitochondrial dysfunction and damage in response to lomitapide treatment.


Benzimidazoles , Mitochondrial Diseases , Multiple Myeloma , Humans , Mitophagy , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Mitochondria/metabolism , Neoplasm Recurrence, Local/pathology , Drug Resistance , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Poly(ADP-ribose) Polymerases/metabolism
13.
Int Immunopharmacol ; 130: 111685, 2024 Mar 30.
Article En | MEDLINE | ID: mdl-38377860

The NET family member, CDGSH iron-sulfur domain-containing protein 1 (CISD1), is located in theoutermembrane of mitochondria, where it regulates energy and iron metabolism. CISD1 has vital functions in certain human diseases; however, its function in acute lung injury (ALI) is unknown. ALI pathogenesis critically involves mitochondrial dysfunction and ferroptosis, which might be regulated by CISD1. Therefore, we investigated CISD1's function in mitochondrial dysfunction and ferroptosis regulation in lipopolysaccharide (LPS)-induced ALI. We found that CISD1 was upregulated in LPS-induced ALI,and silencing Cisd1 prevented cell apoptosis and increased cell viability. When CISD1was inhibited by mitoNEET ligand-1 (NL-1) there was a significant mitigation of pathological injury and lung edema, and reduced numbers of total cells, polymorphonuclear leukocytes, and a decreased protein content in the bronchoalveolar lavage fluid (BALF). Moreover, inhibition of CISD1 markedly decreased the interleukin (IL)6, IL-1ß, and tumor necrosis factor alpha (TNF-α) levels in the lungs and BALF of ALI-model mice. Silencing of Cisd1 prevented LPS-induced mitochondrial membrane potential depolarization, cellular ATP reduction, and reactive oxygen species (ROS) accumulation, suggesting mitochondrial protection. ALI activated ferroptosis, as evidenced by the increased lipid-ROS, intracellular Fe2+ level, reduced Gpx4 (glutathione peroxidase 4) expression, and the glutathione/glutathione disulfide ratio. Interestingly, inhibition of CISD1 reduced LPS-induced ferroptosis in vivo and in vitro. In conclusion, inhibition of CISD1 alleviated mitochondrial dysfunction and ferroptosis in LPS-induced ALI, identifying CISD1 as possible target for therapy of LPS-induced ALI.


Acute Lung Injury , Ferroptosis , Iron-Binding Proteins , Animals , Humans , Mice , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Interleukin-6/metabolism , Iron/metabolism , Iron-Binding Proteins/antagonists & inhibitors , Lipopolysaccharides/metabolism , Lung/pathology , Membrane Proteins/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Reactive Oxygen Species/metabolism
14.
J Pediatr Endocrinol Metab ; 37(3): 260-270, 2024 Mar 25.
Article En | MEDLINE | ID: mdl-38353291

OBJECTIVES: Primary Coenzyme Q10 Deficiency-7 (OMIM 616276) results from bi-allelic pathogenic variants in the COQ4 gene. Common clinical findings include hypotonia, seizures, respiratory distress, and cardiomyopathy. In this report, we present two patients diagnosed with Primary Coenzyme Q10 Deficiency-7 along with a review of previously published cases, with the aim being to provide a better understanding of the clinical and laboratory manifestations of the disease. CASE PRESENTATION: A 3-month-and-22-day-old male was admitted to our outpatient clinic due to poor feeding and restlessness. He was born following an uneventful pregnancy to a nonconsanguineous marriage. A physical examination revealed hypotonia, a dolichocephaly, periorbital edema, and long eyelashes. Blood tests revealed metabolic acidosis and elevated serum lactate levels, while the genetic analysis revealed a variant previously reported as pathogenic, c.437T>G (p.Phe146Cys), in the COQ4 gene. Genetic tests were also conducted on both mother and father, and it revealed heterozygous variant, 0.437T>G (p.Phe146Cys), in the COQ4 gene. As a result of these findings, the patient was diagnosed with neonatal encephalomyopathy-cardiomyopathy-respiratory distress syndrome (Primary Coenzyme Q10 Deficiency-7). A 1-year-old male was admitted to our clinic with complaints of hypotonia, seizures, and feeding difficulties. He was born following an uneventful pregnancy to a nonconsanguineous marriage. On his first day of life, he was admitted to the neonatal intensive care unit due to poor feeding and hypotonia. A physical examination revealed microcephaly, a high palate, poor feeding, weak crying, hypotonia, bilateral horizontal nystagmus, and inability to maintain eye contact. Laboratory findings were within normal limits, while a whole exome sequencing analysis revealed a homozygous variant previously reported as pathogenic, c.458C>T (p.A153V), in the COQ4 gene. The patient was diagnosed with Primary Coenzyme Q10 Deficiency-7. CONCLUSIONS: Primary Coenzyme Q10 Deficiency-7 should be considered in the differential diagnosis of infants presenting with neurological and dysmorphic manifestations.


Ataxia , Cardiomyopathies , Mitochondrial Diseases , Muscle Weakness , Ubiquinone/deficiency , Infant , Infant, Newborn , Pregnancy , Female , Humans , Male , Muscle Hypotonia/etiology , Muscle Hypotonia/genetics , Mitochondrial Diseases/pathology , Ubiquinone/genetics , Seizures/complications , Cardiomyopathies/complications
15.
Circ Res ; 134(5): 482-501, 2024 03.
Article En | MEDLINE | ID: mdl-38323474

BACKGROUND: Mitochondrial dysfunction is a primary driver of cardiac contractile failure; yet, the cross talk between mitochondrial energetics and signaling regulation remains obscure. Ponatinib, a tyrosine kinase inhibitor used to treat chronic myeloid leukemia, is among the most cardiotoxic tyrosine kinase inhibitors and causes mitochondrial dysfunction. Whether ponatinib-induced mitochondrial dysfunction triggers the integrated stress response (ISR) to induce ponatinib-induced cardiotoxicity remains to be determined. METHODS: Using human induced pluripotent stem cells-derived cardiomyocytes and a recently developed mouse model of ponatinib-induced cardiotoxicity, we performed proteomic analysis, molecular and biochemical assays to investigate the relationship between ponatinib-induced mitochondrial stress and ISR and their role in promoting ponatinib-induced cardiotoxicity. RESULTS: Proteomic analysis revealed that ponatinib activated the ISR in cardiac cells. We identified GCN2 (general control nonderepressible 2) as the eIF2α (eukaryotic translation initiation factor 2α) kinase responsible for relaying mitochondrial stress signals to trigger the primary ISR effector-ATF4 (activating transcription factor 4), upon ponatinib exposure. Mechanistically, ponatinib treatment exerted inhibitory effects on ATP synthase activity and reduced its expression levels resulting in ATP deficits. Perturbed mitochondrial function resulting in ATP deficits then acts as a trigger of GCN2-mediated ISR activation, effects that were negated by nicotinamide mononucleotide, an NAD+ precursor, supplementation. Genetic inhibition of ATP synthase also activated GCN2. Interestingly, we showed that the decreased abundance of ATP also facilitated direct binding of ponatinib to GCN2, unexpectedly causing its activation most likely because of a conformational change in its structure. Importantly, administering an ISR inhibitor protected human induced pluripotent stem cell-derived cardiomyocytes against ponatinib. Ponatinib-treated mice also exhibited reduced cardiac function, effects that were attenuated upon systemic ISRIB administration. Importantly, ISRIB does not affect the antitumor effects of ponatinib in vitro. CONCLUSIONS: Neutralizing ISR hyperactivation could prevent or reverse ponatinib-induced cardiotoxicity. The findings that compromised ATP production potentiates GCN2-mediated ISR activation have broad implications across various cardiac diseases. Our results also highlight an unanticipated role of ponatinib in causing direct activation of a kinase target despite its role as an ATP-competitive kinase inhibitor.


Imidazoles , Induced Pluripotent Stem Cells , Mitochondrial Diseases , Pyridazines , Humans , Animals , Mice , Protein Serine-Threonine Kinases/metabolism , Cardiotoxicity/pathology , Proteomics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/toxicity , Mitochondrial Diseases/pathology , Adenosine Triphosphate
16.
Ageing Res Rev ; 96: 102248, 2024 Apr.
Article En | MEDLINE | ID: mdl-38408490

Temporal lobe epilepsy (TLE) is the most common form of epileptic syndrome. It has been established that due to its complex pathogenesis, a considerable proportion of TLE patients often progress to drug-resistant epilepsy. Ferroptosis has emerged as an important neuronal death mechanism in TLE, which is primarily influenced by lipid accumulation and oxidative stress. In previous studies of ferroptosis, more attention has been focused on the impact of changes in the levels of proteins related to the redox equilibrium and signaling pathways on epileptic seizures. However, it is worth noting that the oxidative-reduction changes in different organelles may have different pathophysiological significance in the process of ferroptosis-related diseases. Mitochondria, as a key organelle involved in ferroptosis, its structural damage and functional impairment can lead to energy metabolism disorders and disruption of the excitatory inhibitory balance, significantly increasing the susceptibility to epileptic seizures. Therefore, secondary mitochondrial dysfunction in the process of ferroptosis could play a crucial role in TLE pathogenesis. This review focuses on ferroptosis and mitochondria, discussing the pathogenic role of ferroptosis-related mitochondrial dysfunction in TLE, thus aiming to provide novel insights and potential implications of ferroptosis-related secondary mitochondrial dysfunction in epileptic seizures and to offer new insights for the precise exploration of ferroptosis-related therapeutic targets for TLE patients.


Epilepsy, Temporal Lobe , Ferroptosis , Mitochondrial Diseases , Humans , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Seizures/complications , Seizures/metabolism , Seizures/pathology , Mitochondria/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology
17.
Acta Physiol (Oxf) ; 240(4): e14121, 2024 Apr.
Article En | MEDLINE | ID: mdl-38409944

AIM: Mitochondrial dysfunction, a characteristic pathological feature of renal Ischemic/reperfusion injury (I/RI), predisposes tubular epithelial cells to maintain an inflammatory microenvironment, however, the exact mechanisms through which mitochondrial dysfunction modulates the induction of tubular injury remains incompletely understood. METHODS: ESI-QTRAP-MS/MS approach was used to characterize the targeted metabolic profiling of kidney with I/RI. Tubule injury, mitochondrial dysfunction, and fumarate level were evaluated using qPCR, transmission electron microscopy, ELISA, and immunohistochemistry. RESULTS: We demonstrated that tubule injury occurred at the phase of reperfusion in murine model of I/RI. Meanwhile, enhanced glycolysis and mitochondrial dysfunction were found to be associated with tubule injury. Further, we found that tubular fumarate, which resulted from fumarate hydratase deficiency and released from dysfunctional mitochondria, promoted tubular injury. Mechanistically, fumarate induced tubular injury by causing disturbance of glutathione (GSH) hemostasis. Suppression of GSH with buthionine sulphoximine administration could deteriorate the fumarate inhibition-mediated tubule injury recovery. Reactive oxygen species/NF-κB signaling activation played a vital role in fumarate-mediated tubule injury. CONCLUSION: Our studies demonstrated that the mitochondrial-derived fumarate promotes tubular epithelial cell injury in renal I/RI. Blockade of fumarate-mediated ROS/NF-κB signaling activation may serve as a novel therapeutic approach to ameliorate hypoxic tubule injury.


Acute Kidney Injury , Mitochondrial Diseases , Reperfusion Injury , Mice , Animals , NF-kappa B/metabolism , Tandem Mass Spectrometry , Kidney/metabolism , Mitochondria/metabolism , Reperfusion Injury/metabolism , Reperfusion , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Ischemia/pathology , Apoptosis
18.
Curr Opin Neurobiol ; 85: 102841, 2024 Apr.
Article En | MEDLINE | ID: mdl-38306948

Parkinson's disease (PD) is a growing cause of disability worldwide and there is a critical need for the development of disease-modifying therapies to slow or stop disease progression. Recent advances in characterizing the genetics of PD have expanded our understanding of the cell biology of this disorder. Mitochondrial oxidative stress, defects in synaptic function, and impaired lysosomal activity have been shown to be linked in PD, resulting in a pathogenic feedback cycle involving the accumulation of toxic oxidized dopamine and alpha-synuclein. In this review, we will highlight recent data on a subset of PD-linked genes which have key roles in these pathways and the pathogenic cycle. We will furthermore discuss findings highlighting the importance of dynamic mitochondria-lysosome contact sites that mediate direct inter-organelle cross-talk in the pathogenesis of PD and related disorders.


Mitochondrial Diseases , Parkinson Disease , Humans , Parkinson Disease/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology
19.
Hum Mol Genet ; 33(10): 860-871, 2024 May 04.
Article En | MEDLINE | ID: mdl-38324746

Neuromuscular disorders caused by dysfunction of the mitochondrial respiratory chain are common, severe and untreatable. We recovered a number of mitochondrial genes, including electron transport chain components, in a large forward genetic screen for mutations causing age-related neurodegeneration in the context of proteostasis dysfunction. We created a model of complex I deficiency in the Drosophila retina to probe the role of protein degradation abnormalities in mitochondrial encephalomyopathies. Using our genetic model, we found that complex I deficiency regulates both the ubiquitin/proteasome and autophagy/lysosome arms of the proteostasis machinery. We further performed an in vivo kinome screen to uncover new and potentially druggable mechanisms contributing to complex I related neurodegeneration and proteostasis failure. Reduction of RIOK kinases and the innate immune signaling kinase pelle prevented neurodegeneration in complex I deficiency animals. Genetically targeting oxidative stress, but not RIOK1 or pelle knockdown, normalized proteostasis markers. Our findings outline distinct pathways controlling neurodegeneration and protein degradation in complex I deficiency and introduce an experimentally facile model in which to study these debilitating and currently treatment-refractory disorders.


Disease Models, Animal , Drosophila Proteins , Electron Transport Complex I , Electron Transport Complex I/deficiency , Mitochondria , Mitochondrial Diseases , Proteostasis , Animals , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/pathology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/deficiency , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Autophagy/genetics , Oxidative Stress/genetics , Drosophila melanogaster/genetics , Mutation , Lysosomes/metabolism , Lysosomes/genetics , Drosophila/genetics , Drosophila/metabolism , Signal Transduction
20.
Circulation ; 149(16): 1268-1284, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38362779

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a common heritable heart disease. Although HCM has been reported to be associated with many variants of genes involved in sarcomeric protein biomechanics, pathogenic genes have not been identified in patients with partial HCM. FARS2 (the mitochondrial phenylalanyl-tRNA synthetase), a type of mitochondrial aminoacyl-tRNA synthetase, plays a role in the mitochondrial translation machinery. Several variants of FARS2 have been suggested to cause neurological disorders; however, FARS2-associated diseases involving other organs have not been reported. We identified FARS2 as a potential novel pathogenic gene in cardiomyopathy and investigated its effects on mitochondrial homeostasis and the cardiomyopathy phenotype. METHODS: FARS2 variants in patients with HCM were identified using whole-exome sequencing, Sanger sequencing, molecular docking analyses, and cell model investigation. Fars2 conditional mutant (p.R415L) or knockout mice, fars2-knockdown zebrafish, and Fars2-knockdown neonatal rat ventricular myocytes were engineered to construct FARS2 deficiency models both in vivo and in vitro. The effects of FARS2 and its role in mitochondrial homeostasis were subsequently evaluated using RNA sequencing and mitochondrial functional analyses. Myocardial tissues from patients were used for further verification. RESULTS: We identified 7 unreported FARS2 variants in patients with HCM. Heart-specific Fars2-deficient mice presented cardiac hypertrophy, left ventricular dilation, progressive heart failure accompanied by myocardial and mitochondrial dysfunction, and a short life span. Heterozygous cardiac-specific Fars2R415L mice displayed a tendency to cardiac hypertrophy at age 4 weeks, accompanied by myocardial dysfunction. In addition, fars2-knockdown zebrafish presented pericardial edema and heart failure. FARS2 deficiency impaired mitochondrial homeostasis by directly blocking the aminoacylation of mt-tRNAPhe and inhibiting the synthesis of mitochondrial proteins, ultimately contributing to an imbalanced mitochondrial quality control system by accelerating mitochondrial hyperfragmentation and disrupting mitochondrion-related autophagy. Interfering with the mitochondrial quality control system using adeno-associated virus 9 or specific inhibitors mitigated the cardiac and mitochondrial dysfunction triggered by FARS2 deficiency by restoring mitochondrial homeostasis. CONCLUSIONS: Our findings unveil the previously unrecognized role of FARS2 in heart and mitochondrial homeostasis. This study may provide new insights into the molecular diagnosis and prevention of heritable cardiomyopathy as well as therapeutic options for FARS2-associated cardiomyopathy.


Cardiomyopathy, Hypertrophic , Heart Failure , Mitochondrial Diseases , Phenylalanine-tRNA Ligase , Animals , Humans , Infant, Newborn , Mice , Rats , Cardiomyopathy, Hypertrophic/pathology , Heart Failure/pathology , Homeostasis , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/metabolism , Molecular Docking Simulation , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , Zebrafish/genetics , Mutation
...