ABSTRACT
Acute kidney injury (AKI) is a common complication of cisplatin chemotherapy, which greatly limits its clinical effect and application. This study explored the function of solute Carrier Family 31 Member 1 (SLC31A1) in cisplatin-induced AKI and its possible mechanism. Mice and HK-2 cells were exposed to cisplatin to establish the in vivo and in vitro AKI models. Cell viability was detected by CCK-8. Mitochondrial and oxidative damage was determined by Mito-Tracker Green staining, mtROS level, ATP production, mitochondrial membrane potential, MDA content and CAT activity. AKI was evaluated by renal function and histopathological changes. Apoptosis was detected by TUNEL and caspase-3 expression. Molecule expression was measured by RT-qPCR, Western blotting, and immunohistochemistry. Molecular mechanism was studied by luciferase reporter assay and ChIP. SLC31A1 level was predominantly increased by cisplatin exposure in AKI models. Notably, copper ion (Cu+) level was enhanced by cisplatin challenge. Moreover, Cu+ supplementation intensified cisplatin-induced cell death, mitochondrial dysfunction, and oxidative stress in HK-2 cells, indicating the involvement of cuproptosis in cisplatin-induced AKI, whereas these changes were partially counteracted by SLC31A1 knockdown. E74 like ETS transcription factor 3 (ELF3) could directly bind to SLC31A1 promoter and promote its transcription. ELF3 was up-regulated and positively correlated with SLC31A1 expression upon cisplatin-induced AKI. SLC31A1 silencing restored renal function, alleviated mitochondrial dysfunction, and apoptosis in cisplatin-induced AKI mice. ELF3 transcriptionally activated SLC31A1 to trigger cuproptosis that drove cisplatin-induced AKI through mitochondrial dysfunction, indicating that SLC31A1 might be a promising therapeutic target to mitigate AKI during cisplatin chemotherapy.
Subject(s)
Acute Kidney Injury , Cisplatin , Copper , Animals , Mice , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Apoptosis , Cisplatin/adverse effects , Copper/metabolism , Copper Transport Proteins , Mitochondrial Diseases/complicationsABSTRACT
Septic cardiomyopathy (SCM) has a high incidence and complex pathogenesis, which can significantly increase the mortality of sepsis patients. NOD-like receptor protein 3 (NLRP3) inflammatory corpuscles play an important role in the pathogenesis of SCM. Mitochondrial dysfunction in cardiomyocytes is also one of the important pathogenesis of SCM. Activation of NLRP3 inflammatory corpuscles is closely related to mitochondrial dysfunction. The study of interaction mechanism between the two is helpful to find a new therapeutic scheme for SCM. This article reviews the interaction between NLRP3 inflammatory corpuscles and mitochondrial dysfunction in the pathogenesis of SCM, as well as the related mechanisms of traditional Chinese medicine (TCM) prevention and treatment of SCM, providing theoretical reference for further exploring therapeutic targets for SCM.
Subject(s)
Cardiomyopathies , Mitochondrial Diseases , Sepsis , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Cardiomyopathies/etiology , Sepsis/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolismABSTRACT
Polycystic ovary syndrome(PCOS) is a highly prevalent endocrine and reproductive disorder characterized by ovulatory dysfunction, hyperandrogenism(HA), and polycystic ovarian morphology(PCOM). It is often accompanied by insulin resistance(IR), obesity, and metabolic disorders and can lead to cardiovascular diseases, endometrial carcinoma and many other late complications, seriously affecting the physical and mental health and quality of life in premenopausal women. The etiology of PCOS is still unknown and many scholars assume that mitochondrial dysfunction may represent a major pathogenic factor in PCOS in recent years. With a holistic view, treatment based on syndrome differentiation, and multi-system and multi-target treatment manner, traditional Chinese medicine(TCM) can mitigate the symptoms and signs of PCOS from multiple aspects. Although there have been reviews on the mechanism of mitochondrial dysfunction in PCOS, there is still a lack of reviews on the intervention of mitochondrial function by TCM to treat PCOS. Therefore, this paper focuses on the role of mitochondrial dysfunction in PCOS and summarizes the studies about the TCM intervention of PCOS by regulating the mitochondrial function, inflammation, oxidative stress(OS), autophagy, and apoptosis in the last five years, aiming to shed new light on the prevention and treatment of PCOS with TCM.
Subject(s)
Medicine, Chinese Traditional , Mitochondrial Diseases , Polycystic Ovary Syndrome , Female , Humans , Mitochondrial Diseases/complications , Mitochondrial Diseases/therapy , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/therapyABSTRACT
INTRODUCTION: Primary coenzyme Q10 (CoQ10) deficiency, a recessive disorder associated with various defects of CoQ10 biosynthesis and widely varying clinical presentation, is customarily managed by oral Q10 supplementation but the benefit is debated. METHODS: To address this question, we mapped individual responses in two patients with COQ8A-related ataxia following coenzyme Q10 supplementation using noninvasive imaging. Metabolic 31phosphorus magnetic resonance spectroscopy imaging (31P-MRSI) and volumetric cerebellar neuroimaging were performed to quantify the individual treatment response in two patients with COQ8A-related ataxia, each compared with eight age- and gender-matched healthy control subjects. RESULTS: Post-treatment change in energy metabolite levels differed in the two patients, with higher energy levels and improved dysarthria and leg coordination in one, and decreased energy levels without clinical benefit in the other. CONCLUSIONS: Our results suggest that the cerebellar bioenergetic state may predict treatment response in COQ8A-related ataxia and highlight the potential of pathophysiology-orientated neuroimaging evidence to inform treatment decisions.
Subject(s)
Cerebellar Ataxia , Mitochondrial Diseases , Ataxia/complications , Ataxia/diagnostic imaging , Ataxia/drug therapy , Cerebellar Ataxia/complications , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/drug therapy , Energy Metabolism , Humans , Mitochondrial Diseases/complications , Muscle Weakness/complications , Ubiquinone/deficiency , Ubiquinone/therapeutic useABSTRACT
BACKGROUND: Brown-Vialetto-Van Laere syndrome (BVVLS) is a rare, treatable neurodegenerative disorder with a variable clinical presentation, caused by mutations in three different riboflavin transporter genes. CASE: An 11-year-old-boy presented with respiratory insufficiency and a rapidly progressive muscle weakness. He was the fifth child of a consanguineous marriage with a medical history of hearing loss. He was peripherally week with a reduced muscle tone. Upper extremity muscles were effected more than lower limbs. He deteriorated rapidly and became quadriplegic. Brain magnetic resonance imaging and magnetic resonance spectroscopy were normal. Echocardiography revealed left ventricular non-compaction. A homozygous c.1088C > T (p.363L) missense mutation was identified in SLC52A2 gene. Significant clinical improvement was seen with high dose riboflavin. CONCLUSION: This is the first reported BVVLS case presented with left ventricle-non compaction which may be caused by a secondary respiratory chain deficiency. Riboflavin transporter deficiencies should be considered in the differential diagnosis of mitochondrial disorders and secondary respiratory chain deficiencies should be thought during the follow-up of BVVLS.
Subject(s)
Bulbar Palsy, Progressive , Mitochondrial Diseases , Bulbar Palsy, Progressive/diagnosis , Bulbar Palsy, Progressive/genetics , Child , Hearing Loss, Sensorineural , Heart Ventricles , Humans , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Receptors, G-Protein-CoupledABSTRACT
Coenzyme Q10 (CoQ10) is a ubiquitous cofactor in the body, operating in the inner mitochondrial membrane, where it plays a vital role in the generation of adenosine triphosphate (ATP) through the electron transport chain (ETC). In addition to this, CoQ10 serves as an antioxidant, protecting the cell from oxidative stress by reactive oxygen species (ROS) as well as maintaining a proton (H+) gradient across lysosome membranes to facilitate the breakdown of cellular waste products. Through the process of ageing, the body becomes deficient in CoQ10, resulting in several systemic manifestations. On a cellular level, one of the consequences of CoQ10 deficiency is apoptosis, which can be visualised in tissues of the central nervous system (CNS). Diseases affecting the retina and brain such as age-related macular degeneration (AMD), glaucoma, Alzheimer's disease (AD) and Parkinson's disease (PD) have shown defects in cellular biochemical reactions attributed to reduced levels of CoQ10. Through further research into the pathogenesis of such conditions, the effects of CoQ10 deficiency can be counteracted through supplementation, early detection and intervention.
Subject(s)
Ataxia/metabolism , Brain/metabolism , Mitochondrial Diseases/metabolism , Muscle Weakness/metabolism , Neurodegenerative Diseases/metabolism , Retina/metabolism , Ubiquinone/deficiency , Animals , Ataxia/complications , Ataxia/pathology , Brain/pathology , Humans , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Muscle Weakness/complications , Muscle Weakness/pathology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Retina/pathology , Ubiquinone/analogs & derivatives , Ubiquinone/metabolismABSTRACT
Several lines of evidence implicate mitochondria in the pathophysiology of autism spectrum disorder (ASD). In this review, we outline some of the evidence supporting this notion, as well as discuss novel abnormalities in mitochondrial function that appear to be related to ASD, and treatments that both target mitochondria and have evidence of usefulness in the treatment of ASD in clinical trials. A suspicion of the mitochondrion's involvement in ASD can be traced back to 1985 when lactic acidosis was noted in a subset of children with ASD. A large population-based study in 2007 confirmed this notion and found that a subset of children with ASD (â¼4%) could be diagnosed with a definite mitochondrial disease. Further studies suggested that children with ASD and mitochondrial disease may have certain characteristics such as fatigability, gastrointestinal disorders, unusual types of neurodevelopmental regression, seizures/epilepsy, and motor delay. Further research examining biomarkers of mitochondrial dysfunction and electron transport chain activity suggest that abnormalities of mitochondrial function could affect a much higher number of children with ASD, perhaps up to 80%. Recent research has identified a type of dysfunction of mitochondria in which the activity of the electron transport chain is significantly increased. This novel type of mitochondrial dysfunction may be associated with environmental exposures and neurodevelopmental regression. Several treatments that target mitochondria appear to have evidence for use in children with ASD, including cofactors such as L-Carnitine and the ketogenic diet. Although the understanding of the involvement of mitochondria in ASD is evolving, the mitochondrion is clearly a novel molecular target which can be helpful in understanding the etiology of ASD and treatments that may improve function of children with ASD.
Subject(s)
Autism Spectrum Disorder , Diet, Ketogenic , Dietary Supplements , Mitochondrial Diseases , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/therapy , Child , Humans , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/therapyABSTRACT
Epilepsy is frequently a severe and sinister symptom in primary mitochondrial diseases, a group of more than 350 different genetic disorders characterized by mitochondrial dysfunction and extreme clinical and biochemical heterogeneity. Mitochondrial epilepsy is notoriously difficult to manage, principally because the vast majority of primary mitochondrial diseases currently lack effective therapies. Treating the underlying mitochondrial disorder is likely to be a more effective strategy than using traditional antiepileptic drugs. This review, initially presented at the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures at the Francis Crick Institute in London, summarizes the currently available and emerging therapies for mitochondrial epilepsy. Potentially treatable mitochondrial diseases include disorders of coenzyme Q10 biosynthesis and a group of mitochondrial respiratory chain complex I subunit and assembly factor defects that respond to riboflavin (vitamin B2). Approaches that have been adopted in actively recruiting clinical trials include redox modulation, harnessing mitochondrial biogenesis, using rapamycin to target mitophagy, nucleoside supplementation, and gene and cell therapies. Most of the clinical trials are at an early stage (Phase 1 or 2) and none of the currently active trials is specifically targeting mitochondrial epilepsy. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".
Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/etiology , Epilepsy/therapy , Mitochondrial Diseases/complications , Mitochondrial Diseases/therapy , Animals , Anticonvulsants/pharmacology , Antioxidants/therapeutic use , Humans , Organelle Biogenesis , Status Epilepticus/drug therapyABSTRACT
Cerebellar ataxia is a hallmark of coenzyme Q10 (CoQ10) deficiency associated with COQ8A mutations. We present four patients, one with novel COQ8A pathogenic variants all with early, prominent handwriting impairment, dystonia and only mild ataxia. To better define the phenotypic spectrum and course of COQ8A disease, we review the clinical presentation and evolution in 47 reported cases. Individuals with COQ8A mutation display great clinical variability and unpredictable responses to CoQ10 supplementation. Onset is typically during infancy or childhood with ataxic features associated with developmental delay or regression. When disease onset is later in life, first symptoms can include: incoordination, epilepsy, tremor, and deterioration of writing. The natural history is characterized by a progression to a multisystem brain disease dominated by ataxia, with disease severity inversely correlated with age at onset. Six previously reported cases share with ours, a clinical phenotype characterized by slowly progressive or static writing difficulties, focal dystonia, and speech disorder, with only minimal ataxia. The combination of writing difficulty, dystonia and ataxia is a distinctive constellation that is reminiscent of a previously described clinical entity called Dystonia Ataxia Syndrome (DYTCA) and is an important clinical indicator of COQ8A mutations, even when ataxia is mild or absent.
Subject(s)
Ataxia , Disease Progression , Dystonic Disorders , Handwriting , Heterozygote , Mitochondrial Diseases , Mitochondrial Proteins/genetics , Muscle Weakness , Ubiquinone/deficiency , Adult , Ataxia/complications , Ataxia/epidemiology , Ataxia/etiology , Ataxia/genetics , Ataxia/physiopathology , Child , Dystonic Disorders/epidemiology , Dystonic Disorders/etiology , Dystonic Disorders/genetics , Dystonic Disorders/physiopathology , Female , Humans , Male , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Muscle Weakness/complications , Muscle Weakness/epidemiology , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Ubiquinone/genetics , Young AdultABSTRACT
COQ4 mutations have recently been shown to cause a broad spectrum of mitochondrial disorders in association with CoQ10 deficiency. Herein, we report the clinical phenotype, in silico and biochemical analyses, and intervention for a novel c.370 G > A (p.G124S) COQ4 mutation in a Chinese family. This mutation is exclusively present in the East Asian population (allele frequency of ~0.001). The homozygous mutation caused CoQ10 deficiency-associated Leigh syndrome with an onset at 1-2 months of age, presenting as respiratory distress, lactic acidosis, dystonia, seizures, failure to thrive, and detectable lesions in the midbrain and basal ganglia. No renal impairment was involved. The levels of CoQ10 and mitochondrial respiratory chain complex (C) II + III activity were clearly lower in cultured fibroblasts derived from the patient than in those from unaffected carriers; the decreased CII + III activity could be increased by CoQ10 treatment. Follow-up studies suggested that our patient benefitted from the oral supplementation of CoQ10, which allowed her to maintain a relatively stable health status. Based on the genetic testing, preimplantation and prenatal diagnoses were performed, confirming that the next offspring of this family was unaffected. Our cases expand the phenotypic spectrum of COQ4 mutations and the genotypic spectrum of Leigh syndrome.
Subject(s)
Ataxia/genetics , Genetic Testing , Leigh Disease/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Ubiquinone/deficiency , Asian People/genetics , Ataxia/complications , Child, Preschool , Computer Simulation , Female , Fibroblasts/metabolism , Heterozygote , Homozygote , Humans , Infant , Leigh Disease/complications , Leigh Disease/physiopathology , Male , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/complications , Muscle Weakness/complications , Mutation , Phenotype , Ubiquinone/genetics , Ubiquinone/pharmacokineticsABSTRACT
BACKGROUND: COQ2 mutations cause a rare infantile multisystemic disease with heterogeneous clinical features. Promising results have been reported in response to Coenzyme Q10 treatment, especially for kidney involvement, but little is known about the long-term outcomes. METHODS: We report four new patients from two families with the c.437GâA (p.Ser146Asn) mutation in COQ2 and the outcomes of two patients after long-term coenzyme Q10 treatment. RESULTS: Index cases from two families presented with vomiting, nephrotic range proteinuria, and diabetes in early infancy. These patients were diagnosed with coenzyme Q10 deficiency and died shortly after diagnosis. Siblings of the index cases later presented with neonatal diabetes and proteinuria and were diagnosed at the first day of life. Coenzyme Q10 treatment was started immediately. The siblings responded dramatically to coenzyme Q10 treatment with normalized glucose and proteinuria levels, but they developed refractory focal clonic seizures beginning at three months of life that progressed to encephalopathy. CONCLUSIONS: In our cohort with CoQ10 deficiency, neurological involvement did not improve with oral coenzyme Q10 treatment despite the initial recovery from the diabetes and nephrotic syndrome.
Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Ataxia/diet therapy , Ataxia/genetics , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/genetics , Muscle Weakness/diet therapy , Muscle Weakness/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Ataxia/complications , Ataxia/diagnostic imaging , Cohort Studies , Diabetes Mellitus/etiology , Family Health , Female , Humans , Infant , Kidney/pathology , Kidney/ultrastructure , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnostic imaging , Muscle Weakness/complications , Muscle Weakness/diagnostic imaging , Mutation/genetics , Proteinuria/etiology , Ubiquinone/genetics , Ubiquinone/therapeutic useABSTRACT
Familial Hypercholesterolemia (FH) is an autosomal co-dominant genetic disorder characterized by elevated low-density lipoprotein (LDL) cholesterol levels and increased risk for premature cardiovascular disease. Here, we examined FH pathophysiology in skin fibroblasts derived from FH patients harboring heterozygous mutations in the LDL-receptor. Fibroblasts from FH patients showed a reduced LDL-uptake associated with increased intracellular cholesterol levels and coenzyme Q10 (CoQ10) deficiency, suggesting dysregulation of the mevalonate pathway. Secondary CoQ10 deficiency was associated with mitochondrial depolarization and mitophagy activation in FH fibroblasts. Persistent mitophagy altered autophagy flux and induced inflammasome activation accompanied by increased production of cytokines by mutant cells. All the pathological alterations in FH fibroblasts were also reproduced in a human endothelial cell line by LDL-receptor gene silencing. Both increased intracellular cholesterol and mitochondrial dysfunction in FH fibroblasts were partially restored by CoQ10 supplementation. Dysregulated mevalonate pathway in FH, including increased expression of cholesterogenic enzymes and decreased expression of CoQ10 biosynthetic enzymes, was also corrected by CoQ10 treatment. Reduced CoQ10 content and mitochondrial dysfunction may play an important role in the pathophysiology of early atherosclerosis in FH. The diagnosis of CoQ10 deficiency and mitochondrial impairment in FH patients may also be important to establish early treatment with CoQ10.
Subject(s)
Ataxia/complications , Cholesterol/metabolism , Fibroblasts/pathology , Hyperlipoproteinemia Type II/complications , Mitochondrial Diseases/complications , Muscle Weakness/complications , Ubiquinone/deficiency , Ataxia/metabolism , Ataxia/pathology , Cells, Cultured , Fibroblasts/metabolism , Humans , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/pathology , Lipoproteins, LDL/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitophagy , Muscle Weakness/metabolism , Muscle Weakness/pathology , Reactive Oxygen Species/metabolism , Receptors, LDL/metabolism , Ubiquinone/metabolismABSTRACT
Nephrotic syndrome (NS), a frequent chronic kidney disease in children and young adults, is the most common phenotype associated with primary coenzyme Q10 (CoQ10) deficiency and is very responsive to CoQ10 supplementation, although the pathomechanism is not clear. Here, using a mouse model of CoQ deficiency-associated NS, we show that long-term oral CoQ10 supplementation prevents kidney failure by rescuing defects of sulfides oxidation and ameliorating oxidative stress, despite only incomplete normalization of kidney CoQ levels and lack of rescue of CoQ-dependent respiratory enzymes activities. Liver and kidney lipidomics, and urine metabolomics analyses, did not show CoQ metabolites. To further demonstrate that sulfides metabolism defects cause oxidative stress in CoQ deficiency, we show that silencing of sulfide quinone oxido-reductase (SQOR) in wild-type HeLa cells leads to similar increases of reactive oxygen species (ROS) observed in HeLa cells depleted of the CoQ biosynthesis regulatory protein COQ8A. While CoQ10 supplementation of COQ8A depleted cells decreases ROS and increases SQOR protein levels, knock-down of SQOR prevents CoQ10 antioxidant effects. We conclude that kidney failure in CoQ deficiency-associated NS is caused by oxidative stress mediated by impaired sulfides oxidation and propose that CoQ supplementation does not significantly increase the kidney pool of CoQ bound to the respiratory supercomplexes, but rather enhances the free pool of CoQ, which stabilizes SQOR protein levels rescuing oxidative stress.
Subject(s)
Antioxidants/pharmacology , Ataxia/drug therapy , Hydrogen Sulfide/metabolism , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Nephrotic Syndrome/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Alkyl and Aryl Transferases/genetics , Animals , Antioxidants/therapeutic use , Ataxia/complications , Ataxia/metabolism , Disease Models, Animal , HeLa Cells , Humans , Kidney/metabolism , Kidney/pathology , Metabolic Networks and Pathways/drug effects , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Muscle Weakness/complications , Muscle Weakness/metabolism , Nephrotic Syndrome/etiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Reactive Oxygen Species/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology , Ubiquinone/therapeutic useABSTRACT
BACKGROUND: Nephrotic syndrome can be caused by a subgroup of mitochondrial diseases classified as primary coenzyme Q10 (CoQ10) deficiency. Pathogenic COQ2 variants are a cause of primary CoQ10 deficiency and present with phenotypes ranging from isolated nephrotic syndrome to fatal multisystem disease. CASE-DIAGNOSIS/TREATMENT: We report three pediatric patients with COQ2 variants presenting with nephrotic syndrome. Two of these patients had normal leukocyte CoQ10 levels prior to treatment. Pathologic findings varied from mesangial sclerosis to focal segmental glomerulosclerosis, with all patients having abnormal appearing mitochondria on kidney biopsy. In two of the three patients treated with CoQ10 supplementation, the nephrotic syndrome resolved; and at follow-up, both have normal renal function and stable proteinuria. CONCLUSIONS: COQ2 nephropathy should be suspected in patients presenting with nephrotic syndrome, although less common than disease due to mutations in NPHS1, NPHS2, and WT1. The index of suspicion should remain high, and we suggest that providers consider genetic evaluation even in patients with normal leukocyte CoQ10 levels, as levels may be within normal range even with significant clinical disease. Early molecular diagnosis and specific treatment are essential in the management of this severe yet treatable condition.
Subject(s)
Alkyl and Aryl Transferases/genetics , Ataxia/drug therapy , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Nephrotic Syndrome/therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Ataxia/complications , Ataxia/diagnosis , Ataxia/genetics , Biopsy , Child , Child, Preschool , Genetic Testing , Humans , Kidney/pathology , Kidney Transplantation , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Muscle Weakness/complications , Muscle Weakness/diagnosis , Muscle Weakness/genetics , Nephrotic Syndrome/blood , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/etiology , Treatment Outcome , Ubiquinone/administration & dosage , Ubiquinone/geneticsSubject(s)
Biotin/therapeutic use , Diagnostic Errors , Hypothyroidism/diagnosis , Immunoassay , Mitochondrial Diseases/drug therapy , Thyroid Function Tests , Vitamin B Complex/therapeutic use , Biomarkers/blood , Biotin/immunology , Humans , Hypothyroidism/blood , Hypothyroidism/complications , Infant , Male , Mitochondrial Diseases/blood , Mitochondrial Diseases/complications , Vitamin B Complex/immunologyABSTRACT
BACKGROUND: A 7-month-old male infant was admitted because he was suffering from nephrotic syndrome, along with encephalomyopathy, hypertrophic cardiomyopathy, clinically suspected deafness and retinitis pigmentosa, and an elevated serum lactate level. METHODS: Coenzyme Q10 supplementation was started because of the clinical suspicion of primary CoQ10 deficiency. Despite intensive efforts, he passed away 4 weeks after admission. RESULTS: The results of genetic tests, available postmortem, explored two hitherto undescribed mutations in the PDSS2 gene. Both were located within the polyprenyl synthetase domain. Clinical exome sequencing revealed a heterozygous missense mutation in exon 3, and our in-house joint-analysis algorithm detected a heterozygous large 2923-bp deletion that affected the 5 prime end of exon 8. Other causative defects in the CoQ10 and infantile nephrosis-related genes examined were not found. A postmortem histological, immunohistochemical, and electron microscopic evaluation of the glomeruli revealed collapsing-sclerosing lesions consistent with diffuse mesangial sclerosis. The extrarenal alterations included hypertrophic cardiomyopathy and diffuse alveolar damage. A histological evaluation of the central nervous system and skeletal muscles did not demonstrate any obvious abnormality. CONCLUSIONS: Until now, the clinical features and the mutational status of 6 patients with a PDSS2 gene defect have been reported in the English literature. Here, we describe for the first time detailed kidney morphology features in a patient with nephrotic syndrome carrying mutations in the PDSS2 gene.
Subject(s)
Alkyl and Aryl Transferases/genetics , Ataxia/genetics , Kidney/pathology , Mitochondrial Diseases/genetics , Muscle Weakness/genetics , Nephrotic Syndrome/genetics , Sclerosis/genetics , Ubiquinone/deficiency , Ataxia/complications , Autopsy , Fatal Outcome , Genetic Testing/methods , Humans , Infant , Male , Mitochondrial Diseases/complications , Muscle Weakness/complications , Mutation , Nephrotic Syndrome/complications , Nephrotic Syndrome/etiology , Sclerosis/complications , Ubiquinone/analogs & derivatives , Ubiquinone/genetics , Ubiquinone/therapeutic useABSTRACT
Leukoencephalopathy with thalamus and brainstem involvement and high lactate (LTBL) is a recently described autosomal recessive mitochondrial disease characterized by early onset of neurological symptoms, a biphasic clinical course, and distinctive neuroimaging. Pathogenic variants in the EARS2 gene that encode for mitochondrial glutamyl-tRNA synthetase are responsible for LTBL. Here, we describe the clinical course of an infant diagnosed with an acute crisis of LTBL and severe liver disease. This article illustrates the utility of blood lactate quantification in addition to basic metabolic testing and brain imaging in a child with low tone and poor growth. In addition, this case demonstrates the utility of current genetic diagnostic testing, in lieu of more invasive procedures, in obtaining rapid answers in this very complicated group of disorders.
Subject(s)
Acidosis/diagnosis , Brain Stem/diagnostic imaging , Lactic Acid/blood , Leukoencephalopathies/diagnosis , Mitochondrial Diseases/diagnosis , Thalamus/diagnostic imaging , Acidosis/complications , Acidosis/therapy , Diagnosis, Differential , Glutamate-tRNA Ligase/genetics , Humans , Infant , Leukoencephalopathies/complications , Leukoencephalopathies/therapy , Liver Diseases/blood , Liver Diseases/complications , Liver Diseases/diagnostic imaging , Liver Diseases/therapy , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/therapyABSTRACT
In familial and sporadic multiple system atrophy (MSA) patients, deficiency of coenzyme Q10 (CoQ10) has been associated with mutations in COQ2, which encodes the second enzyme in the CoQ10 biosynthetic pathway. Cerebellar ataxia is the most common presentation of CoQ10 deficiency, suggesting that the cerebellum might be selectively vulnerable to low levels of CoQ10 To investigate whether CoQ10 deficiency represents a common feature in the brains of MSA patients independent of the presence of COQ2 mutations, we studied CoQ10 levels in postmortem brains of 12 MSA, 9 Parkinson disease (PD), 9 essential tremor (ET) patients, and 12 controls. We also assessed mitochondrial respiratory chain enzyme activities, oxidative stress, mitochondrial mass, and levels of enzymes involved in CoQ biosynthesis. Our studies revealed CoQ10 deficiency in MSA cerebellum, which was associated with impaired CoQ biosynthesis and increased oxidative stress in the absence of COQ2 mutations. The levels of CoQ10 in the cerebella of ET and PD patients were comparable or higher than in controls. These findings suggest that CoQ10 deficiency may contribute to the pathogenesis of MSA. Because no disease modifying therapies are currently available, increasing CoQ10 levels by supplementation or upregulation of its biosynthesis may represent a novel treatment strategy for MSA patients.
Subject(s)
Ataxia/metabolism , Cerebellum/metabolism , Mitochondrial Diseases/metabolism , Multiple System Atrophy/metabolism , Muscle Weakness/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Aged , Aged, 80 and over , Ataxia/complications , Ataxia/pathology , Case-Control Studies , Cerebellum/pathology , Female , Humans , Male , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Multiple System Atrophy/complications , Multiple System Atrophy/pathology , Muscle Weakness/complications , Muscle Weakness/pathology , Oxidative Stress/physiology , Ubiquinone/metabolismABSTRACT
Inherited ataxias are a group of heterogeneous disorders in children or adults but their genetic definition remains still undetermined in almost half of the patients. However, CoQ10 deficiency is a rare cause of cerebellar ataxia and ADCK3 is the most frequent gene associated with this defect. We herein report a 48 year old man, who presented with dysarthria and walking difficulties. Brain magnetic resonance imaging showed a marked cerebellar atrophy. Serum lactate was elevated. Tissues obtained by muscle and skin biopsies were studied for biochemical and genetic characterization. Skeletal muscle biochemistry revealed decreased activities of complexes I+III and II+III and a severe reduction of CoQ10 , while skin fibroblasts showed normal CoQ10 levels. A mild loss of maximal respiration capacity was also found by high-resolution respirometry. Molecular studies identified a novel homozygous deletion (c.504del_CT) in ADCK3, causing a premature stop codon. Western blot analysis revealed marked reduction of ADCK3 protein levels. Treatment with CoQ10 was started and, after 1 year follow-up, patient neurological condition slightly improved. This report suggests the importance of investigating mitochondrial function and, in particular, muscle CoQ10 levels, in patients with adult-onset cerebellar ataxia. Moreover, clinical stabilization by CoQ10 supplementation emphasizes the importance of an early diagnosis.