Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 443
1.
J Prim Care Community Health ; 14: 21501319231172697, 2023.
Article En | MEDLINE | ID: mdl-37162197

Bilateral lower extremity weakness and swelling can have several causes. Although often underdiagnosed, mitochondrial myopathy is more prevalent in the general population than more commonly suspected diseases, such as Guillain-Barre syndrome. The clinical manifestations of mitochondrial disease can be broadly classified into 3 categories: chronic progressive external ophthalmoplegia, skeletal muscle-central nervous system syndromes, or pure myopathy. Cardiac abnormalities occur in 30% to 32% of cases, mostly in the form of hypertrophic cardiomyopathy, dilated cardiomyopathy, or conduction abnormalities. We report a case of a 21-year-old student who developed bilateral lower limb weakness, pain, and swelling diagnosed with mitochondrial myopathy on muscle biopsy. Initial laboratory tests revealed elevated creatinine kinase, brain natriuretic peptide, troponin, myoglobin, and lactic acid and reduced serum bicarbonate. Cardiac workup revealed systolic heart failure with a reduced ejection fraction. Endomyocardial biopsy revealed punctate foci of lymphocytic myocarditis. However, cardiac magnetic resonance imaging did not reveal either myocarditis or an infiltrative cardiac disease. An extensive autoimmune and infection work-up was negative. A muscle biopsy from the patient's rectus femoris revealed scattered ragged-blue fibers (stained with NADH dehydrogenase), scattered ragged-red fibers on modified Gomori trichrome stain, and cytochrome-c oxidase negative fibers with increased perimysial and endomysial connective tissue, consistent with active and chronic primary mitochondrial myopathy. The patient was treated successfully with furosemide, metoprolol, and methylprednisolone. Adult-onset mitochondrial myopathy is a rare clinical disorder, and our experience stresses the importance of using an inter-disciplinary team approach to diagnose uncommon clinical disorders with widely variable multisystem involvement.


Mitochondrial Myopathies , Myocarditis , Ophthalmoplegia, Chronic Progressive External , Adult , Male , Humans , Young Adult , Myocarditis/pathology , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/pathology , Muscle, Skeletal/pathology , Ophthalmoplegia, Chronic Progressive External/diagnosis , Ophthalmoplegia, Chronic Progressive External/pathology , Lower Extremity/pathology
2.
Neuromuscul Disord ; 32(11-12): 923-930, 2022 12.
Article En | MEDLINE | ID: mdl-36428163

Mitochondrial diseases are a heterogeneous group of pathologies, caused by missense mutations, sporadic large-scale deletions of mitochondrial DNA (mtDNA) or mutations of nuclear maintenance genes. We report the case of a patient in whom extended muscle pathology, biochemical and genetic mtDNA analyses have proven to be essential to elucidate a unique asymmetrical myopathic presentation. From the age of 34 years on, the patient has presented with oculomotor disorders, right facial peripheral palsy and predominantly left upper limb muscle weakness and atrophy. By contrast, he displayed no motor weakness on the right hemi-body, and no sensory symptoms, cerebellar syndrome, hypoacusis, or parkinsonism. Cardiac function was normal. CK levels were elevated (671 UI/L). Electroneuromyography (ENMG) and muscle MRI showed diffuse myogenic alterations, more pronounced on the left side muscles. Biopsy of the left deltoid muscle showed multiple mitochondrial defects, whereas in the right deltoid, mitochondrial defects were much less marked. Extended mitochondrial biochemical and molecular workup revealed a unique mtDNA deletion, with a 63.4% heteroplasmy load in the left deltoid, versus 8.1% in the right one. This case demonstrates that, in mitochondrial myopathies, heteroplasmy levels may drastically vary for the same type of muscle, rising the hypothesis of a new pathophysiological mechanism explaining asymmetry in hereditary myopathies.


Heteroplasmy , Mitochondrial Myopathies , Male , Humans , Adult , Mitochondrial Myopathies/pathology , DNA, Mitochondrial/genetics , Muscular Atrophy/pathology , Muscles/pathology
3.
Int J Mol Sci ; 23(19)2022 Sep 20.
Article En | MEDLINE | ID: mdl-36232299

Thymidine kinase (TK2) deficiency causes mitochondrial DNA depletion syndrome. We aimed to report the clinical, biochemical, genetic, histopathological, and ultrastructural features of a cohort of paediatric patients with TK2 deficiency. Mitochondrial DNA was isolated from muscle biopsies to assess depletions and deletions. The TK2 genes were sequenced using Sanger sequencing from genomic DNA. All muscle biopsies presented ragged red fibres (RRFs), and the prevalence was greater in younger ages, along with an increase in succinate dehydrogenase (SDH) activity and cytochrome c oxidase (COX)-negative fibres. An endomysial inflammatory infiltrate was observed in younger patients and was accompanied by an overexpression of major histocompatibility complex type I (MHC I). The immunofluorescence study for complex I and IV showed a greater number of fibres than those that were visualized by COX staining. In the ultrastructural analysis, we found three major types of mitochondrial alterations, consisting of concentrically arranged lamellar cristae, electrodense granules, and intramitochondrial vacuoles. The pathological features in the muscle showed substantial differences in the youngest patients when compared with those that had a later onset of the disease. Additional ultrastructural features are described in the muscle biopsy, such as sarcomeric de-structuration in the youngest patients with a more severe phenotype.


Mitochondrial Myopathies , Thymidine Kinase/metabolism , DNA, Mitochondrial/analysis , DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Succinate Dehydrogenase , Thymidine Kinase/genetics
4.
Arch Oral Biol ; 140: 105448, 2022 Aug.
Article En | MEDLINE | ID: mdl-35550192

OBJECTIVE: This study aims to explore and compare the histopathology of upper cleft lip tissue in order to identify the abnormalities and orientation of muscle and collagen fibers in patients affected with non-syndromic cleft lip with or without cleft palate (NSCL±P) and normal controls. MATERIALS AND METHODS: Eight fresh lip tissues of consented patients with NSCL±P and two controls were fixed and stained with hematoxylin and eosin (H&E), Masson's trichrome (MT), and modified Gomori trichrome techniques. The images were captured and examined using imaging cellSens software (Olympus, Tokyo, Japan) and Mirax Scan (Carl Zeiss, Germany). The H&E stained tissues were analysed for muscle fiber size using image processing program (imageJ, USA). Histopathological appearance of epidermal and dermal layers including collagen orientation, as well as muscle fibers abnormalities were observed. RESULTS: Tissues stained with H&E exhibit pseudoepitheliomatous hyperplasia, epidermal and sebaceous glands hyperplasia. Morphometric analysis of muscle fibers showed the diameter was between 6.912 and 10.246 µm. Collagen fibers were densely packed in cleft tissue, but muscle fibers were sparse in MT stain. Modified Gomori trichrome stain revealed hypoplastic muscle with fibrosis, including ragged red fibers. CONCLUSION: Disoriented collagen fibers, significant sparse and disorganized orbicularis oris muscle fibers with classical myopathic appearances proved that cleft tissue had abnormal histology findings. These findings further support the mechanism of collagen and muscle fibers during embryonic development that causing cleft formation.


Cleft Lip , Cleft Palate , Mitochondrial Myopathies , Coloring Agents , Facial Muscles , Female , Fibrosis , Humans , Hyperplasia/pathology , Mitochondrial Myopathies/pathology , Muscle Fibers, Skeletal , Pregnancy
6.
Brain Pathol ; 32(4): e13038, 2022 07.
Article En | MEDLINE | ID: mdl-34806237

Two homoplasmic variants in tRNAGlu (m.14674T>C/G) are associated with reversible infantile respiratory chain deficiency. This study sought to further characterize the expression of the individual mitochondrial respiratory chain complexes and to describe the natural history of the disease. Seven patients from four families with mitochondrial myopathy associated with the homoplasmic m.14674T>C variant were investigated. All patients underwent skeletal muscle biopsy and mtDNA sequencing. Whole-genome sequencing was performed in one family. Western blot and immunohistochemical analyses were used to characterize the expression of the individual respiratory chain complexes. Patients presented with hypotonia and feeding difficulties within the first weeks or months of life, except for one patient who first showed symptoms at 4 years of age. Histopathological findings in muscle included lipid accumulation, numerous COX-deficient fibers, and mitochondrial proliferation. Ultrastructural abnormalities included enlarged mitochondria with concentric cristae and dense mitochondrial matrix. The m.14674T>C variant in MT-TE was identified in all patients. Immunohistochemistry and immunoblotting demonstrated pronounced deficiency of the complex I subunit NDUFB8. The expression of MTCO1, a complex IV subunit, was also decreased, but not to the same extent as NDUFB8. Longitudinal follow-up data demonstrated that not all features of the disorder are entirely transient, that the disease may be progressive, and that signs and symptoms of myopathy may develop during childhood. This study sheds new light on the involvement of complex I in reversible infantile respiratory chain deficiency, it shows that the disorder may be progressive, and that myopathy can develop without an infantile episode.


Cytochrome-c Oxidase Deficiency , Mitochondrial Myopathies , Cytochrome-c Oxidase Deficiency/genetics , Cytochrome-c Oxidase Deficiency/pathology , DNA, Mitochondrial/genetics , Electron Transport , Humans , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology , Muscle, Skeletal/pathology , Mutation
7.
Neuromuscul Disord ; 31(6): 551-557, 2021 06.
Article En | MEDLINE | ID: mdl-33832841

Reversible infantile respiratory chain deficiency is a severe neonatal mitochondrial myopathy that resolves spontaneously. It is caused by the homoplasmic m.14674T>C mtDNA mutation and additional nuclear variants in genes interacting with mt-tRNAGlu have been detected in some patients. We present detailed clinical, imaging, and muscle biopsy findings in a boy and a girl with neonatal hypotonia, feeding difficulties, lactic acidosis, and ragged red fibers. Both patients show fat replacement on muscle imaging, which was mild in the boy, but severe in the girl, affecting mostly the posterior leg muscles. In addition to the homoplasmic m.14674T>C, both patients carried heterozygous variants in QRSL1 (c. 686T>G; p.Val299Gly) and EARS2 (c.358C>T; p.Arg120Trp), respectively. It is very important to recognize the clinical and morphological signs of reversible infantile respiratory chain deficiency as patients should receive intensive supportive care in the first 6 months of life. Understanding the mechanism of the spontaneous recovery may lead to novel therapeutic perspectives in other mitochondrial diseases.


Mitochondrial Diseases/pathology , Muscle, Skeletal/pathology , Biopsy , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Myopathies/pathology , Muscle Hypotonia/etiology
8.
Mol Genet Metab ; 133(1): 1-7, 2021 05.
Article En | MEDLINE | ID: mdl-33744096

Mutations in the HADHB gene lead to Mitochondrial Trifunctional Protein (MTP) deficiency. MTP deficiency is a rare autosomal recessive disorder affecting long-chain fatty acid oxidation. Patients affected by MTP deficiency are unable to metabolize long-chain fatty-acids and suffer a variety of symptoms exacerbated during fasting. The three phenotypes associated with complete MTP deficiency are an early-onset cardiomyopathy and early death, an intermediate form with recurrent hypoketotic hypoglycemia and a sensorimotor neuropathy with episodic rhabdomyolysis with small amount of residual enzyme activities. This review aims to discuss the pathophysiological mechanisms and clinical manifestations of each phenotype, which appears different and linked to HADHB expression levels. Notably, the pathophysiology of the sensorimotor neuropathy is relatively unknown and we provide a hypothesis on the qualitative aspect of the role of acylcarnitine buildup in Schwann cells in MTP deficiency patients. We propose that acylcarnitine may exit the Schwann cell and alter membrane properties of nearby axons leading to axonal degeneration based on recent findings in different metabolic disorders.


Cardiomyopathies/genetics , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein/deficiency , Mitochondrial Trifunctional Protein/genetics , Nervous System Diseases/genetics , Rhabdomyolysis/genetics , Cardiomyopathies/pathology , Humans , Lipid Metabolism, Inborn Errors/pathology , Mitochondrial Myopathies/pathology , Mutation/genetics , Nervous System Diseases/pathology , Phenotype , Rhabdomyolysis/pathology
9.
J Inherit Metab Dis ; 44(4): 893-902, 2021 07.
Article En | MEDLINE | ID: mdl-33638202

Peripheral neuropathy is a known irreversible long-term complication of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) and mitochondrial trifunctional protein deficiency (MTPD), two inherited disorders of mitochondrial long-chain fatty acid oxidation. The underlying pathophysiology of neuropathy is still not fully understood. We report electrophysiological studies and neurological findings in a series of 8 LCHAD-deficient and 11 MTP-deficient patients. The median age at time of the study was 8.0 years (0.5-25 years). The overall prevalence of neuropathy was 58% with neuropathic symptoms being slightly more common in MTPD compared to LCHADD (70% vs 50%, respectively). Onset of neuropathy was significantly earlier in MTPD patients compared to LCHADD patients (median age at onset 4.7 vs 15.3 years, respectively, P = .047). In four patients, isolated peripheral neuropathy was the first and only presenting symptom, and in all four the diagnosis was missed by newborn screening. About half of the patients (45.5%) had a sensorimotor neuropathy, while 27.3% showed a pure motor form and another 27.3% an isolated sensory form. Despite early diagnosis by newborn screening and early initiation of therapy, peripheral neuropathy cannot be prevented in all patients with LCHADD/MTPD and has severe impact on the life of affected patients. Electrophysiology classifies LCHADD/MTPD neuropathy as axonal with secondary demyelination. A novel observation is that in patients with acute, fulminant onset of neuropathy, symptoms can be partly reversible. Further studies are needed to elucidate the underlying pathophysiology of axonal damage and possible therapeutic targets.


Cardiomyopathies/complications , Lipid Metabolism, Inborn Errors/complications , Mitochondrial Myopathies/complications , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/complications , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/etiology , Rhabdomyolysis/complications , Adolescent , Adult , Age Factors , Cardiomyopathies/diagnosis , Cardiomyopathies/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/pathology , Male , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/pathology , Nervous System Diseases/diagnosis , Nervous System Diseases/pathology , Peripheral Nervous System Diseases/pathology , Phenotype , Rhabdomyolysis/diagnosis , Rhabdomyolysis/pathology , Young Adult
10.
Ophthalmic Genet ; 42(1): 23-27, 2021 02.
Article En | MEDLINE | ID: mdl-33107778

Introduction: LCHADD causes retinopathy associated with low vision, visual field defects, nyctalopia and myopia. We report a retrospective long-term single-center study of 6 LCHADD patients trying to clarify if early diagnosis has an impact on the course and outcome of chorioretinal degeneration. Methods: Long-term follow-up of visual acuity and staging of chorioretinal degeneration by fundus photography, optical coherence tomography (OCT) and autofluorescence (AF) in all six patients. Three patients (2 m/1 f; age 8-14.8 years) were diagnosed by newborn screening, a single patient early within the first year of life and treated promptly while the other two (1 m/1 f; age 23-24 years) were diagnosed later after developing symptoms. All carried HADHA variants; five were homozygous for the common p.E510Q variant, in one from the symptomatically diagnosed group p.[E510Q]; [R291*] was detected. Results: All patients showed retinal alterations, but early diagnosis was associated with a milder phenotype and a longer preservation of visual function. Among symptomatic patients, only one showed mild retinal involvement at the time of diagnosis. Conclusion: Despite the small number our study suggests that early diagnosis does not prevent retinopathy but might contribute to a milder phenotype with retained good visual acuity over time. OCT and AF are reliable non-invasive diagnostic tools to estimate the progression of early-stage retinal changes in LCHADD patients.


Cardiomyopathies/pathology , Lipid Metabolism, Inborn Errors/pathology , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/deficiency , Mitochondrial Myopathies/pathology , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/pathology , Rhabdomyolysis/pathology , Visual Acuity , Adolescent , Adult , Cardiomyopathies/genetics , Child , Female , Humans , Lipid Metabolism, Inborn Errors/genetics , Male , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein/genetics , Multimodal Imaging , Nervous System Diseases/genetics , Prognosis , Retrospective Studies , Rhabdomyolysis/genetics , Young Adult
11.
J Med Genet ; 58(3): 155-167, 2021 03.
Article En | MEDLINE | ID: mdl-32439808

BACKGROUND: Mitochondria provide ATP through the process of oxidative phosphorylation, physically located in the inner mitochondrial membrane (IMM). The mitochondrial contact site and organising system (MICOS) complex is known as the 'mitoskeleton' due to its role in maintaining IMM architecture. APOO encodes MIC26, a component of MICOS, whose exact function in its maintenance or assembly has still not been completely elucidated. METHODS: We have studied a family in which the most affected subject presented progressive developmental delay, lactic acidosis, muscle weakness, hypotonia, weight loss, gastrointestinal and body temperature dysautonomia, repetitive infections, cognitive impairment and autistic behaviour. Other family members showed variable phenotype presentation. Whole exome sequencing was used to screen for pathological variants. Patient-derived skin fibroblasts were used to confirm the pathogenicity of the variant found in APOO. Knockout models in Drosophila melanogaster and Saccharomyces cerevisiae were employed to validate MIC26 involvement in MICOS assembly and mitochondrial function. RESULTS: A likely pathogenic c.350T>C transition was found in APOO predicting an I117T substitution in MIC26. The mutation caused impaired processing of the protein during import and faulty insertion into the IMM. This was associated with altered MICOS assembly and cristae junction disruption. The corresponding mutation in MIC26 or complete loss was associated with mitochondrial structural and functional deficiencies in yeast and D. melanogaster models. CONCLUSION: This is the first case of pathogenic mutation in APOO, causing altered MICOS assembly and neuromuscular impairment. MIC26 is involved in the assembly or stability of MICOS in humans, yeast and flies.


Apolipoproteins/genetics , Autistic Disorder/genetics , Cognitive Dysfunction/genetics , Membrane Proteins/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Animals , Autistic Disorder/pathology , Cognitive Dysfunction/pathology , Drosophila melanogaster/genetics , Fibroblasts/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Humans , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Mitochondrial Myopathies/epidemiology , Mitochondrial Myopathies/pathology , Protein Binding , Saccharomyces cerevisiae/genetics
12.
Muscle Nerve ; 63(3): 304-310, 2021 03.
Article En | MEDLINE | ID: mdl-33146414

BACKGROUND: The diagnosis of uncommon pediatric neuromuscular disease (NMD) is challenging due to genetic and phenotypic heterogeneity, yet is important to guide treatment, prognosis, and recurrence risk. Patients with diagnostically challenging presentations typically undergo extensive testing with variable molecular diagnostic yield. Given the advancement in next generation sequencing (NGS), we investigated the value of clinical whole exome sequencing (ES) in uncommon pediatric NMD. METHODS: A retrospective cohort study of 106 pediatric NMD patients with a combination of ES, chromosomal microarray (CMA), and candidate gene testing was completed at a large tertiary referral center. RESULTS: A molecular diagnosis was achieved in 37/79 (46%) patients with ES, 4/44 (9%) patients with CMA, and 15/74 (20%) patients with candidate gene testing. In 2/79 (3%) patients, a dual molecular diagnosis explaining the neuromuscular disease process was identified. A total of 42 patients (53%) who received ES remained without a molecular diagnosis at the conclusion of the study. CONCLUSIONS: Due to NGS, molecular diagnostic yield of rare neurological diseases is at an all-time high. We show that ES has a higher diagnostic rate compared to other genetic tests in a complex pediatric neuromuscular disease cohort and should be considered early in the diagnostic journey for select NMD patients with challenging presentations in which a clinical diagnosis is not evident.


Exome Sequencing , Neuromuscular Diseases/diagnosis , Adolescent , Biopsy , Child , Child, Preschool , Cohort Studies , Electromyography , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Microarray Analysis , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology , Molecular Diagnostic Techniques , Muscular Dystrophies/diagnosis , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Myopathy, Central Core/diagnosis , Myopathy, Central Core/genetics , Myopathy, Central Core/pathology , Myositis/diagnosis , Myositis/genetics , Myositis/pathology , Neural Conduction , Neuromuscular Diseases/genetics , Neuromuscular Diseases/pathology , Retrospective Studies , Sequence Analysis, DNA , Spinal Muscular Atrophies of Childhood/diagnosis , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/pathology , Spinocerebellar Ataxias/diagnosis , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology
13.
Biochim Biophys Acta Gen Subj ; 1865(3): 129834, 2021 03.
Article En | MEDLINE | ID: mdl-33358864

BACKGROUND: Mitochondria is a key organelle for energy production and cellular adaptive response to intracellular and extracellular stresses. Mitochondrial stress can be evoked by various stimuli such as metabolic stressors or pathogen infection, which may lead to expression of 'mitokines' such as growth differentiation factor 15 (GDF15). SCOPE OF REVIEW: This review summarizes the mechanism of GDF15 expression in response to organelle stress such as mitochondrial stress, and covers pathophysiological conditions or diseases that are associated with elevated GDF15 level. This review also illustrates the in vivo role of GDF15 expression in those stress conditions or diseases, and a potential of GDF15 as a therapeutic agent against metabolic disorders such as NASH. MAJOR CONCLUSIONS: Mitochondrial unfolded protein response (UPRmt) is a critical process to recover from mitochondrial stress. UPRmt can induce expression of secretory proteins that can exert systemic effects (mitokines) as well as mitochondrial chaperons. GDF15 can have either protective or detrimental systemic effects in response to mitochondrial stresses, suggesting its role as a mitokine. Mounting evidence shows that GDF15 is also induced by stresses of organelles other than mitochondria such as endoplasmic reticulum (ER). GDF15 level is increased in serum or tissue of mice and human subjects with metabolic diseases such as obesity or NASH. GDF15 can modulate metabolic features of those diseases. GENERAL SIGNIFICANCE: GDF15 play a role as an integrated stress response (ISR) beyond mitochondrial stress response. GDF15 is involved in the pathogenesis of metabolic diseases such as NASH, and also could be a candidate for therapeutic agent against those diseases.


Aging/genetics , Growth Differentiation Factor 15/genetics , Mitochondrial Myopathies/genetics , Muscular Atrophy/genetics , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics , Aging/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/therapeutic use , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Myopathies/metabolism , Mitochondrial Myopathies/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism , Obesity/pathology , Unfolded Protein Response
14.
Biochim Biophys Acta Gen Subj ; 1865(3): 129832, 2021 03.
Article En | MEDLINE | ID: mdl-33358866

BACKGROUND: Increasing evidence from pathological and biochemical investigations suggests that mitochondrial metabolic impairment and oxidative stress play a crucial role in the pathogenesis of mitochondrial diseases, such as mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome, and various neurodegenerative disorders. Recent advances in molecular imaging technology with positron emission tomography (PET) and functional magnetic resonance imaging (MRI) have accomplished a direct and non-invasive evaluation of the pathophysiological changes in living patients. SCOPE OF REVIEW: In this review, we focus on the latest achievements of molecular imaging for mitochondrial metabolism and oxidative stress in mitochondrial diseases and neurodegenerative disorders. MAJOR CONCLUSIONS: Molecular imaging with PET and MRI exhibited mitochondrial metabolic changes, such as enhanced glucose utilization with lactic acid fermentation, suppressed fatty acid metabolism, decreased TCA-cycle metabolism, impaired respiratory chain activity, and increased oxidative stress, in patients with MELAS syndrome. In addition, PET imaging clearly demonstrated enhanced cerebral oxidative stress in patients with Parkinson's disease or amyotrophic lateral sclerosis. The magnitude of oxidative stress correlated well with clinical severity in patients, indicating that oxidative stress based on mitochondrial dysfunction is associated with the neurodegenerative changes in these diseases. GENERAL SIGNIFICANCE: Molecular imaging is a promising tool to improve our knowledge regarding the pathogenesis of diseases associated with mitochondrial dysfunction and oxidative stress, and this would facilitate the development of potential antioxidants and mitochondrial therapies.


Acidosis, Lactic/diagnostic imaging , Brain Diseases/diagnostic imaging , MELAS Syndrome/diagnostic imaging , Mitochondria/metabolism , Mitochondrial Myopathies/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , Acidosis, Lactic/metabolism , Acidosis, Lactic/pathology , Brain Diseases/metabolism , Brain Diseases/pathology , Electron Transport/genetics , Fatty Acids/metabolism , Glucose/metabolism , Humans , Lactic Acid/metabolism , MELAS Syndrome/metabolism , MELAS Syndrome/pathology , Magnetic Resonance Imaging , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Myopathies/metabolism , Mitochondrial Myopathies/pathology , Molecular Imaging , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Positron-Emission Tomography , Severity of Illness Index
15.
Mitochondrion ; 56: 52-61, 2021 01.
Article En | MEDLINE | ID: mdl-33220502

Mitochondrial diseases (MIDs) involve multiple organs including peripheral nerves and skeletal muscle. Mitochondrial neuropathy (MN) and mitochondrial myopathy (MM) are commonly associated and linked at the neuromuscular junction (NMJ). Herein we review MN in connection with neurogenic features of MM, and pathological evidence for the involvement of the peripheral nerve and NMJ in MID patients traditionally assumed to have predominantly MM. MN is not uncommon, but still likely under-reported, and muscle biopsies of MM commonly exhibit neurogenic features. Pathological examination remains the gold standard to assess the nerve and muscle changes in patients with MIDs. Ultrastructural studies by electron microscopy are often necessary to fully characterize the pathology of mitochondrial cytopathy in MN and MM.


Mitochondrial Diseases/pathology , Mitochondrial Myopathies/pathology , Neuromuscular Junction/pathology , Peripheral Nerves/pathology , Animals , Biopsy , Diagnosis, Differential , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Myopathies/metabolism , Neuromuscular Junction/metabolism , Peripheral Nerves/metabolism
16.
Genes (Basel) ; 11(12)2020 12 19.
Article En | MEDLINE | ID: mdl-33352713

Primary mitochondrial myopathies (PMM) are a group of mitochondrial disorders characterized by a predominant skeletal muscle involvement. The aim of this study was to evaluate whether the biochemical profile determined by Fourier-transform infrared (FTIR) spectroscopic technique would allow to distinguish among patients affected by progressive external ophthalmoplegia (PEO), the most common PMM presentation, oculopharyngeal muscular dystrophy (OPMD), and healthy controls. Thirty-four participants were enrolled in the study. FTIR spectroscopy was found to be a sensitive and specific diagnostic marker for PEO. In particular, FTIR spectroscopy was able to distinguish PEO patients from those affected by OPMD, even in the presence of histological findings similar to mitochondrial myopathy. At the same time, FTIR spectroscopy differentiated single mtDNA deletion and mutations in POLG, the most common nuclear gene associated with mitochondrial diseases, with high sensitivity and specificity. In conclusion, our data suggest that FTIR spectroscopy is a valuable biodiagnostic tool for the differential diagnosis of PEO with a high ability to also distinguish between single mtDNA deletion and mutations in POLG gene based on specific metabolic transitions.


DNA, Mitochondrial/genetics , Deltoid Muscle/chemistry , Mitochondrial Myopathies/diagnosis , Spectroscopy, Fourier Transform Infrared , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Child , DNA Polymerase gamma/genetics , DNA, Mitochondrial/analysis , Deltoid Muscle/pathology , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology , Muscular Dystrophy, Oculopharyngeal/diagnosis , Muscular Dystrophy, Oculopharyngeal/pathology , Ophthalmoplegia, Chronic Progressive External/diagnosis , Ophthalmoplegia, Chronic Progressive External/pathology , Sensitivity and Specificity , Young Adult
17.
Mol Genet Metab ; 131(1-2): 90-97, 2020.
Article En | MEDLINE | ID: mdl-32928639

BACKGROUND: The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS: We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS: Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS: We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.


Cardiomyopathies/blood , Carnitine O-Palmitoyltransferase/deficiency , Carnitine/analogs & derivatives , Congenital Bone Marrow Failure Syndromes/blood , Lipid Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/blood , Mitochondrial Diseases/blood , Mitochondrial Myopathies/blood , Mitochondrial Trifunctional Protein/deficiency , Muscular Diseases/blood , Nervous System Diseases/blood , Rhabdomyolysis/blood , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/blood , Carbon-Carbon Double Bond Isomerases/genetics , Carbon-Carbon Double Bond Isomerases/metabolism , Cardiomyopathies/diet therapy , Cardiomyopathies/pathology , Cardiomyopathies/therapy , Carnitine/blood , Carnitine/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/blood , Congenital Bone Marrow Failure Syndromes/diet therapy , Congenital Bone Marrow Failure Syndromes/pathology , Congenital Bone Marrow Failure Syndromes/therapy , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Exercise Therapy , Fasting , Female , Humans , Lipid Metabolism, Inborn Errors/diet therapy , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/therapy , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/blood , Male , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/pathology , Mitochondrial Diseases/therapy , Mitochondrial Myopathies/diet therapy , Mitochondrial Myopathies/pathology , Mitochondrial Myopathies/therapy , Mitochondrial Trifunctional Protein/blood , Muscular Diseases/diet therapy , Muscular Diseases/pathology , Muscular Diseases/therapy , Nervous System Diseases/diet therapy , Nervous System Diseases/pathology , Nervous System Diseases/therapy , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Rhabdomyolysis/diet therapy , Rhabdomyolysis/pathology , Rhabdomyolysis/therapy
18.
J Am Heart Assoc ; 9(18): e017091, 2020 09 15.
Article En | MEDLINE | ID: mdl-32892688

Background People with chronic heart failure (CHF) experience severe skeletal muscle dysfunction, characterized by mitochondrial abnormalities, which exacerbates the primary symptom of exercise intolerance. However, the molecular triggers and characteristics underlying mitochondrial abnormalities caused by CHF remain poorly understood. Methods and Results We recruited 28 patients with CHF caused by reduced ejection fraction and 9 controls. We simultaneously biopsied skeletal muscle from the pectoralis major in the upper limb and from the vastus lateralis in the lower limb. We phenotyped mitochondrial function in permeabilized myofibers from both sites and followed this by complete RNA sequencing to identify novel molecular abnormalities in CHF skeletal muscle. Patients with CHF presented with upper and lower limb skeletal muscle impairments to mitochondrial function that were of a similar deficit and indicative of a myopathy. Mitochondrial abnormalities were strongly correlated to symptoms. Further RNA sequencing revealed a unique transcriptome signature in CHF skeletal muscle characterized by a novel triad of differentially expressed genes related to deficits in energy metabolism including adenosine monophosphate deaminase 3, pyridine nucleotide-disulphide oxidoreductase domain 2, and lactate dehydrogenase C. Conclusions Our data suggest an upper and lower limb metabolic myopathy that is characterized by a unique transcriptome signature in skeletal muscle of humans with CHF.


Heart Failure/metabolism , Mitochondrial Myopathies/metabolism , Transcriptome , Aged , Biopsy , Case-Control Studies , Female , Heart Failure/diagnosis , Humans , Male , Mitochondria, Muscle/metabolism , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Sequence Analysis, RNA
19.
J Clin Lipidol ; 14(5): 646-648, 2020.
Article En | MEDLINE | ID: mdl-32800583

A 48-year-old man presented to our lipid clinic with statin intolerance and elevated serum creatine kinase levels, being affected by mitochondrial myopathy because of heteroplasmic mitochondrial DNA missense mutation in MTCO1 gene (m.7671T>A). He had just been treated with a coronary artery bypass 4 years before because of acute coronary syndrome, and he had consistently high levels of both low-density lipoprotein cholesterol and triglycerides. Dyslipidemia was successfully treated using 75 mg of alirocumab subcutaneously every 2 weeks, 10 mg of ezetimibe daily, 2 g of marine omega-3 fatty acids daily, and 145 mg of micronized fenofibrate every 2 days. Although muscle weakness persisted, myalgia did not reoccur and serum creatine kinase levels remained almost stable over the time.


Antibodies, Monoclonal, Humanized/therapeutic use , Hyperlipidemias/drug therapy , Mitochondrial Myopathies/drug therapy , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Male , Middle Aged , Mitochondrial Myopathies/metabolism , Mitochondrial Myopathies/pathology , Mutation, Missense , Prognosis
20.
Mol Genet Genomic Med ; 8(10): e1427, 2020 10.
Article En | MEDLINE | ID: mdl-32749073

BACKGROUND: Mitochondrial DNA depletion syndrome (MTDPS) is part of a group of mitochondrial diseases characterized by a reduction in mitochondrial DNA copy number. Most MTDPS is caused by mutations in genes that disrupt deoxyribonucleotide metabolism. METHODS: We performed the whole-exome sequencing of a hepato-encephalopathy patient with MTDPS and functional analyses to determine the clinical significance of the identified variant. RESULTS: Here, whole-exome sequencing of a patient presenting with hepato-encephalopathy and MTDPS identified a novel homozygous frameshift variant, c.13_29del (p.Trp6Profs*71) in MICOS13. MICOS13 (also known as QIL1, MIC13, or C19orf70) is a component of the MICOS complex, which plays crucial roles in the maintenance of cristae junctions at the mitochondrial inner membrane. We found loss of MICOS13 protein and fewer cristae structures in the mitochondria of fibroblasts derived from the patient. Stable expression of a wild-type MICOS13 cDNA in the patients fibroblasts using a lentivirus system rescued mitochondrial respiratory chain complex deficiencies. CONCLUSION: Our findings suggest that the novel c.13_29del (p.Trp6Profs*71) MICOS13 variant causes hepato-encephalopathy with MTDPS. We propose that MICOS13 is classified as the cause of MTDPS.


Membrane Proteins/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Proteins/genetics , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Frameshift Mutation , Homozygote , Humans , Infant , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Myopathies/pathology , Mitochondrial Proteins/metabolism
...