Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468672

ABSTRACT

The pathogenesis of chemotherapy-induced peripheral neuropathy (CIPN) is poorly understood. Here, we report that the CIPN-causing drug bortezomib (Bort) promotes delta 2 tubulin (D2) accumulation while affecting microtubule stability and dynamics in sensory neurons in vitro and in vivo and that the accumulation of D2 is predominant in unmyelinated fibers and a hallmark of bortezomib-induced peripheral neuropathy (BIPN) in humans. Furthermore, while D2 overexpression was sufficient to cause axonopathy and inhibit mitochondria motility, reduction of D2 levels alleviated both axonal degeneration and the loss of mitochondria motility induced by Bort. Together, our data demonstrate that Bort, a compound structurally unrelated to tubulin poisons, affects the tubulin cytoskeleton in sensory neurons in vitro, in vivo, and in human tissue, indicating that the pathogenic mechanisms of seemingly unrelated CIPN drugs may converge on tubulin damage. The results reveal a previously unrecognized pathogenic role for D2 in BIPN that may occur through altered regulation of mitochondria motility.


Subject(s)
Bortezomib/adverse effects , Neoplasms/drug therapy , Peripheral Nervous System Diseases/genetics , Tubulin/genetics , Animals , Antineoplastic Agents/adverse effects , Axons/drug effects , Axons/pathology , Disease Models, Animal , Drosophila melanogaster/genetics , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Larva/drug effects , Larva/genetics , Microtubules/drug effects , Microtubules/genetics , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Neoplasms/genetics , Neoplasms/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Zebrafish/genetics
2.
Mult Scler ; 29(7): 892-897, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37227101

ABSTRACT

Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune disease primarily affecting the optic nerves and spinal cord, which is usually associated with anti-aquaporin-4 antibodies. Here, we present two individuals who were negative for anti-aquaporin-4 antibodies and were initially diagnosed with seronegative NMOSD. Each patient's clinical course and radiographic features raised suspicion for an alternative disease process. Both individuals were found to have pathogenic variants of MT-ND5, encoding subunit 5 of mitochondrial complex I, ultimately leading to a revised diagnosis of a primary mitochondrial disorder. These cases illustrate the importance of biochemical and genetic testing in atypical cases of NMOSD.


Subject(s)
Mitochondrial Diseases , Neuromyelitis Optica , Humans , Aquaporin 4 , Autoantibodies , Magnetic Resonance Imaging , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Diagnostic Errors
3.
J Inherit Metab Dis ; 45(5): 996-1012, 2022 09.
Article in English | MEDLINE | ID: mdl-35621276

ABSTRACT

Mitochondrial complex V plays an important role in oxidative phosphorylation by catalyzing the generation of ATP. Most complex V subunits are nuclear encoded and not yet associated with recognized Mendelian disorders. Using exome sequencing, we identified a rare homozygous splice variant (c.87+3A>G) in ATP5PO, the complex V subunit which encodes the oligomycin sensitivity conferring protein, in three individuals from two unrelated families, with clinical suspicion of a mitochondrial disorder. These individuals had a similar, severe infantile and often lethal multi-systemic disorder that included hypotonia, developmental delay, hypertrophic cardiomyopathy, progressive epileptic encephalopathy, progressive cerebral atrophy, and white matter abnormalities on brain MRI consistent with Leigh syndrome. cDNA studies showed a predominant shortened transcript with skipping of exon 2 and low levels of the normal full-length transcript. Fibroblasts from the affected individuals demonstrated decreased ATP5PO protein, defective assembly of complex V with markedly reduced amounts of peripheral stalk proteins, and complex V hydrolytic activity. Further, expression of human ATP5PO cDNA without exon 2 (hATP5PO-∆ex2) in yeast cells deleted for yATP5 (ATP5PO homolog) was unable to rescue growth on media which requires oxidative phosphorylation when compared to the wild type construct (hATP5PO-WT), indicating that exon 2 deletion leads to a non-functional protein. Collectively, our findings support the pathogenicity of the ATP5PO c.87+3A>G variant, which significantly reduces but does not eliminate complex V activity. These data along with the recent report of an affected individual with ATP5PO variants, add to the evidence that rare biallelic variants in ATP5PO result in defective complex V assembly, function and are associated with Leigh syndrome.


Subject(s)
Brain Diseases , Leigh Disease , Mitochondrial Proton-Translocating ATPases , Brain Diseases/metabolism , DNA, Complementary/metabolism , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mutation , Proteins/metabolism
4.
Brain ; 144(9): 2696-2708, 2021 10 22.
Article in English | MEDLINE | ID: mdl-33856027

ABSTRACT

Many patients with SARS-CoV-2 infection develop neurological signs and symptoms; although, to date, little evidence exists that primary infection of the brain is a significant contributing factor. We present the clinical, neuropathological and molecular findings of 41 consecutive patients with SARS-CoV-2 infections who died and underwent autopsy in our medical centre. The mean age was 74 years (38-97 years), 27 patients (66%) were male and 34 (83%) were of Hispanic/Latinx ethnicity. Twenty-four patients (59%) were admitted to the intensive care unit. Hospital-associated complications were common, including eight patients (20%) with deep vein thrombosis/pulmonary embolism, seven (17%) with acute kidney injury requiring dialysis and 10 (24%) with positive blood cultures during admission. Eight (20%) patients died within 24 h of hospital admission, while 11 (27%) died more than 4 weeks after hospital admission. Neuropathological examination of 20-30 areas from each brain revealed hypoxic/ischaemic changes in all brains, both global and focal; large and small infarcts, many of which appeared haemorrhagic; and microglial activation with microglial nodules accompanied by neuronophagia, most prominently in the brainstem. We observed sparse T lymphocyte accumulation in either perivascular regions or in the brain parenchyma. Many brains contained atherosclerosis of large arteries and arteriolosclerosis, although none showed evidence of vasculitis. Eighteen patients (44%) exhibited pathologies of neurodegenerative diseases, which was not unexpected given the age range of our patients. We examined multiple fresh frozen and fixed tissues from 28 brains for the presence of viral RNA and protein, using quantitative reverse-transcriptase PCR, RNAscope® and immunocytochemistry with primers, probes and antibodies directed against the spike and nucleocapsid regions. The PCR analysis revealed low to very low, but detectable, viral RNA levels in the majority of brains, although they were far lower than those in the nasal epithelia. RNAscope® and immunocytochemistry failed to detect viral RNA or protein in brains. Our findings indicate that the levels of detectable virus in coronavirus disease 2019 brains are very low and do not correlate with the histopathological alterations. These findings suggest that microglial activation, microglial nodules and neuronophagia, observed in the majority of brains, do not result from direct viral infection of brain parenchyma, but more likely from systemic inflammation, perhaps with synergistic contribution from hypoxia/ischaemia. Further studies are needed to define whether these pathologies, if present in patients who survive coronavirus disease 2019, might contribute to chronic neurological problems.


Subject(s)
Brain Infarction/pathology , Brain/pathology , COVID-19/pathology , Hypoxia-Ischemia, Brain/pathology , Intracranial Hemorrhages/pathology , Acute Kidney Injury/complications , Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Adult , Aged , Aged, 80 and over , Bacteremia/complications , Brain/metabolism , Brain Infarction/complications , COVID-19/complications , COVID-19/physiopathology , Coronavirus Nucleocapsid Proteins/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/complications , Inflammation , Intensive Care Units , Intracranial Hemorrhages/complications , Male , Microglia/pathology , Middle Aged , Neurons/pathology , Phagocytosis , Phosphoproteins/metabolism , Pulmonary Embolism/complications , Pulmonary Embolism/physiopathology , RNA, Viral/metabolism , Renal Dialysis , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Survival Rate , T-Lymphocytes/pathology , Venous Thrombosis/complications , Venous Thrombosis/physiopathology
5.
Hum Mol Genet ; 28(15): 2486-2500, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31009944

ABSTRACT

Mutations in LMNA encoding lamin A/C and EMD encoding emerin cause cardiomyopathy and muscular dystrophy. Lmna null mice develop these disorders and have a lifespan of 7-8 weeks. Emd null mice show no overt pathology and have normal skeletal muscle but with regeneration defects. We generated mice with germline deletions of both Lmna and Emd to determine the effects of combined loss of the encoded proteins. Mice without lamin A/C and emerin are born at the expected Mendelian ratio, are grossly normal at birth but have shorter lifespans than those lacking only lamin A/C. However, there are no major differences between these mice with regards to left ventricular function, heart ultrastructure or electrocardiographic parameters except for slower heart rates in the mice lacking both lamin A/C and emerin. Skeletal muscle is similarly affected in both of these mice. Lmna+/- mice also lacking emerin live to at least 1 year and have no significant differences in growth, heart or skeletal muscle compared to Lmna+/- mice. Deletion of the mouse gene encoding lamina-associated protein 1 leads to prenatal death; however, mice with heterozygous deletion of this gene lacking both lamin A/C and emerin are born at the expected Mendelian ratio but had a shorter lifespan than those only lacking lamin A/C and emerin. These results show that mice with combined deficiencies of three interacting nuclear envelope proteins have normal embryonic development and that early postnatal defects are primarily driven by loss of lamin A/C or lamina-associated polypeptide 1 rather than emerin.


Subject(s)
Carrier Proteins/genetics , Heart/physiopathology , Lamin Type A/genetics , Membrane Proteins/genetics , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Nuclear Proteins/genetics , Animals , Animals, Newborn , Disease Models, Animal , Female , Haploinsufficiency , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Myocardium/metabolism , Myocardium/pathology
6.
Hum Mol Genet ; 28(2): 209-219, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30260394

ABSTRACT

X-linked scapuloperoneal myopathy (X-SM), one of Four-and-a-half LIM 1 (FHL1) related diseases, is an adult-onset slowly progressive myopathy, often associated with cardiomyopathy. We previously generated a knock-in mouse model that has the same mutation (c.365 G > C, p.W122S) as human X-SM patients. The mutant male mouse developed late-onset slowly progressive myopathy without cardiomyopathy. In this study, we observed that heterozygous (Het) and homozygous (Homo) female mice did not show alterations of skeletal muscle function or histology. In contrast, 20-month-old mutant female mice showed signs of cardiomyopathy on echocardiograms with increased systolic diameter [wild-type (WT): 2.74 ± 0.22 mm, mean ± standard deviation (SD); Het: 3.13 ± 0.11 mm, P < 0.01; Homo: 3.08 ± 0.37 mm, P < 0.05) and lower fractional shortening (WT: 31.1 ± 4.4%, mean ± SD; Het: 22.7 ± 2.5%, P < 0.01; Homo: 22.4 ± 6.9%, P < 0.01]. Histological analysis of cardiac muscle revealed frequent extraordinarily large rectangular nuclei in mutant female mice that were also observed in human cardiac muscle from X-SM patients. Western blot demonstrated decreased Fhl1 protein levels in cardiac muscle, but not in skeletal muscle, of Homo mutant female mice. Proteomic analysis of cardiac muscle from 20-month-old Homo mutant female mice indicated abnormalities of the integrin signaling pathway (ISP) in association with cardiac dysfunction. The ISP dysregulation was further supported by altered levels of a subunit of the ISP downstream effectors Arpc1a in Fhl1 mutant mice and ARPC1A in X-SM patient muscles. This study reveals the first mouse model of FHL1-related cardiomyopathy and implicates ISP dysregulation in the pathogenesis of FHL1 myopathy.


Subject(s)
Actins/metabolism , Cardiomyopathies/genetics , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Animals , Body Composition , Body Weight , Cardiomyopathies/pathology , Echocardiography , Female , Heterozygote , Homozygote , Male , Mice , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation, Missense , Myocardium/pathology , Phenotype , Proteomics , Signal Transduction
7.
J Neurol Neurosurg Psychiatry ; 92(11): 1186-1196, 2021 11.
Article in English | MEDLINE | ID: mdl-34103343

ABSTRACT

BACKGROUND: We used a multimodal approach including detailed phenotyping, whole exome sequencing (WES) and candidate gene filters to diagnose rare neurological diseases in individuals referred by tertiary neurology centres. METHODS: WES was performed on 66 individuals with neurogenetic diseases using candidate gene filters and stringent algorithms for assessing sequence variants. Pathogenic or likely pathogenic missense variants were interpreted using in silico prediction tools, family segregation analysis, previous publications of disease association and relevant biological assays. RESULTS: Molecular diagnosis was achieved in 39% (n=26) including 59% of childhood-onset cases and 27% of late-onset cases. Overall, 37% (10/27) of myopathy, 41% (9/22) of neuropathy, 22% (2/9) of MND and 63% (5/8) of complex phenotypes were given genetic diagnosis. Twenty-seven disease-associated variants were identified including ten novel variants in FBXO38, LAMA2, MFN2, MYH7, PNPLA6, SH3TC2 and SPTLC1. Single-nucleotide variants (n=10) affected conserved residues within functional domains and previously identified mutation hot-spots. Established pathogenic variants (n=16) presented with atypical features, such as optic neuropathy in adult polyglucosan body disease, facial dysmorphism and skeletal anomalies in cerebrotendinous xanthomatosis, steroid-responsive weakness in congenital myasthenia syndrome 10. Potentially treatable rare diseases were diagnosed, improving the quality of life in some patients. CONCLUSIONS: Integrating deep phenotyping, gene filter algorithms and biological assays increased diagnostic yield of exome sequencing, identified novel pathogenic variants and extended phenotypes of difficult to diagnose rare neurogenetic disorders in an outpatient clinic setting.


Subject(s)
Exome Sequencing , Genetic Diseases, Inborn/diagnosis , Mutation , Nervous System Diseases/diagnosis , Rare Diseases/diagnosis , Adolescent , Adult , Aged , Genetic Diseases, Inborn/genetics , Humans , Middle Aged , Molecular Diagnostic Techniques , Nervous System Diseases/genetics , Pedigree , Phenotype , Rare Diseases/genetics , Young Adult
8.
Hum Mol Genet ; 27(19): 3305-3312, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29917077

ABSTRACT

Leigh syndrome is a frequent, heterogeneous pediatric presentation of mitochondrial oxidative phosphorylation (OXPHOS) disease, manifesting with psychomotor retardation and necrotizing lesions in brain deep gray matter. OXPHOS occurs at the inner mitochondrial membrane through the integrated activity of five protein complexes, of which complex V (CV) functions in a dimeric form to directly generate adenosine triphosphate (ATP). Mutations in several different structural CV subunits cause Leigh syndrome; however, dimerization defects have not been associated with human disease. We report four Leigh syndrome subjects from three unrelated Ashkenazi Jewish families harboring a homozygous splice-site mutation (c.87 + 1G>C) in a novel CV subunit disease gene, USMG5. The Ashkenazi population allele frequency is 0.57%. This mutation produces two USMG5 transcripts, wild-type and lacking exon 3. Fibroblasts from two Leigh syndrome probands had reduced wild-type USMG5 mRNA expression and undetectable protein. The mutation did not alter monomeric CV expression, but reduced both CV dimer expression and ATP synthesis rate. Rescue with wild-type USMG5 cDNA in proband fibroblasts restored USMG5 protein, increased CV dimerization and enhanced ATP production rate. These data demonstrate that a recurrent USMG5 splice-site founder mutation in the Ashkenazi Jewish population causes autosomal recessive Leigh syndrome by reduction of CV dimerization and ATP synthesis.


Subject(s)
Leigh Disease/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Adenosine Triphosphate/biosynthesis , Child , Child, Preschool , Dimerization , Exons/genetics , Founder Effect , Gene Frequency , Haplotypes , Humans , Infant , Infant, Newborn , Jews/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Male , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation , Oxidative Phosphorylation , RNA Splice Sites/genetics , Exome Sequencing
9.
Mol Genet Metab ; 131(4): 398-404, 2020 12.
Article in English | MEDLINE | ID: mdl-33279411

ABSTRACT

Genetic defects in mitochondrial DNA encoded tRNA genes impair mitochondrial translation with resultant defects in the mitochondrial respiratory chain and oxidative phosphorylation system. The phenotypic spectrum of disease seen in mitochondrial tRNA defects is variable and proving pathogenicity of new variants is challenging. Only three pathogenic variants have been described previously in the mitochondrial tRNATyr gene MT-TY, with the reported phenotypes consisting largely of adult onset myopathy and ptosis. We report a patient with a novel MT-TY acceptor stem variant m.5889A>G at high heteroplasmy in muscle, low in blood, and absent in the mother's blood. The phenotype consisted of a childhood-onset severe multi-system disorder characterized by a neurodegenerative course including ataxia and seizures, failure-to-thrive, combined myopathy and neuropathy, and hearing and vision loss. Brain imaging showed progressive atrophy and basal ganglia calcifications. Mitochondrial biomarkers lactate and GDF15 were increased. Functional studies showed a deficient activity of the respiratory chain enzyme complexes containing mtDNA-encoded subunits I, III and IV. There were decreased steady state levels of these mitochondrial complex proteins, and presence of incompletely assembled complex V forms in muscle. These changes are typical of a mitochondrial translational defect. These data support the pathogenicity of this novel variant. Careful review of variants in MT-TY additionally identified two other pathogenic variants, one likely pathogenic variant, nine variants of unknown significance, five likely benign and four benign variants.


Subject(s)
DNA, Mitochondrial/genetics , Muscular Diseases/genetics , RNA, Transfer/genetics , Tyrosine/genetics , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation/genetics , Oxidative Phosphorylation , Phenotype
10.
Acta Neuropathol ; 139(6): 1089-1104, 2020 06.
Article in English | MEDLINE | ID: mdl-32236737

ABSTRACT

RYR1 encodes the type 1 ryanodine receptor, an intracellular calcium release channel (RyR1) on the skeletal muscle sarcoplasmic reticulum (SR). Pathogenic RYR1 variations can destabilize RyR1 leading to calcium leak causing oxidative overload and myopathy. However, the effect of RyR1 leak has not been established in individuals with RYR1-related myopathies (RYR1-RM), a broad spectrum of rare neuromuscular disorders. We sought to determine whether RYR1-RM affected individuals exhibit pathologic, leaky RyR1 and whether variant location in the channel structure can predict pathogenicity. Skeletal muscle biopsies were obtained from 17 individuals with RYR1-RM. Mutant RyR1 from these individuals exhibited pathologic SR calcium leak and increased activity of calcium-activated proteases. The increased calcium leak and protease activity were normalized by ex-vivo treatment with S107, a RyR stabilizing Rycal molecule. Using the cryo-EM structure of RyR1 and a new dataset of > 2200 suspected RYR1-RM affected individuals we developed a method for assigning pathogenicity probabilities to RYR1 variants based on 3D co-localization of known pathogenic variants. This study provides the rationale for a clinical trial testing Rycals in RYR1-RM affected individuals and introduces a predictive tool for investigating the pathogenicity of RYR1 variants of uncertain significance.


Subject(s)
Calcium/metabolism , Muscular Diseases/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Cytoplasm/metabolism , Humans , Muscle, Skeletal/metabolism , Muscular Diseases/therapy , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism
11.
Hum Mol Genet ; 26(1): 65-78, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27798115

ABSTRACT

Lamina-associated polypeptide 1 (LAP1) is an integral protein of the inner nuclear membrane that has been implicated in striated muscle maintenance. Mutations in its gene have been linked to muscular dystrophy and cardiomyopathy. As germline deletion of the gene encoding LAP1 is perinatal lethal, we explored its potential role in myogenic differentiation and development by generating a conditional knockout mouse in which the protein is depleted from muscle progenitors at embryonic day 8.5 (Myf5-Lap1CKO mice). Although cultured myoblasts lacking LAP1 demonstrated defective terminal differentiation and altered expression of muscle regulatory factors, embryonic myogenesis and formation of skeletal muscle occurred in both mice with a Lap1 germline deletion and Myf5-Lap1CKO mice. However, skeletal muscle fibres were hypotrophic and their nuclei were morphologically abnormal with a wider perinuclear space than normal myonuclei. Myf5-Lap1CKO mouse skeletal muscle contained fewer satellite cells than normal and these cells had evidence of reduced myogenic potential. Abnormalities in signalling pathways required for postnatal hypertrophic growth were also observed in skeletal muscles of these mice. Our results demonstrate that early embryonic depletion of LAP1 does not impair myogenesis but that it is necessary for postnatal skeletal muscle growth.


Subject(s)
Carrier Proteins/physiology , Membrane Proteins/physiology , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscular Dystrophies/embryology , Myoblasts/cytology , Animals , Cell Differentiation , Cell Proliferation , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Myogenic Regulatory Factors
12.
Hum Mol Genet ; 24(3): 714-26, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274776

ABSTRACT

A member of the four-and-a-half-LIM (FHL) domain protein family, FHL1, is highly expressed in human adult skeletal and cardiac muscle. Mutations in FHL1 have been associated with diverse X-linked muscle diseases: scapuloperoneal (SP) myopathy, reducing body myopathy, X-linked myopathy with postural muscle atrophy, rigid spine syndrome (RSS) and Emery-Dreifuss muscular dystrophy. In 2008, we identified a missense mutation in the second LIM domain of FHL1 (c.365 G>C, p.W122S) in a family with SP myopathy. We generated a knock-in mouse model harboring the c.365 G>C Fhl1 mutation and investigated the effects of this mutation at three time points (3-5 months, 7-10 months and 18-20 months) in hemizygous male and heterozygous female mice. Survival was comparable in mutant and wild-type animals. We observed decreased forelimb strength and exercise capacity in adult hemizygous male mice starting from 7 to 10 months of age. Western blot analysis showed absence of Fhl1 in muscle at later stages. Thus, adult hemizygous male, but not heterozygous female, mice showed a slowly progressive phenotype similar to human patients with late-onset muscle weakness. In contrast to SP myopathy patients with the FHL1 W122S mutation, mutant mice did not manifest cytoplasmic inclusions (reducing bodies) in muscle. Because muscle weakness was evident prior to loss of Fhl1 protein and without reducing bodies, our findings indicate that loss of function is responsible for the myopathy in the Fhl1 W122S knock-in mice.


Subject(s)
Forelimb/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Emery-Dreifuss/pathology , Myocardium/pathology , Age of Onset , Animals , Disease Models, Animal , Female , Gene Knock-In Techniques , Hemizygote , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Emery-Dreifuss/epidemiology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Mutation, Missense
13.
Muscle Nerve ; 51(5): 767-72, 2015 May.
Article in English | MEDLINE | ID: mdl-25430424

ABSTRACT

INTRODUCTION: Nemaline myopathy (NM) is a congenital neuromuscular disorder often characterized by hypotonia, facial weakness, skeletal muscle weakness, and the presence of rods on muscle biopsy. A rare form of nemaline myopathy known as Amish Nemaline Myopathy has only been seen in a genetically isolated cohort of Old Order Amish patients who may additionally present with tremors in the first 2-3 months of life. METHODS: We describe an Hispanic male diagnosed with nemaline myopathy histopathologically and subsequently confirmed by next generation gene sequencing. RESULTS: Direct sequencing revealed that he is homozygous for a pathogenic nonsense variant c.323C>G (p.S108X) in exon 9 of the TNNT1 gene. CONCLUSIONS: This report describes a novel pathogenic variant in the TNNT1 gene and represents a nemaline myopathy-causing variant in the TNNT1 gene outside of the Old Order Amish and Dutch ancestry.


Subject(s)
Mutation/genetics , Myopathies, Nemaline/genetics , Troponin I/genetics , Biopsy , Child, Preschool , Exons/genetics , Hispanic or Latino/genetics , Homozygote , Humans , Male , Muscle, Skeletal/pathology , Myopathies, Nemaline/diagnosis , Pedigree
14.
Brain ; 137(Pt 5): 1337-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24727567

ABSTRACT

Balanced pools of deoxyribonucleoside triphosphate precursors are required for DNA replication, and alterations of this balance are relevant to human mitochondrial diseases including mitochondrial neurogastrointestinal encephalopathy. In this disease, autosomal recessive TYMP mutations cause severe reductions of thymidine phosphorylase activity; marked elevations of the pyrimidine nucleosides thymidine and deoxyuridine in plasma and tissues, and somatic multiple deletions, depletion and site-specific point mutations of mitochondrial DNA. Thymidine phosphorylase and uridine phosphorylase double knockout mice recapitulated several features of these patients including thymidine phosphorylase activity deficiency, elevated thymidine and deoxyuridine in tissues, mitochondrial DNA depletion, respiratory chain defects and white matter changes. However, in contrast to patients with this disease, mutant mice showed mitochondrial alterations only in the brain. To test the hypothesis that elevated levels of nucleotides cause unbalanced deoxyribonucleoside triphosphate pools and, in turn, pathogenic mitochondrial DNA instability, we have stressed double knockout mice with exogenous thymidine and deoxyuridine, and assessed clinical, neuroradiological, histological, molecular, and biochemical consequences. Mutant mice treated with exogenous thymidine and deoxyuridine showed reduced survival, body weight, and muscle strength, relative to untreated animals. Moreover, in treated mutants, leukoencephalopathy, a hallmark of the disease, was enhanced and the small intestine showed a reduction of smooth muscle cells and increased fibrosis. Levels of mitochondrial DNA were depleted not only in the brain but also in the small intestine, and deoxyribonucleoside triphosphate imbalance was observed in the brain. The relative proportion, rather than the absolute amount of deoxyribonucleoside triphosphate, was critical for mitochondrial DNA maintenance. Thus, our results demonstrate that stress of exogenous pyrimidine nucleosides enhances the mitochondrial phenotype of our knockout mice. Our mouse studies provide insights into the pathogenic role of thymidine and deoxyuridine imbalance in mitochondrial neurogastrointestinal encephalopathy and an excellent model to study new therapeutic approaches.


Subject(s)
Deoxyribonucleosides/adverse effects , Intestinal Pseudo-Obstruction/chemically induced , Intestinal Pseudo-Obstruction/genetics , Mitochondrial Encephalomyopathies/chemically induced , Mitochondrial Encephalomyopathies/genetics , Age Factors , Animals , Body Weight/drug effects , Body Weight/genetics , Brain/pathology , Deoxyribonucleosides/metabolism , Disease Models, Animal , Intestinal Pseudo-Obstruction/mortality , Intestinal Pseudo-Obstruction/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Mitochondrial Encephalomyopathies/mortality , Mitochondrial Encephalomyopathies/physiopathology , Motor Activity/drug effects , Muscle Strength/drug effects , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal , Ophthalmoplegia/congenital , Psychomotor Disorders/etiology , Psychomotor Disorders/genetics , Succinate Dehydrogenase/metabolism , Thymidine/adverse effects , Thymidine/metabolism , Thymidine Phosphorylase/deficiency , Uridine Phosphorylase/deficiency
15.
Muscle Nerve ; 50(2): 292-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24711008

ABSTRACT

INTRODUCTION: A 61-year-old woman with a 5-year history of progressive muscle weakness and atrophy had a muscle biopsy characterized by a combination of dystrophic features (necrotic fibers and endomysial fibrosis) and mitochondrial alterations [ragged-red, cytochrome c oxidase (COX)-negative fibers]. METHODS: Sequencing of the whole mtDNA, assessment of the mutation load in muscle and accessible nonmuscle tissues, and single fiber polymerase chain reaction. RESULTS: Muscle mitochondrial DNA (mtDNA) sequencing revealed a novel heteroplasmic mutation (m.4403G>A) in the gene (MTTM) that encodes tRNA(Met). The mutation was not present in accessible nonmuscle tissues from the patient or 2 asymptomatic sisters. CONCLUSIONS: The clinical features and muscle morphology in this patient are very similar to those described in a previous patient with a different mutation, also in MTTM, which suggests that mutations in this gene confer a distinctive "dystrophic" quality. This may be a diagnostic clue in patients with isolated mitochondrial myopathy.


Subject(s)
Dystonia/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics , RNA, Transfer/genetics , Dystonia/complications , Female , Humans , Middle Aged , Mitochondrial Myopathies/complications
16.
Brain ; 136(Pt 5): 1508-17, 2013 May.
Article in English | MEDLINE | ID: mdl-23543484

ABSTRACT

In 2001, we reported linkage of an autosomal dominant form of limb-girdle muscular dystrophy, limb-girdle muscular dystrophy 1F, to chromosome 7q32.1-32.2, but the identity of the mutant gene was elusive. Here, using a whole genome sequencing strategy, we identified the causative mutation of limb-girdle muscular dystrophy 1F, a heterozygous single nucleotide deletion (c.2771del) in the termination codon of transportin 3 (TNPO3). This gene is situated within the chromosomal region linked to the disease and encodes a nuclear membrane protein belonging to the importin beta family. TNPO3 transports serine/arginine-rich proteins into the nucleus, and has been identified as a key factor in the HIV-import process into the nucleus. The mutation is predicted to generate a 15-amino acid extension of the C-terminus of the protein, segregates with the clinical phenotype, and is absent in genomic sequence databases and a set of >200 control alleles. In skeletal muscle of affected individuals, expression of the mutant messenger RNA and histological abnormalities of nuclei and TNPO3 indicate altered TNPO3 function. Our results demonstrate that the TNPO3 mutation is the cause of limb-girdle muscular dystrophy 1F, expand our knowledge of the molecular basis of muscular dystrophies and bolster the importance of defects of nuclear envelope proteins as causes of inherited myopathies.


Subject(s)
Gene Deletion , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/genetics , beta Karyopherins/genetics , Adolescent , Adult , Aged , Base Sequence , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Muscular Dystrophies, Limb-Girdle/metabolism , Pedigree , beta Karyopherins/biosynthesis
17.
Circ Heart Fail ; 17(4): e011110, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38567527

ABSTRACT

BACKGROUND: Mutations in LMNA encoding nuclear envelope proteins lamin A/C cause dilated cardiomyopathy. Activation of the AKT/mTOR (RAC-α serine/threonine-protein kinase/mammalian target of rapamycin) pathway is implicated as a potential pathophysiologic mechanism. The aim of this study was to assess whether pharmacological inhibition of mTOR signaling has beneficial effects on heart function and prolongs survival in a mouse model of the disease, after onset of heart failure. METHODS: We treated male LmnaH222P/H222P mice, after the onset of heart failure, with placebo or either of 2 orally bioavailable mTOR inhibitors: everolimus or NV-20494, a rapamycin analog highly selective against mTORC1. We examined left ventricular remodeling, and the cell biological, biochemical, and histopathologic features of cardiomyopathy, potential drug toxicity, and survival. RESULTS: Everolimus treatment (n=17) significantly reduced left ventricular dilatation and increased contractility on echocardiography, with a 7% (P=0.018) reduction in left ventricular end-diastolic diameter and a 39% (P=0.0159) increase fractional shortening compared with placebo (n=17) after 6 weeks of treatment. NV-20494 treatment (n=15) yielded similar but more modest and nonsignificant changes. Neither drug prevented the development of cardiac fibrosis. Drug treatment reactivated suppressed autophagy and inhibited mTORC1 signaling in the heart, although everolimus was more potent. With regards to drug toxicity, everolimus alone led to a modest degree of glucose intolerance during glucose challenge. Everolimus (n=20) and NV-20494 (n=20) significantly prolonged median survival in LmnaH222P/H222P mice, by 9% (P=0.0348) and 11% (P=0.0206), respectively, compared with placebo (n=20). CONCLUSIONS: These results suggest that mTOR inhibitors may be beneficial in patients with cardiomyopathy caused by LMNA mutations and that further study is warranted.


Subject(s)
Cardiomyopathies , Drug-Related Side Effects and Adverse Reactions , Heart Failure , Mice , Humans , Male , Animals , Everolimus/pharmacology , Everolimus/therapeutic use , Lamin Type A/genetics , Lamin Type A/metabolism , MTOR Inhibitors , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Mutation , TOR Serine-Threonine Kinases , Mechanistic Target of Rapamycin Complex 1/genetics , Mammals/metabolism
18.
bioRxiv ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38659958

ABSTRACT

GDF15 (growth differentiation factor 15) is a marker of cellular energetic stress linked to physical-mental illness, aging, and mortality. However, questions remain about its dynamic properties and measurability in human biofluids other than blood. Here, we examine the natural dynamics and psychobiological regulation of plasma and saliva GDF15 in four human studies representing 4,749 samples from 188 individuals. We show that GDF15 protein is detectable in saliva (8% of plasma concentration), likely produced by salivary glands secretory duct cells. Using a brief laboratory socio-evaluative stressor paradigm, we find that psychosocial stress increases plasma (+3.5-5.9%) and saliva GDF15 (+43%) with distinct kinetics, within minutes. Moreover, saliva GDF15 exhibits a robust awakening response, declining by ~40-89% within 30-45 minutes from its peak level at the time of waking up. Clinically, individuals with genetic mitochondrial OxPhos diseases show elevated baseline plasma and saliva GDF15, and post-stress GDF15 levels in both biofluids correlate with multi-system disease severity, exercise intolerance, and the subjective experience of fatigue. Taken together, our data establish that saliva GDF15 is dynamic, sensitive to psychological states, a clinically relevant endocrine marker of mitochondrial diseases. These findings also point to a shared psychobiological pathway integrating metabolic and mental stress.

19.
Neurobiol Dis ; 54: 349-61, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23333625

ABSTRACT

Autism spectrum disorder (ASD) consists of a group of complex developmental disabilities characterized by impaired social interactions, deficits in communication and repetitive behavior. Multiple lines of evidence implicate mitochondrial dysfunction in ASD. In postmortem BA21 temporal cortex, a region that exhibits synaptic pathology in ASD, we found that compared to controls, ASD patients exhibited altered protein levels of mitochondria respiratory chain protein complexes, decreased Complex I and IV activities, decreased mitochondrial antioxidant enzyme SOD2, and greater oxidative DNA damage. Mitochondrial membrane mass was higher in ASD brain, as indicated by higher protein levels of mitochondrial membrane proteins Tom20, Tim23 and porin. No differences were observed in either mitochondrial DNA or levels of the mitochondrial gene transcription factor TFAM or cofactor PGC1α, indicating that a mechanism other than alterations in mitochondrial genome or mitochondrial biogenesis underlies these mitochondrial abnormalities. We further identified higher levels of the mitochondrial fission proteins (Fis1 and Drp1) and decreased levels of the fusion proteins (Mfn1, Mfn2 and Opa1) in ASD patients, indicating altered mitochondrial dynamics in ASD brain. Many of these changes were evident in cortical pyramidal neurons, and were observed in ASD children but were less pronounced or absent in adult patients. Together, these findings provide evidence that mitochondrial function and intracellular redox status are compromised in pyramidal neurons in ASD brain and that mitochondrial dysfunction occurs during early childhood when ASD symptoms appear.


Subject(s)
Autistic Disorder/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Temporal Lobe/metabolism , Adolescent , Adult , Autistic Disorder/pathology , Blotting, Western , Child , Child, Preschool , Electron Transport Chain Complex Proteins/analysis , Electron Transport Chain Complex Proteins/metabolism , Female , Humans , Male , Middle Aged , Mitochondria/pathology , Temporal Lobe/pathology , Young Adult
20.
Acta Neuropathol ; 126(4): 595-601, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23942639

ABSTRACT

We report a rare case of peripheral T-cell lymphoma arising in a 52-year-old man with biopsy-proven aggressive polymyositis, who had cardiac involvement, progressive bulbar symptoms, and died 11 months post diagnosis due to multiorgan failure. Using a multimodality approach including immunohistochemistry, genome-wide single nucleotide polymorphism (SNP)-array analysis, and high-throughput sequencing of the complementary determining region 3 (CDR3) of T-cell receptor beta (TCRß) genes, our study demonstrates a molecular link between polymyositis and T-cell lymphoma, and provides evidence of the rapid and possibly late occurrence of genomic instability during neoplastic transformation of an oligoclonal T-cell population. Immunohistochemical analysis revealed loss of CD5, CD7, and CD8 antigen expression in autopsy tissue samples, as well as the occurrence of aberrant CD56 expression, not seen in pre-mortem biopsies, supporting the emergence of a neoplastic T-cell population. Multiplex polymerase chain reaction and next-generation sequencing of the TCRß CDR3 region displayed two unique T-cell clones in both the diagnostic biopsy confirming polymyositis and the autopsy muscle tissue exhibiting T-cell lymphoma, linking the two pathological processes. SNP-array analysis revealed complex genomic abnormalities at autopsy but not in the pre-mortem muscle biopsies displaying polymyositis, confirming malignant transformation of the oligoclonal T-cell infiltrate. Our findings raise the possibility that clinically aggressive polymyositis might represent a preneoplastic condition in some instances, similar to certain other autoimmune and inflammatory disorders.


Subject(s)
Cell Transformation, Neoplastic/pathology , Lymphoma, T-Cell, Peripheral/complications , Lymphoma, T-Cell, Peripheral/pathology , Polymyositis/complications , T-Lymphocytes/pathology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Autopsy , Base Sequence , Brain/pathology , Cardiomegaly/pathology , Fatal Outcome , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Sequence Data , Muscle Weakness/etiology , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Myocardium/pathology , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Polymyositis/pathology , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL