Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 77
1.
J Transl Med ; 22(1): 449, 2024 May 13.
Article En | MEDLINE | ID: mdl-38741129

Inherited deficiency of thymidine phosphorylase (TP), encoded by TYMP, leads to a rare disease with multiple mitochondrial DNA (mtDNA) abnormalities, mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). However, the impact of TP deficiency on lysosomes remains unclear, which are important for mitochondrial quality control and nucleic acid metabolism. Muscle biopsy tissue and skin fibroblasts from MNGIE patients, patients with m.3243 A > G mitochondrial encephalopathy, lactic acidosis and stroke-like episodes (MELAS) and healthy controls (HC) were collected to perform mitochondrial and lysosomal functional analyses. In addition to mtDNA abnormalities, compared to controls distinctively reduced expression of LAMP1 and increased mitochondrial content were detected in the muscle tissue of MNGIE patients. Skin fibroblasts from MNGIE patients showed decreased expression of LAMP2, lowered lysosomal acidity, reduced enzyme activity and impaired protein degradation ability. TYMP knockout or TP inhibition in cells can also induce the similar lysosomal dysfunction. Using lysosome immunoprecipitation (Lyso- IP), increased mitochondrial proteins, decreased vesicular proteins and V-ATPase enzymes, and accumulation of various nucleosides were detected in lysosomes with TP deficiency. Treatment of cells with high concentrations of dThd and dUrd also triggers lysosomal dysfunction and disruption of mitochondrial homeostasis. Therefore, the results provided evidence that TP deficiency leads to nucleoside accumulation in lysosomes and lysosomal dysfunction, revealing the widespread disruption of organelles underlying MNGIE.


DNA, Mitochondrial , Fibroblasts , Lysosomes , Mitochondria , Mitochondrial Encephalomyopathies , Nucleosides , Thymidine Phosphorylase , Humans , Lysosomes/metabolism , Thymidine Phosphorylase/metabolism , Thymidine Phosphorylase/deficiency , Thymidine Phosphorylase/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Encephalomyopathies/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Nucleosides/metabolism , Intestinal Pseudo-Obstruction/metabolism , Intestinal Pseudo-Obstruction/pathology , Intestinal Pseudo-Obstruction/enzymology , Intestinal Pseudo-Obstruction/genetics , Ophthalmoplegia/metabolism , Ophthalmoplegia/pathology , Ophthalmoplegia/congenital , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology , Male , Female , Skin/pathology , Skin/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism
2.
Neuromuscul Disord ; 34: 27-31, 2024 Jan.
Article En | MEDLINE | ID: mdl-38052666

Genetic variation at HNRNPA2B1 is associated with inclusion body myopathy, Paget's disease and paediatric onset oculopharyngeal muscular dystrophy. We present a pedigree where a mother and two daughters presented with adolescent to early-adulthood onset of symptoms reminiscent of oculopharyngeal muscular dystrophy or chronic progressive external ophthalmoplegia, with a later limb-girdle pattern of weakness. Creatine Kinase was ∼1000 U/L. Myoimaging identified fatty replacement of sartorius, adductors longus and magnus, biceps femoris, semitendinosus and gastrocnemii. Muscle biopsies showed a variation of fibre size, occasional rimmed vacuoles and increased internalised myonuclei. Cases were heterozygous for a frameshift variant at HNRNPA2B1, consistent with a dominant and fully-penetrant mode of inheritance. Genetic variation at HNRNPA2B1 should be considered in adults with an oculopharyngeal muscular dystrophy-like or chronic progressive external ophthalmoplegia-like myopathy where initial testing fails to identify a cause.


Muscular Diseases , Muscular Dystrophy, Oculopharyngeal , Ophthalmoplegia, Chronic Progressive External , Adolescent , Adult , Child , Humans , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Muscular Dystrophy, Oculopharyngeal/diagnosis , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Ophthalmoplegia, Chronic Progressive External/pathology , Pedigree , Phenotype
3.
Open Biol ; 13(4): 230008, 2023 04.
Article En | MEDLINE | ID: mdl-37042114

Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant disease characterized by the progressive degeneration of specific muscles. OPMD is due to a mutation in the gene encoding poly(A) binding protein nuclear 1 (PABPN1) leading to a stretch of 11 to 18 alanines at N-terminus of the protein, instead of 10 alanines in the normal protein. This alanine tract extension induces the misfolding and aggregation of PABPN1 in muscle nuclei. Here, using Drosophila OPMD models, we show that the unfolded protein response (UPR) is activated in OPMD upon endoplasmic reticulum stress. Mutations in components of the PERK branch of the UPR reduce muscle degeneration and PABPN1 aggregation characteristic of the disease. We show that oral treatment of OPMD flies with Icerguastat (previously IFB-088), a Guanabenz acetate derivative that shows lower side effects, also decreases muscle degeneration and PABPN1 aggregation. Furthermore, the positive effect of Icerguastat depends on GADD34, a key component of the phosphatase complex in the PERK branch of the UPR. This study reveals a major contribution of the ER stress in OPMD pathogenesis and provides a proof-of-concept for Icerguastat interest in future pharmacological treatments of OPMD.


Muscular Dystrophy, Oculopharyngeal , Animals , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology , Muscle, Skeletal/metabolism , Unfolded Protein Response , Cell Nucleus/metabolism , Endoplasmic Reticulum Stress , Drosophila
4.
J Neuromuscul Dis ; 10(3): 459-463, 2023.
Article En | MEDLINE | ID: mdl-36847015

Immediately after the initial methionine codon, the PABPN1 gene encodes a stretch of 10 alanines, 1 glycine, and 2 alanines. Oculopharyngeal muscular dystrophy (OPMD) is caused by the expansion of the first 10 alanine stretches. The only exception is the missense mutation of glycine at the 12th residue into alanine, which elongates the stretch to 13 alanines by connecting the first and second stretch with the addition of one alanine in between, indicating that the expansion or elongation of the alanine stretch results in OPMD. We report a 77-year-old man with the novel missense mutation c.34G > T (p.Gly12Trp) in PABPN1 gene whose clinicopathological findings were compatible with OPMD. He presented with slowly progressive bilateral ptosis, dysphagia, and symmetrical proximal dominant muscle weakness. Magnetic resonance imaging revealed selective fat replacement of the tongue, bilateral adductor magnus, and soleus muscles. Immunohistochemistry studies of the muscle biopsy sample revealed PABPN1-posibive aggregates in the myonuclei which have been reported to be specific to OPMD. This is the first OPMD case caused by neither the expansion nor the elongation of alanine stretch. The present case suggests that OPMD may be caused not only by triplet repeats but also by point mutations.


Muscular Dystrophy, Oculopharyngeal , Male , Humans , Aged , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Point Mutation , Alanine/genetics , Glycine/genetics , Poly(A)-Binding Protein I/genetics
5.
Ann Clin Transl Neurol ; 10(3): 426-439, 2023 03.
Article En | MEDLINE | ID: mdl-36691350

OBJECTIVE: Oculopharyngeal muscular dystrophy (OPMD) is a late-onset inherited neuromuscular disorder, with progressive ptosis and dysphagia as common manifestations. To date, OPMD has rarely been reported among East Asians. The present study summarizes the phenotypic and genotypic features of Chinese patients with OPMD. METHODS: Twenty-one patients with molecularly confirmed OPMD from 9 unrelated families were identified by direct sequencing of the polyadenlyate binding protein nuclear-1 (PABPN1) gene. Immunofluorescence staining of muscle biopsies was conducted to identify the components of protein degradation pathways involved in OPMD. RESULTS: In our cohort, the genetically confirmed OPMD group had a mean age at onset of 50.6 ± 4.2 years (range 45-60 years). Ptosis (42.9%) was the most common initial symptom; patients with ptosis as the first symptom subsequently developed dysphagia within a median time of 5.5 years (range 1-19 years). Evidence of external ophthalmoplegia was found in 38.1% of patients. A total of 33.3% of the patients developed muscle weakness at a median age at onset of 66 years (range 50-70 years), with neck flexor involvement in all patients. Five genotypes were observed in our cohort, including classical (GCG)9-11 repeats in 7 families and non-GCG elongations with additional GCA expansions in 2 families. OPMD muscle biopsies revealed rimmed vacuoles and intranuclear filamentous inclusions. The PABPN1 protein showed substantial accumulation in the nuclei of muscle fiber aggregates and closely colocalized with p62, LC3B and FK2. INTERPRETATION: Our findings indicate wide genetic heterogeneity in OPMD in the Chinese population and demonstrate abnormalities in protein degradation pathways in this disease.


Deglutition Disorders , Muscular Dystrophy, Oculopharyngeal , Humans , Middle Aged , Aged , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology , East Asian People , Genotype , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein I/genetics
6.
Methods Mol Biol ; 2587: 557-568, 2023.
Article En | MEDLINE | ID: mdl-36401050

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset rare muscle disease affecting approximately 1 in 80,000 individuals worldwide. However, it can affect as much as 1:600 individuals in some populations due to a strong founder effect. The muscle pathology is characterized by progressive eyelid drooping (ptosis), swallowing difficulties (dysphagia), and limb weakness at later stages of disease progression. The genetic defect is associated with significant fibrotic deposition and atrophy in affected muscles. No treatments are available to cure the disease. Only surgical techniques to correct ptosis and swallowing are currently possible, though they carry a risk of recurrence. Myostatin is a negative regulator of muscle growth, and several strategies to downregulate its expression have been developed with the aim of improving muscle mass and strength in muscular pathologies. We recently showed that weekly systemic treatment of the A17 murine model of OPMD with a monoclonal antibody for myostatin improves body and muscle mass, increases muscle strength, and reduces muscle fibrosis. Here, we describe the methodology for repeated intraperitoneal delivery of myostatin antibody in the murine model. Furthermore, we detail the most relevant analyses to assess histopathological and functional improvements of this treatment in this mouse model.


Muscular Dystrophy, Oculopharyngeal , Mice , Animals , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Myostatin , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Immunotherapy
7.
Acta Neuropathol ; 144(6): 1157-1170, 2022 12.
Article En | MEDLINE | ID: mdl-36197469

Oculopharyngeal muscular dystrophy (OPMD) is a rare muscle disease characterized by an onset of weakness in the pharyngeal and eyelid muscles. The disease is caused by the extension of a polyalanine tract in the Poly(A) Binding Protein Nuclear 1 (PABPN1) protein leading to the formation of intranuclear inclusions or aggregates in the muscle of OPMD patients. Despite numerous studies stressing the deleterious role of nuclear inclusions in cellular and animal OPMD models, their exact contribution to human disease is still unclear. In this study, we used a large and unique collection of human muscle biopsy samples to perform an in-depth analysis of PABPN1 aggregates in relation to age, genotype and muscle status with the final aim to improve our understanding of OPMD physiopathology. Here we demonstrate that age and genotype influence PABPN1 aggregates: the percentage of myonuclei containing PABPN1 aggregates increases with age and the chaperone HSP70 co-localize more frequently with PABPN1 aggregates with a larger polyalanine tract. In addition to the previously described PRMT1 and HSP70 co-factors, we identified new components of PABPN1 aggregates including GRP78/BiP, RPL24 and p62. We also observed that myonuclei containing aggregates are larger than myonuclei without. When comparing two muscles from the same patient, a similar amount of aggregates is observed in different muscles, except for the pharyngeal muscle where fewer aggregates are observed. This could be due to the peculiar nature of this muscle which has a low level of PAPBN1 and contains regenerating fibers. To confirm the fate of PABPN1 aggregates in a regenerating muscle, we generated a xenograft model by transplanting human OPMD muscle biopsy samples into the hindlimb of an immunodeficient mouse. Xenografts from subjects with OPMD displayed regeneration of human myofibers and PABPN1 aggregates were rapidly present-although to a lower extent-after muscle fiber regeneration. Our data obtained on human OPMD samples add support to the dual non-exclusive models in OPMD combining toxic PABPN1 intranuclear inclusions together with PABPN1 loss of function which altogether result in this late-onset and muscle selective disease.


Muscular Dystrophy, Oculopharyngeal , Humans , Mice , Animals , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Intranuclear Inclusion Bodies/metabolism , Intranuclear Inclusion Bodies/pathology , Heterografts , Disease Models, Animal , Molecular Chaperones/metabolism , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism
8.
Otol Neurotol ; 43(4): e442-e445, 2022 04 01.
Article En | MEDLINE | ID: mdl-35120077

OBJECTIVES: To describe cases of patulous Eustachian tube (PET) or patent ET conditions in oculopharyngeal muscular dystrophy (OPMD). PATIENTS: Four cases of PET or patent ET conditions with OPMD. MAIN OUTCOME MEASURES: Clinical case records, objective ET function tests (tubo-tympano-aerodynamic graphy and sonotubometry), and swallowing function (videoendoscopic examination and Food Intake Level Scale) were analyzed. RESULTS: Two cases of definite PET, one case of possible PET, and one case lacking aural symptoms with findings of patent ET. All patients have ptosis, and three cases have dysphagia. Body mass index indicated that three cases were underweight. Magnetic resonance imaging in case 4 showed atrophy and fat replacement of palatine and masticatory muscles. CONCLUSIONS: It is important to consider PET or patent ET conditions when OPMD patients describe aural symptoms.


Ear Diseases , Eustachian Tube , Muscular Dystrophy, Oculopharyngeal , Otitis Media , Ear Diseases/pathology , Humans , Muscular Dystrophy, Oculopharyngeal/complications , Muscular Dystrophy, Oculopharyngeal/pathology , Otitis Media/pathology
9.
PLoS Genet ; 18(1): e1010015, 2022 01.
Article En | MEDLINE | ID: mdl-35025870

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder characterized by progressive weakness and degeneration of specific muscles. OPMD is due to extension of a polyalanine tract in poly(A) binding protein nuclear 1 (PABPN1). Aggregation of the mutant protein in muscle nuclei is a hallmark of the disease. Previous transcriptomic analyses revealed the consistent deregulation of the ubiquitin-proteasome system (UPS) in OPMD animal models and patients, suggesting a role of this deregulation in OPMD pathogenesis. Subsequent studies proposed that UPS contribution to OPMD involved PABPN1 aggregation. Here, we use a Drosophila model of OPMD to address the functional importance of UPS deregulation in OPMD. Through genome-wide and targeted genetic screens we identify a large number of UPS components that are involved in OPMD. Half dosage of UPS genes reduces OPMD muscle defects suggesting a pathological increase of UPS activity in the disease. Quantification of proteasome activity confirms stronger activity in OPMD muscles, associated with degradation of myofibrillar proteins. Importantly, improvement of muscle structure and function in the presence of UPS mutants does not correlate with the levels of PABPN1 aggregation, but is linked to decreased degradation of muscle proteins. Oral treatment with the proteasome inhibitor MG132 is beneficial to the OPMD Drosophila model, improving muscle function although PABPN1 aggregation is enhanced. This functional study reveals the importance of increased UPS activity that underlies muscle atrophy in OPMD. It also provides a proof-of-concept that inhibitors of proteasome activity might be an attractive pharmacological approach for OPMD.


Muscular Atrophy/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Poly(A)-Binding Protein I/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Disease Models, Animal , Drosophila melanogaster , Gene Expression Regulation , Genetic Testing , Humans , Leupeptins/pharmacology , Leupeptins/therapeutic use , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Dystrophy, Oculopharyngeal/drug therapy , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Mutation , Poly(A)-Binding Protein I/chemistry , Proof of Concept Study , Protein Aggregates/drug effects
10.
Med Sci (Paris) ; 38 Hors série n° 1: 13-16, 2022 Dec.
Article Fr | MEDLINE | ID: mdl-36649629

Oculopharyngeal muscular dystrophy (OPMD) is one of the diseases related to pathological expansions of trinucleotides. Its pathogenesis remains unclear although the presence of aggregates within the nuclei of the muscle fiber seems to play an important role. The basic research studies presented here help understand their composition and their deleterious role. These elements may result in new therapeutic avenues.


Title: Les agrégats nucléaires dans la dystrophie musculaire oculopharyngée. Abstract: La dystrophie musculaire oculopharyngée est une des maladies en rapport avec des expansions pathologiques de triplets nucléotidiques. Sa physiopathologie est encore imparfaitement connue même si la présence d'agrégats au niveau des noyaux de la fibre musculaire semble jouer un rôle déterminant. Les travaux fondamentaux présentés ici permettent de mieux comprendre leur composition et leur rôle délétère. Autant d'éléments qui pourraient déboucher sur des voies thérapeutiques nouvelles.


Muscular Dystrophy, Oculopharyngeal , Humans , Muscular Dystrophy, Oculopharyngeal/pathology , Cell Nucleus , Muscle Fibers, Skeletal/pathology
11.
J Mol Neurosci ; 71(12): 2526-2533, 2021 Dec.
Article En | MEDLINE | ID: mdl-33825174

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a multi-system disorder caused by several homozygous or compound heterozygous mutations, mostly in the nuclear gene of TYMP. Our current knowledge on the underlying pathology of the disease is derived through the study of about 200 cases of different ethnicities. Clinical presentations include severe cachexia, weakness, ptosis, diplopia, abdominal cramps or digestive tract disorders, hearing impairment, and paresthesia.Herein, we aim to present five novel mutations of the nuclear gene of TYMP in six Iranian patients diagnosed with MNGIE. In our population, age at the time of diagnosis was 18 to 49 years, while the onset of the symptoms varied from 13 to 20 years. We detected two pathogenic non-frameshift nonsense premature stop codon mutations (c.1013C > A, and c.130C > T), one variant of uncertain significance (VUS) non-frameshift missense mutation (c.345G > T), one likely pathogenic frameshift insertion (c.801_802insCGCG), and one likely benign homozygous non-frameshift deletion (c.1176_1187del) from two siblings. Our findings also confirm the autosomal recessive inheritance pattern of MNGIE in the Iranian population. The lack of knowledge in the area of nuclear gene-modifier genes shadows the genotype-phenotype relationships of MNGIE.


Intestinal Pseudo-Obstruction/genetics , Muscular Dystrophy, Oculopharyngeal/genetics , Ophthalmoplegia/congenital , Phenotype , Thymidine Phosphorylase/genetics , Adolescent , Codon, Nonsense , Female , Genes, Recessive , Humans , Intestinal Pseudo-Obstruction/pathology , Iran , Male , Muscular Dystrophy, Oculopharyngeal/pathology , Ophthalmoplegia/genetics , Ophthalmoplegia/pathology , Thymidine Phosphorylase/metabolism , Young Adult
12.
Am J Physiol Gastrointest Liver Physiol ; 320(5): G768-G779, 2021 05 01.
Article En | MEDLINE | ID: mdl-33655764

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a rare autosomal recessive disease caused by thymidine phosphorylase (TP) enzyme defect. As gastrointestinal changes do not revert in patients undergone TP replacement therapy, one can postulate that other unexplored mechanisms contribute to MNGIE pathophysiology. Hence, we focused on the local TP angiogenic potential that has never been considered in MNGIE. In this study, we investigated the enteric submucosal microvasculature and the effect of hypoxia on fibrosis and enteric neurons density in jejunal full-thickness biopsies collected from patients with MNGIE. Orcein staining was used to count blood vessels based on their size. Fibrosis was assessed using the Sirius Red and Fast Green method. Hypoxia and neoangiogenesis were determined via hypoxia-inducible-factor-1α (HIF-1α) and vascular endothelial cell growth factor (VEGF) protein expression, respectively. Neuron-specific enolase was used to label enteric neurons. Compared with controls, patients with MNGIE showed a decreased area of vascular tissue, but a twofold increase of submucosal vessels/mm2 with increased small size and decreased medium and large size vessels. VEGF positive vessels, fibrosis index, and HIF-1α protein expression were increased, whereas there was a diminished thickness of the longitudinal muscle layer with an increased interganglionic distance and reduced number of myenteric neurons. We demonstrated the occurrence of an angiopathy in the GI tract of patients with MNGIE. Neoangiogenetic changes, as detected by the abundance of small size vessels in the jejunal submucosa, along with hypoxia provide a morphological basis to explain neuromuscular alterations, vasculature breakdown, and ischemic abnormalities in MNGIE.NEW & NOTEWORTHY Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is characterized by a genetically driven defect of thymidine phosphorylase, a multitask enzyme playing a role also in angiogenesis. Indeed, major gastrointestinal bleedings are life-threatening complications of MNGIE. Thus, we focused on jejunal submucosal vasculature and showed intestinal microangiopathy as a novel feature occurring in this disease. Notably, vascular changes were associated with neuromuscular abnormalities, which may explain gut dysfunction and help to develop future therapeutic approaches in MNGIE.


Gastrointestinal Tract/metabolism , Intestinal Pseudo-Obstruction/metabolism , Mitochondrial Encephalomyopathies/metabolism , Muscular Dystrophy, Oculopharyngeal/metabolism , Neovascularization, Pathologic/metabolism , Ophthalmoplegia/congenital , Gastrointestinal Tract/pathology , Humans , Intestinal Pseudo-Obstruction/pathology , Mitochondrial Encephalomyopathies/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Neovascularization, Pathologic/pathology , Ophthalmoplegia/metabolism , Ophthalmoplegia/pathology , Thymidine Phosphorylase/metabolism
13.
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
14.
J Neuromuscul Dis ; 7(4): 495-504, 2020.
Article En | MEDLINE | ID: mdl-32925090

BACKGROUND: Muscle MRI is increasingly used as a diagnostic and research tool in muscle disorders. However, the correlation between MRI abnormalities and histopathological severity is largely unknown. OBJECTIVE: To investigate correlations between muscle MRI abnormalities and histopathological severity in healthy controls and patients with muscle disease. METHODS: We performed quantitative MRI and histopathological analysis in 35 patients with inclusion body myositis, facioscapulohumeral muscular dystrophy or oculopharyngeal muscular dystrophy and 12 healthy controls. Participants contributed needle biopsies of the vastus lateralis and/or tibialis anterior, yielding 77 muscle biopsies with matched T1, T2 and TIRM MRI imaging. Muscle biopsies were evaluated with a semi-quantitative histopathology severity grading scale (range 0-12) and an inflammation severity grading scale (range 0-3). RESULTS: In muscle disease, histopathology sum scores ranged from 0 to 11 and correlated significantly with fat percentage as measured on MRI (Spearman's rho = 0.594, p < 0.001). Muscle edema on muscle MRI was associated with increased amounts of inflammation (p < 0.001). Mild abnormalities occured in 95% of control biopsies and were more pronounced in tibialis anterior (median sum score of 1±1 in vastus lateralis and 2±1 in tibialis anterior (p = 0.048)). CONCLUSION: In muscle disease, fatty infiltration on MRI correlates moderately with muscle histopathology. Histopathological abnormalities can occur prior to the onset of fatty infiltration. In middle-aged controls, almost all biopsies showed some histopathological abnormalities. The findings from this study may facilitate the choice for appropriate imaging sequences as outcome measures in therapeutic trials.


Biopsy/standards , Magnetic Resonance Imaging/standards , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/diagnosis , Muscular Dystrophy, Oculopharyngeal/diagnosis , Myositis, Inclusion Body/diagnosis , Female , Humans , Male , Middle Aged , Muscular Dystrophy, Facioscapulohumeral/diagnostic imaging , Muscular Dystrophy, Facioscapulohumeral/pathology , Muscular Dystrophy, Oculopharyngeal/diagnostic imaging , Muscular Dystrophy, Oculopharyngeal/pathology , Myositis, Inclusion Body/diagnostic imaging , Myositis, Inclusion Body/pathology , Severity of Illness Index
15.
Eur J Neurol ; 27(4): 709-715, 2020 04.
Article En | MEDLINE | ID: mdl-31769567

BACKGROUND AND PURPOSE: The aim was to assess the value of insoluble PABPN1 muscle fibre nuclei accumulation in the diagnosis of atypical cases of oculopharyngeal muscular dystrophy (OPMD). METHODS: Muscle biopsies from a selected cohort of 423 adult patients from several Italian neuromuscular centres were analysed by immunofluorescence: 30 muscle biopsies of genetically proven OPMD, 30 biopsies from patients not affected by neuromuscular disorders, 220 from genetically undiagnosed patients presenting ptosis or swallowing disturbances, progressive lower proximal weakness and/or isolated rimmed vacuoles at muscle biopsy and 143 muscle biopsies of patients affected by other neuromuscular diseases. RESULTS: The detection of insoluble nuclear PABPN1 accumulation is rapid, sensitive (100%) and specific (96%). The revision of our cohort allowed us to discover 23 new OPMD cases out of 220 patients affected with nonspecific muscle diseases. CONCLUSIONS: Oculopharyngeal muscular dystrophy is often misdiagnosed leading to diagnosis delay, causing waste of time and resources. A great number of these cases present symptoms and histological findings frequently overlapping with other muscle diseases, i.e. inclusion body myositis and progressive external ophthalmoplegia. PABPN1 nuclear accumulation is a reliable method for diagnostic purposes and it is safe and useful in helping pathologists and clinicians to direct genetic analysis in the case of suspected OPMD, even when clinical and histological clues are deceptive.


Cell Nucleus/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Oculopharyngeal/diagnosis , Poly(A)-Binding Protein I/metabolism , Cell Nucleus/pathology , Fluorescent Antibody Technique , Humans , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology
16.
J Cachexia Sarcopenia Muscle ; 10(5): 1016-1026, 2019 10.
Article En | MEDLINE | ID: mdl-31066242

BACKGROUND: Oculopharyngeal muscular dystrophy (OPMD) is a late-onset muscle disease affecting one per 80 000 of the general population characterized by profound dysphagia and ptosis, and limb weakness at later stages. Affected muscles are characterized by increased fibrosis and atrophy. Myostatin is a negative regulator of muscle mass, and inhibition of myostatin has been demonstrated to ameliorate symptoms in dystrophic muscles. METHODS: In this study, we performed a systemic delivery of a monoclonal antibody to immunologically block myostatin in the A17 mouse model of OPMD. The mice were administered a weekly dose of 10 mg/kg RK35 intraperitonially for 10 weeks, following which histological analyses were performed on the samples. RESULTS: This treatment significantly (P < 0.01) improved body mass (11%) and muscle mass (for the tibialis anterior and extensor digitorum longus by 19% and 41%) in the A17 mice treated with RK35 when compared to saline controls. Similarly, a significantly (P < 0.01) increased muscle strength (18% increase in maximal tetanic force) and myofibre diameter (17% and 44% for the tibialis anterior and extensor digitorum longus), and reduced expression of markers of muscle fibrosis (40% reduction in area of expression), was also observed. No change in the density of intranuclear inclusions (a hallmark of disease progression of OPMD) was however observed. CONCLUSIONS: Our study supports the clinical translation of such antibody-mediated inhibition of myostatin as a treatment of OPMD. This strategy has implications to be used as adjuvant therapies with gene therapy based approaches, or to stabilize the muscle prior to myoblast transplantation.


Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology , Myostatin/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers , Body Composition , Immunohistochemistry , Mice , Muscle Strength , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Oculopharyngeal/etiology , Myoblasts/metabolism
17.
Lab Invest ; 99(11): 1728-1740, 2019 11.
Article En | MEDLINE | ID: mdl-30894671

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder characterized by ptosis, dysphagia, and weakness of proximal limbs. OPMD is caused by the expansion of polyalanine in poly(A)-binding protein, nuclear 1 (PABPN1). Although mitochondrial abnormality has been proposed as the possible etiology, the molecular pathogenesis is still poorly understood. The aim of the study was to specify the mechanism by which expanded PABPN1 causes mitochondrial dysfunction in OPMD. We evaluated whether transgenic mouse model of OPMD, by expressing expanded PABPN1, indeed causes mitochondrial abnormality associated with muscle degeneration. We also investigated the mechanism by which expanded PABPN1 would cause mitochondrial dysfunction in the mouse and cell models of OPMD. Mitochondrial localization of PABPN1 was observed in the muscle fibers of patients with OPMD. Moreover, abnormal accumulation of PABPN1 on the inner membrane of mitochondria and reduced expression of OXPHOS complexes were detected in the muscle fibers of the transgenic mice expressing expanded human PABPN1 with a 13-alanine stretch. In cells expressing PABPN1 with a 10-alanine or 18-alanine stretch, both types of PABPN1 accumulated in the mitochondria and interacted with TIM23 mitochondrial protein import complex, but PABPN1 with 18-alanine stretch decreased the cell viability and aggresome formation. We proposed that the abnormal accumulation of expanded PABPN1 in mitochondria may be associated with mitochondrial abnormality in OPMD.


Mitochondria, Muscle/metabolism , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Trinucleotide Repeat Expansion , Animals , Case-Control Studies , Cell Survival , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria, Muscle/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Mutant Proteins/chemistry , Oxidative Phosphorylation , Poly(A)-Binding Protein I/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Orphanet J Rare Dis ; 14(1): 33, 2019 02 08.
Article En | MEDLINE | ID: mdl-30736844

BACKGROUND: MNGIE is a rare and fatal disease in which absence of the enzyme thymidine phosphorylase induces systemic accumulation of thymidine and deoxyuridine and secondary mitochondrial DNA alterations. Gastrointestinal (GI) symptoms are frequently reported in MNGIE patients, however, they are not resolved with the current treatment interventions. Recently, our understanding of the GI pathology has increased, which rationalizes the pursuit of more targeted therapeutic strategies. In particular, interstitial cells of Cajal (ICC) play key roles in GI physiology and are involved in the pathogenesis of the GI dysmotility. However, understanding of the triggers of ICC deficits in MNGIE is lacking. Herein, we review the current knowledge about the pathology of GI dysmotility in MNGIE, discuss potential mechanisms in relation to ICC loss/dysfunction, remark on the limited contribution of the current treatments, and propose intervention strategies to overcome ICC deficits. Finally, we address the advances and new research avenues offered by organoids and tissue engineering technologies, and propose schemes to implement to further our understanding of the GI pathology and utility in regenerative and personalized medicine in MNGIE. CONCLUSION: Interstitial cells of Cajal play key roles in the physiology of the gastrointestinal motility. Evaluation of their status in the GI dysmotility related to MNGIE would be valuable for diagnosis of MNGIE. Understanding the underlying pathological and molecular mechanisms affecting ICC is an asset for the development of targeted prevention and treatment strategies for the GI dysmotility related to MNGIE.


Gastrointestinal Diseases/pathology , Interstitial Cells of Cajal/pathology , Thymidine Phosphorylase/deficiency , Female , Gastrointestinal Diseases/metabolism , Humans , Interstitial Cells of Cajal/metabolism , Intestinal Pseudo-Obstruction/metabolism , Intestinal Pseudo-Obstruction/pathology , Male , Muscular Dystrophy, Oculopharyngeal/metabolism , Muscular Dystrophy, Oculopharyngeal/pathology , Mutation/genetics , Ophthalmoplegia/congenital , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/metabolism
19.
Hum Mol Genet ; 28(10): 1694-1708, 2019 05 15.
Article En | MEDLINE | ID: mdl-30649389

Oculopharyngeal muscular dystrophy (OPMD) is a rare late onset genetic disease leading to ptosis, dysphagia and proximal limb muscles at later stages. A short abnormal (GCN) triplet expansion in the polyA-binding protein nuclear 1 (PABPN1) gene leads to PABPN1-containing aggregates in the muscles of OPMD patients. Here we demonstrate that treating mice with guanabenz acetate (GA), an FDA-approved antihypertensive drug, reduces the size and number of nuclear aggregates, improves muscle force, protects myofibers from the pathology-derived turnover and decreases fibrosis. GA targets various cell processes, including the unfolded protein response (UPR), which acts to attenuate endoplasmic reticulum (ER) stress. We demonstrate that GA increases both the phosphorylation of the eukaryotic translation initiation factor 2α subunit and the splicing of Xbp1, key components of the UPR. Altogether these data show that modulation of protein folding regulation is beneficial for OPMD and promote the further development of GA or its derivatives for treatment of OPMD in humans. Furthermore, they support the recent evidences that treating ER stress could be therapeutically relevant in other more common proteinopathies.


Guanabenz/pharmacology , Muscular Dystrophy, Oculopharyngeal/drug therapy , Poly(A)-Binding Protein I/genetics , X-Box Binding Protein 1/genetics , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Fibrosis/drug therapy , Fibrosis/genetics , Fibrosis/pathology , Humans , Mice , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Phosphorylation/drug effects , Protein Aggregates/drug effects , Protein Aggregates/genetics , Protein Folding , Unfolded Protein Response/drug effects
20.
J Neurol Neurosurg Psychiatry ; 90(5): 576-585, 2019 05.
Article En | MEDLINE | ID: mdl-30530568

BACKGROUND AND OBJECTIVE: Oculopharyngeal muscular dystrophy (OPMD) is a genetic disorder caused by an abnormal expansion of GCN triplets within the PABPN1 gene. Previous descriptions have focused on lower limb muscles in small cohorts of patients with OPMD, but larger imaging studies have not been performed. Previous imaging studies have been too small to be able to correlate imaging findings to genetic and clinical data. METHODS: We present cross-sectional, T1-weighted muscle MRI and CT-scan data from 168 patients with genetically confirmed OPMD. We have analysed the pattern of muscle involvement in the disease using hierarchical analysis and presented it as heatmaps. Results of the scans were correlated with genetic and clinical data. RESULTS: Fatty replacement was identified in 96.7% of all symptomatic patients. The tongue, the adductor magnus and the soleus were the most commonly affected muscles. Muscle pathology on MRI correlated positively with disease duration and functional impairment. CONCLUSIONS: We have described a pattern that can be considered characteristic of OPMD. An early combination of fat replacement in the tongue, adductor magnus and soleus can be helpful for differential diagnosis. The findings suggest the natural history of the disease from a radiological point of view. The information generated by this study is of high diagnostic value and important for clinical trial development.


Muscle, Skeletal/diagnostic imaging , Muscular Dystrophy, Oculopharyngeal/diagnostic imaging , Adult , Cohort Studies , Cross-Sectional Studies , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal/complications , Muscular Dystrophy, Oculopharyngeal/pathology , Tomography, X-Ray Computed
...