Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Ann Clin Transl Neurol ; 11(3): 629-640, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38311799

ABSTRACT

OBJECTIVE: ACTN2, encoding alpha-actinin-2, is essential for cardiac and skeletal muscle sarcomeric function. ACTN2 variants are a known cause of cardiomyopathy without skeletal muscle involvement. Recently, specific dominant monoallelic variants were reported as a rare cause of core myopathy of variable clinical onset, although the pathomechanism remains to be elucidated. The possibility of a recessively inherited ACTN2-myopathy has also been proposed in a single series. METHODS: We provide clinical, imaging, and histological characterization of a series of patients with a novel biallelic ACTN2 variant. RESULTS: We report seven patients from five families with a recurring biallelic variant in ACTN2: c.1516A>G (p.Arg506Gly), all manifesting with a consistent phenotype of asymmetric, progressive, proximal, and distal lower extremity predominant muscle weakness. None of the patients have cardiomyopathy or respiratory insufficiency. Notably, all patients report Palestinian ethnicity, suggesting a possible founder ACTN2 variant, which was confirmed through haplotype analysis in two families. Muscle biopsies reveal an underlying myopathic process with disruption of the intermyofibrillar architecture, Type I fiber predominance and atrophy. MRI of the lower extremities demonstrate a distinct pattern of asymmetric muscle involvement with selective involvement of the hamstrings and adductors in the thigh, and anterior tibial group and soleus in the lower leg. Using an in vitro splicing assay, we show that c.1516A>G ACTN2 does not impair normal splicing. INTERPRETATION: This series further establishes ACTN2 as a muscle disease gene, now also including variants with a recessive inheritance mode, and expands the clinical spectrum of actinopathies to adult-onset progressive muscle disease.


Subject(s)
Cardiomyopathies , Muscular Diseases , Adult , Humans , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Actinin/genetics , Phenotype
2.
Article in English | MEDLINE | ID: mdl-38083393

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a genetic neuromuscular progressive multisystem disease that results in a broad spectrum of clinical central nervous system (CNS) involvement, including problems with memory, attention, executive functioning, and social cognition. Fractional anisotropy and mean diffusivity along-tract data calculated using diffusion tensor imaging techniques play a vital role in assessing white matter microstructural changes associated with neurodegeneration caused by DM1. In this work, a novel spectrogram-based deep learning method is proposed to characterize white matter network alterations in DM1 with the goal of building a deep learning model as neuroimaging biomarkers of DM1. The proposed method is evaluated on fractional anisotropies and mean diffusivities along-tract data calculated for 25 major white matter tracts of 46 DM1 patients and 96 unaffected controls. The evaluation data consists of a total of 7100 spectrogram images. The model achieved 91% accuracy in identifying DM1, a significant improvement compared to previous methods.Clinical relevance- Clinical care of DM1 is particularly challenging due to DM1 multisystem involvement and the disease variability. Patients with DM1 often experience neurological and psychological symptoms, such as excessive sleepiness and apathy, that greatly impact their quality of life. Some of DM1 CNS symptoms may be responsive to treatment. The goal of this research is to gain a deeper understanding of the impact of DM1 on the CNS and to develop a deep learning model that can serve as a biomarker for the disease, with the potential to be used in future clinical trials as an outcome measure.


Subject(s)
Myotonic Dystrophy , White Matter , Humans , White Matter/diagnostic imaging , Myotonic Dystrophy/diagnostic imaging , Myotonic Dystrophy/complications , Myotonic Dystrophy/psychology , Diffusion Tensor Imaging , Anisotropy , Quality of Life , Neuroimaging
3.
J Clin Med ; 12(20)2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37892834

ABSTRACT

Disease-modifying treatments have transformed the natural history of spinal muscular atrophy (SMA), but the cellular pathways altered by SMN restoration remain undefined and biomarkers cannot yet precisely predict treatment response. We performed an exploratory cerebrospinal fluid (CSF) proteomic study in a diverse sample of SMA patients treated with nusinersen to elucidate therapeutic pathways and identify predictors of motor improvement. Proteomic analyses were performed on CSF samples collected before treatment (T0) and at 6 months (T6) using an Olink panel to quantify 1113 peptides. A supervised machine learning approach was used to identify proteins that discriminated patients who improved functionally from those who did not after 2 years of treatment. A total of 49 SMA patients were included (10 type 1, 18 type 2, and 21 type 3), ranging in age from 3 months to 65 years. Most proteins showed a decrease in CSF concentration at T6. The machine learning algorithm identified ARSB, ENTPD2, NEFL, and IFI30 as the proteins most predictive of improvement. The machine learning model was able to predict motor improvement at 2 years with 79.6% accuracy. The results highlight the potential application of CSF biomarkers to predict motor improvement following SMA treatment. Validation in larger datasets is needed.

4.
Brain ; 146(10): 4217-4232, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37143315

ABSTRACT

Myotonic dystrophy type 1 is a dominantly inherited multisystemic disease caused by CTG tandem repeat expansions in the DMPK 3' untranslated region. These expanded repeats are transcribed and produce toxic CUG RNAs that sequester and inhibit activities of the MBNL family of developmental RNA processing factors. Although myotonic dystrophy is classified as a muscular dystrophy, the brain is also severely affected by an unusual cohort of symptoms, including hypersomnia, executive dysfunction, as well as early onsets of tau/MAPT pathology and cerebral atrophy. To address the molecular and cellular events that lead to these pathological outcomes, we recently generated a mouse Dmpk CTG expansion knock-in model and identified choroid plexus epithelial cells as particularly affected by the expression of toxic CUG expansion RNAs. To determine if toxic CUG RNAs perturb choroid plexus functions, alternative splicing analysis was performed on lateral and hindbrain choroid plexi from Dmpk CTG knock-in mice. Choroid plexus transcriptome-wide changes were evaluated in Mbnl2 knockout mice, a developmental-onset model of myotonic dystrophy brain dysfunction. To determine if transcriptome changes also occurred in the human disease, we obtained post-mortem choroid plexus for RNA-seq from neurologically unaffected (two females, three males; ages 50-70 years) and myotonic dystrophy type 1 (one female, three males; ages 50-70 years) donors. To test that choroid plexus transcriptome alterations resulted in altered CSF composition, we obtained CSF via lumbar puncture from patients with myotonic dystrophy type 1 (five females, five males; ages 35-55 years) and non-myotonic dystrophy patients (three females, four males; ages 26-51 years), and western blot and osmolarity analyses were used to test CSF alterations predicted by choroid plexus transcriptome analysis. We determined that CUG RNA induced toxicity was more robust in the lateral choroid plexus of Dmpk CTG knock-in mice due to comparatively higher Dmpk and lower Mbnl RNA levels. Impaired transitions to adult splicing patterns during choroid plexus development were identified in Mbnl2 knockout mice, including mis-splicing previously found in Dmpk CTG knock-in mice. Whole transcriptome analysis of myotonic dystrophy type 1 choroid plexus revealed disease-associated RNA expression and mis-splicing events. Based on these RNA changes, predicted alterations in ion homeostasis, secretory output and CSF composition were confirmed by analysis of myotonic dystrophy type 1 CSF. Our results implicate choroid plexus spliceopathy and concomitant alterations in CSF homeostasis as an unappreciated contributor to myotonic dystrophy type 1 CNS pathogenesis.


Subject(s)
Myotonic Dystrophy , Humans , Female , Mice , Animals , Myotonic Dystrophy/genetics , Choroid Plexus/metabolism , Choroid Plexus/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Alternative Splicing , RNA/genetics , Mice, Knockout , Trinucleotide Repeat Expansion
5.
Annu Int Conf IEEE Eng Med Biol Soc ; 2022: 4377-4382, 2022 07.
Article in English | MEDLINE | ID: mdl-36086274

ABSTRACT

The myotonic dystrophies (DM1 and DM2) are dominantly inherited disorders that cause pathological changes throughout the body. Many individuals with DM experience cognitive, behavioral and other functional central nervous system effects that impact their quality of life. The extent of psychological impairment that will develop in each patient is variable and unpredictable. Hence, it is difficult to get strong supervision information like fully ground truth labels for all cognitive involvement patterns. This study is to assess cognitive involvement among healthy controls and patients with DM. The DM cognitive impairment pattern observation is modeled in a weakly supervised setting and supervision information is used to transform the input feature space to a more discriminative representation suitable for pattern observation. This study incorporated results from 59 adults with DM and 92 control subjects. The developed system categorized the neuropsychological testing data into five cognitive clusters. The quality of the obtained clustering solution was assessed using an internal validity metric. The experimental results show that the proposed algorithm can discover interesting patterns and useful information from neuropsychological data, which will be be crucial in planning clinical trials and monitoring clinical performance. The proposed system resulted in an average classification accuracy of 88%, which is very promising considering the unique challenges present in this population.


Subject(s)
Cognitive Dysfunction , Myotonic Dystrophy , Adult , Cluster Analysis , Cognitive Dysfunction/diagnosis , Humans , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/pathology , Neuropsychological Tests , Quality of Life
7.
Orphanet J Rare Dis ; 17(1): 79, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197080

ABSTRACT

BACKGROUND: Myotonic dystrophy (DM) is a rare, inherited disorder with multi-systemic effects that impact the skeletal muscles, eyes, heart, skin and gastrointestinal, endocrine, respiratory, and central nervous systems. DM is divided into two subtypes: DM1 can present from early childhood through adulthood and also has a congenital form (cDM) while DM2 typically manifests during mid-adulthood. Both forms are progressive with no approved treatments, and unmet need for disease-modifying therapies remains high. This study interrogated health insurance claims data to explore the clinical experience, healthcare resource utilization (HCRU), and all-cause costs for DM. RESULTS: A total of 8541 patients with DM and 242 patients with cDM and their matched controls were selected from a database of over 200 million claimants. HCRU and all-cause costs, including pharmacy, outpatient, and inpatient services, were analyzed across four years in 12-month follow-up periods. Mean all-cause costs per DM patient were high in each of the four periods (range $14,640-$16,704) and showed a steady increase from 13 to 23 months on, while the control group mean costs declined from $9671 in the first 12 months after the index event, to approach the US population average ($5193) over time. For cDM, the highest mean costs were in the first 12-months ($66,496 vs. $2818 for controls), and remained high (above $17,944) across all subsequent periods, while control mean costs approached $0. For DM and cDM, HCRU was higher compared to controls across all study periods and all-cause healthcare costs were mostly driven by inpatient and outpatient encounters. Analysis of all diagnosis codes over the study period (comorbidities) demonstrated an elevated comorbidity profile consistent with the clinical profile of DM. CONCLUSIONS: This study is among the first to utilize claims data to increase understanding of the clinical experience and health economic outcomes associated with DM. The markedly elevated HCRU patterns and comorbidity profile presented here add to the broad body of scientific and clinical knowledge on DM. These insights can inform clinical care and support the development of disease modifying and/or symptom-targeting therapies that address the multi-systemic, progressive nature of DM.


Subject(s)
Myotonic Dystrophy , Adult , Child, Preschool , Comorbidity , Delivery of Health Care , Health Care Costs , Humans , Insurance, Health , Retrospective Studies
8.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34291734

ABSTRACT

Mutations in HNRNPA1 encoding heterogeneous nuclear ribonucleoprotein (hnRNP) A1 are a rare cause of amyotrophic lateral sclerosis (ALS) and multisystem proteinopathy (MSP). hnRNPA1 is part of the group of RNA-binding proteins (RBPs) that assemble with RNA to form RNPs. hnRNPs are concentrated in the nucleus and function in pre-mRNA splicing, mRNA stability, and the regulation of transcription and translation. During stress, hnRNPs, mRNA, and other RBPs condense in the cytoplasm to form stress granules (SGs). SGs are implicated in the pathogenesis of (neuro-)degenerative diseases, including ALS and inclusion body myopathy (IBM). Mutations in RBPs that affect SG biology, including FUS, TDP-43, hnRNPA1, hnRNPA2B1, and TIA1, underlie ALS, IBM, and other neurodegenerative diseases. Here, we characterize 4 potentially novel HNRNPA1 mutations (yielding 3 protein variants: *321Eext*6, *321Qext*6, and G304Nfs*3) and 2 known HNRNPA1 mutations (P288A and D262V), previously connected to ALS and MSP, in a broad spectrum of patients with hereditary motor neuropathy, ALS, and myopathy. We establish that the mutations can have different effects on hnRNPA1 fibrillization, liquid-liquid phase separation, and SG dynamics. P288A accelerated fibrillization and decelerated SG disassembly, whereas *321Eext*6 had no effect on fibrillization but decelerated SG disassembly. By contrast, G304Nfs*3 decelerated fibrillization and impaired liquid phase separation. Our findings suggest different underlying pathomechanisms for HNRNPA1 mutations with a possible link to clinical phenotypes.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Muscular Atrophy, Spinal/genetics , Adolescent , Adult , Child , DNA Mutational Analysis , Female , Genetic Association Studies , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Heterozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Stress Granules/metabolism , Exome Sequencing , Young Adult
9.
Muscle Nerve ; 64(2): 219-224, 2021 08.
Article in English | MEDLINE | ID: mdl-34037996

ABSTRACT

INTRODUCTION/AIMS: We studied a patient with a congenital myasthenic syndrome (CMS) caused by a dominant mutation in the synaptotagmin 2 gene (SYT2) and compared the clinical features of this patient with those of a previously described patient with a recessive mutation in the same gene. METHODS: We performed electrodiagnostic (EDX) studies, genetic studies, muscle biopsy, microelectrode recordings and electron microscopy (EM). RESULTS: Both patients presented with muscle weakness and bulbar deficits, which were worse in the recessive form. EDX studies showed presynaptic failure, which was more prominent in the recessive form. Microelectrode studies in the dominant form showed a marked reduction of the quantal content, which increased linearly with higher frequencies of nerve stimulation. The MEPP frequencies were normal at rest but increased markedly with higher frequencies of nerve stimulation. The EM demonstrated overdeveloped postsynaptic folding, and abundant endosomes, multivesicular bodies and degenerative lamellar bodies inside small nerve terminals. DISCUSSION: The recessive form of CMS caused by a SYT2 mutation showed far more severe clinical manifestations than the dominant form. The pathogenesis of the dominant form likely involves a dominant-negative effect due to disruption of the dual function of synaptotagmin as a Ca2+ -sensor and modulator of synaptic vesicle exocytosis.


Subject(s)
Mutation/genetics , Myasthenic Syndromes, Congenital/genetics , Neuromuscular Junction/genetics , Synaptotagmin II/genetics , Adult , Child, Preschool , Female , Humans , Lambert-Eaton Myasthenic Syndrome/genetics , Lambert-Eaton Myasthenic Syndrome/physiopathology , Male , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Myasthenic Syndromes, Congenital/diagnosis , Neuromuscular Junction/physiopathology
10.
Cell Rep ; 34(3): 108634, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33472074

ABSTRACT

Myotonic dystrophy (DM) is caused by expanded CTG/CCTG repeats, causing symptoms in skeletal muscle, heart, and central nervous system (CNS). CNS issues are debilitating and include hypersomnolence, executive dysfunction, white matter atrophy, and neurofibrillary tangles. Here, we generate RNA-seq transcriptomes from DM and unaffected frontal cortex and identify 130 high-confidence splicing changes, most occurring only in cortex, not skeletal muscle or heart. Mis-spliced exons occur in neurotransmitter receptors, ion channels, and synaptic scaffolds, and GRIP1 mis-splicing modulates kinesin association. Optical mapping of expanded CTG repeats reveals extreme mosaicism, with some alleles showing >1,000 CTGs. Mis-splicing severity correlates with CTG repeat length across individuals. Upregulated genes tend to be microglial and endothelial, suggesting neuroinflammation, and downregulated genes tend to be neuronal. Many gene expression changes strongly correlate with mis-splicing, suggesting candidate biomarkers of disease. These findings provide a framework for mechanistic and therapeutic studies of the DM CNS.


Subject(s)
Frontal Lobe/physiopathology , Myotonic Dystrophy/genetics , Transcriptome/genetics , Humans
11.
J Comp Eff Res ; 9(14): 973-984, 2020 10.
Article in English | MEDLINE | ID: mdl-32851872

ABSTRACT

Aim: Assess the totality of efficacy evidence for ataluren in patients with nonsense mutation Duchenne muscular dystrophy (nmDMD). Materials & methods: Data from the two completed randomized controlled trials (ClinicalTrials.gov: NCT00592553; NCT01826487) of ataluren in nmDMD were combined to examine the intent-to-treat (ITT) populations and two patient subgroups (baseline 6-min walk distance [6MWD] ≥300-<400 or <400 m). Meta-analyses examined 6MWD change from baseline to week 48. Results: Statistically significant differences in 6MWD change with ataluren versus placebo were observed across all three meta-analyses. Least-squares mean difference (95% CI): ITT (n = 342), +17.2 (0.2-34.1) m, p = 0.0473; ≥300-<400 m (n = 143), +43.9 (18.2-69.6) m, p = 0.0008; <400 m (n = 216), +27.7 (6.4-49.0) m, p = 0.0109. Conclusion: These meta-analyses support previous evidence for ataluren in slowing disease progression versus placebo in patients with nmDMD over 48 weeks. Treatment benefit was most evident in patients with a baseline 6MWD ≥300-<400 m (the ambulatory transition phase), thereby informing future trial design.


Subject(s)
Codon, Nonsense/genetics , Muscular Dystrophy, Duchenne/drug therapy , Oxadiazoles/therapeutic use , Randomized Controlled Trials as Topic , Humans , Muscular Dystrophy, Duchenne/genetics
12.
Mol Genet Metab ; 130(1): 58-64, 2020 05.
Article in English | MEDLINE | ID: mdl-32173240

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a fatal disorder characterized by progressive gastrointestinal dysmotility, peripheral neuropathy, leukoencephalopathy, skeletal myopathy, ophthalmoparesis, and ptosis. MNGIE stems from deficient thymidine phosphorylase activity (TP) leading to toxic elevations of plasma thymidine. Hematopoietic stem cell transplant (HSCT) restores TP activity and halts disease progression but has high transplant-related morbidity and mortality. Liver transplant (LT) was reported to restore TP activity in two adult MNGIE patients. We report successful LT in four additional MNGIE patients, including a pediatric patient. Our patients were diagnosed between ages 14 months and 36 years with elevated thymidine levels and biallelic pathogenic variants in TYMP. Two patients presented with progressive gastrointestinal dysmotility, and three demonstrated progressive peripheral neuropathy with two suffering limitations in ambulation. Two patients, including the child, had liver dysfunction and cirrhosis. Following LT, thymidine levels nearly normalized in all four patients and remained low for the duration of follow-up. Disease symptoms stabilized in all patients, with some manifesting improvements, including intestinal function. No patient died, and LT appeared to have a more favorable safety profile than HSCT, especially when liver disease is present. Follow-up studies will need to document the long-term impact of this new approach on disease outcome. Take Home Message: Liver transplantation is effective in stabilizing symptoms and nearly normalizing thymidine levels in patients with mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) and may have an improved safety profile over hematopoietic stem cell transplant.


Subject(s)
Liver Transplantation/methods , Mitochondria/metabolism , Mitochondrial Encephalomyopathies/therapy , Thymidine Phosphorylase/genetics , Adolescent , Adult , Esophageal Motility Disorders/genetics , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Liver Transplantation/mortality , Magnetic Resonance Imaging , Male , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Encephalomyopathies/diagnostic imaging , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/physiopathology , Peripheral Nervous System Diseases/genetics , Thymidine/blood , Exome Sequencing
13.
Am J Hum Genet ; 100(4): 659-665, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28318499

ABSTRACT

Arthrogryposis multiplex congenita (AMC) is a developmental condition characterized by multiple joint contractures resulting from reduced or absent fetal movements. Through genetic mapping of disease loci and whole-exome sequencing in four unrelated multiplex families presenting with severe AMC, we identified biallelic loss-of-function mutations in LGI4 (leucine-rich glioma-inactivated 4). LGI4 is a ligand secreted by Schwann cells that regulates peripheral nerve myelination via its cognate receptor ADAM22 expressed by neurons. Immunolabeling experiments and transmission electron microscopy of the sciatic nerve from one of the affected individuals revealed a lack of myelin. Functional tests using affected individual-derived iPSCs showed that these germline mutations caused aberrant splicing of the endogenous LGI4 transcript and in a cell-based assay impaired the secretion of truncated LGI4 protein. This is consistent with previous studies reporting arthrogryposis in Lgi4-deficient mice due to peripheral hypomyelination. This study adds to the recent reports implicating defective axoglial function as a key cause of AMC.


Subject(s)
Arthrogryposis/genetics , Extracellular Matrix Proteins/genetics , Mutation , Schwann Cells/metabolism , Arthrogryposis/diagnosis , Arthrogryposis/pathology , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Myelin Sheath/metabolism , Nerve Tissue Proteins , Pedigree
16.
Orphanet J Rare Dis ; 10: 135, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26471370

ABSTRACT

BACKGROUND: Hereditary Fibrosing Poikiloderma (HFP) with tendon contractures, myopathy and pulmonary fibrosis (POIKTMP [MIM 615704]) is a very recently described entity of syndromic inherited poikiloderma. Previously by using whole exome sequencing in five families, we identified the causative gene, FAM111B (NM_198947.3), the function of which is still unknown. Our objective in this study was to better define the specific features of POIKTMP through a larger series of patients. METHODS: Clinical and molecular data of two families and eight independent sporadic cases, including six new cases, were collected. RESULTS: Key features consist of: (i) early-onset poikiloderma, hypotrichosis and hypohidrosis; (ii) multiple contractures, in particular triceps surae muscle contractures; (iii) diffuse progressive muscular weakness; (iv) pulmonary fibrosis in adulthood and (v) other features including exocrine pancreatic insufficiency, liver impairment and growth retardation. Muscle magnetic resonance imaging was informative and showed muscle atrophy and fatty infiltration. Histological examination of skeletal muscle revealed extensive fibroadipose tissue infiltration. Microscopy of the skin showed a scleroderma-like aspect with fibrosis and alterations of the elastic network. FAM111B gene analysis identified five different missense variants (two recurrent mutations were found respectively in three and four independent families). All the mutations were predicted to localize in the trypsin-like cysteine/serine peptidase domain of the protein. We suggest gain-of-function or dominant-negative mutations resulting in FAM111B enzymatic activity changes. CONCLUSIONS: HFP with tendon contractures, myopathy and pulmonary fibrosis, is a multisystemic disorder due to autosomal dominant FAM111B mutations. Future functional studies will help in understanding the specific pathological process of this fibrosing disorder.


Subject(s)
Cell Cycle Proteins/genetics , Contracture/genetics , Muscular Diseases/genetics , Pulmonary Fibrosis/genetics , Sclerosis/genetics , Skin Abnormalities/genetics , Skin Diseases, Genetic/genetics , Tendons/pathology , Adolescent , Adult , Amino Acid Sequence , Child , Child, Preschool , Contracture/complications , Contracture/diagnosis , Female , Humans , Infant , Male , Middle Aged , Molecular Sequence Data , Muscular Diseases/complications , Muscular Diseases/diagnosis , Mutation/genetics , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/diagnosis , Sclerosis/complications , Sclerosis/diagnosis , Skin Abnormalities/complications , Skin Abnormalities/diagnosis , Skin Diseases, Genetic/complications , Skin Diseases, Genetic/diagnosis
17.
Curr Neurol Neurosci Rep ; 15(9): 59, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26198888

ABSTRACT

This review article focuses on the cognitive profile associated with the C9orf72 gene with GGGGCC (G4C2) hexanucleotide repeat expansions that is commonly found in both familial and sporadic forms of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) in order to aid clinicians in the screening process. In this growing clinical continuum between FTD and ALS, understanding and recognizing a neurocognitive profile is important for diagnosis. Key features of this profile include executive dysfunction with memory impairment and language deficits as the disease progresses. Behaviorally, patients are prone to disinhibition, apathy, and psychosis. With the discovery of this mutation, studies have begun to characterize the different phenotypes associated with this mutation in terms of epidemiology, clinical presentation, imaging, and pathology. Greater awareness and increased surveillance for this mutation will benefit patients and their families in terms of access to genetic counseling, research studies, and improved understanding of the disease process.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Cognition , Frontotemporal Dementia/enzymology , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , C9orf72 Protein , Frontotemporal Dementia/genetics , Frontotemporal Dementia/physiopathology , Humans , Mutation , Phenotype , Proteins/metabolism
18.
Article in English | MEDLINE | ID: mdl-24809826

ABSTRACT

Abstract Sporadic amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no established biological marker. Recent observation of a reduced number of gems (survival motor neuron protein (SMN)-positive nuclear bodies) in cells from patients with familial ALS and the mouse models suggests an involvement of SMN in ALS pathology. At a molecular level, fused in sarcoma (FUS), one of the familial ALS-linked proteins, has been demonstrated to directly interact with SMN, while impaired nuclear localization of mutated FUS causes defective gem formation. Our objective was to determine whether gems and/or nuclear FUS levels in skin derived fibroblasts from sporadic ALS patients are consistently reduced and thus could constitute a novel and readily available biomarker of the disease. Fibroblasts from 20 patients and 17 age-matched healthy controls were cultured and co-immunostained for SMN and FUS. Results showed that no difference was detected between the two groups in the number of gems and in expression pattern of FUS. The number of gems negatively correlated with the age at biopsy in both ALS and control subjects. In conclusion, the expression pattern of SMN and FUS in fibroblasts cannot serve as a biomarker for sporadic ALS. Donor age-dependent gem reduction is a novel observation that links SMN with cellular senescence.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Nucleus/metabolism , Fibroblasts/ultrastructure , RNA-Binding Protein FUS/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Aged , Biopsy , Case-Control Studies , Cells, Cultured , Female , Fibroblasts/pathology , Humans , Male , Middle Aged , RNA-Binding Protein FUS/genetics , Skin/pathology , Statistics as Topic , Survival of Motor Neuron 1 Protein/genetics
19.
Neurohospitalist ; 2(3): 87-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23983869

ABSTRACT

OBJECTIVE: Our aim was to describe the clinical features of infective endocarditis (IE) in the acute stroke setting and outcomes following thrombolytic therapy. METHODS: This is a single-center, retrospective, descriptive case series of IE-related stroke (IES). Infective endocarditis diagnosis was based on the modified Duke criteria. RESULTS: From 2001 to 2007, 18 patients with acute stroke had definite or possible IE. Presenting stroke subtypes were: 11 ischemic stroke; 2 intracerebral hemorrhage; and 5 with a combination of ischemia and subarachnoid hemorrhage. On presentation, 6 had objective fever and 5 had subjective fever, 8 had heart murmur, and 3 had classic IE stigmata. The most common laboratory abnormalities were leukocytosis (n = 11) and anemia (n = 10). Sixteen patients had valvular vegetations on echocardiogram; 6 of 8 patients had vegetations visualized on transesophageal echocardiogram that were not detected by transthoracic echocardiogram. Two of the 3 patients with valve replacements had vegetations only on their native valves. Of 11 patients with pure ischemic stroke, 4 received thrombolytics and had hemorrhagic conversion. Overall mortality of IES was 56% (10 of 18). Mortality in pure ischemic IES patients was 29% (2 of 7, median National Institute of Health Stroke Scale [NIHSS] 13) in those not receiving thrombolytics and 75% (3 of 4, median NIHSS 14) in those receiving thrombolytics. CONCLUSIONS: Though diagnosis of IE in the acute stroke setting is difficult, features of the history, examination, and laboratory data may raise concern for IE. In this case series, thrombolytics in patients with IE-associated stroke were associated with very poor outcomes.

20.
Hum Mutat ; 32(3): 299-308, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21972111

ABSTRACT

Nonsense mutations are usually predicted to function as null alleles due to premature termination of protein translation. However, nonsense mutations in the DMD gene, encoding the dystrophin protein, have been associated with both the severe Duchenne Muscular Dystrophy (DMD) and milder Becker Muscular Dystrophy (BMD) phenotypes. In a large survey, we identified 243 unique nonsense mutations in the DMD gene, and for 210 of these we could establish definitive phenotypes. We analyzed the reading frame predicted by exons flanking those in which nonsense mutations were found, and present evidence that nonsense mutations resulting in BMD likely do so by inducing exon skipping, confirming that exonic point mutations affecting exon definition have played a significant role in determining phenotype. We present a new model based on the combination of exon definition and intronic splicing regulatory elements for the selective association of BMD nonsense mutations with a subset of DMD exons prone to mutation-induced exon skipping.


Subject(s)
Codon, Nonsense , Dystrophin/genetics , Exons , Muscular Dystrophy, Duchenne/genetics , RNA Splicing , Female , Humans , Male , Muscular Dystrophy, Duchenne/metabolism , Phenotype , RNA Splicing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...