Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 890
Filter
1.
Cells ; 13(4)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38391932

ABSTRACT

Cerebellar ataxias are a wide heterogeneous group of movement disorders. Within this broad umbrella of diseases, there are both genetics and sporadic forms. The clinical presentation of these conditions can exhibit a diverse range of symptoms across different age groups, spanning from pure cerebellar manifestations to sensory ataxia and multisystemic diseases. Over the last few decades, advancements in our understanding of genetics and molecular pathophysiology related to both dominant and recessive ataxias have propelled the field forward, paving the way for innovative therapeutic strategies aimed at preventing and arresting the progression of these diseases. Nevertheless, the rarity of certain forms of ataxia continues to pose challenges, leading to limited insights into the etiology of the disease and the identification of target pathways. Additionally, the lack of suitable models hampers efforts to comprehensively understand the molecular foundations of disease's pathophysiology and test novel therapeutic interventions. In the following review, we describe the epidemiology, symptomatology, and pathological progression of hereditary ataxia, including both the prevalent and less common forms of these diseases. Furthermore, we illustrate the diverse molecular pathways and therapeutic approaches currently undergoing investigation in both pre-clinical studies and clinical trials. Finally, we address the existing and anticipated challenges within this field, encompassing both basic research and clinical endeavors.


Subject(s)
Cerebellar Ataxia , Spinocerebellar Degenerations , Humans , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Ataxia/pathology , Cerebellum/pathology
2.
Mov Disord Clin Pract ; 11(1): 45-52, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38291837

ABSTRACT

BACKGROUND: RFC1-related disorder (RFC1/CANVAS) shares clinical features with other late-onset ataxias, such as spinocerebellar ataxias (SCA) and multiple system atrophy cerebellar type (MSA-C). Thinning of cranial nerves V (CNV) and VIII (CNVIII) has been reported in magnetic resonance imaging (MRI) scans of RFC1/CANVAS, but its specificity remains unclear. OBJECTIVES: To assess the usefulness of CNV and CNVIII thinning to differentiate RFC1/CANVAS from SCA and MSA-C. METHODS: Seventeen individuals with RFC1/CANVAS, 57 with SCA (types 2, 3 and 6), 11 with MSA-C and 15 healthy controls were enrolled. The Balanced Fast Field Echo sequence was used for assessment of cranial nerves. Images were reviewed by a neuroradiologist, who classified these nerves as atrophic or normal, and subsequently the CNV was segmented manually by an experienced neurologist. Both assessments were blinded to patient and clinical data. Non-parametric tests were used to assess between-group comparisons. RESULTS: Atrophy of CNV and CNVIII, both alone and in combination, was significantly more frequent in the RFC1/CANVAS group than in healthy controls and all other ataxia groups. Atrophy of CNV had the highest sensitivity (82%) and combined CNV and CNVIII atrophy had the best specificity (92%) for diagnosing RFC1/CANVAS. In the quantitative analyses, CNV was significantly thinner in the RFC1/CANVAS group relative to all other groups. The cutoff CNV diameter that best identified RFC1/CANVAS was ≤2.2 mm (AUC = 0.91; sensitivity 88.2%, specificity 95.6%). CONCLUSION: MRI evaluation of CNV and CNVIII using a dedicated sequence is an easy-to-use tool that helps to distinguish RFC1/CANVAS from SCA and MSA-C.


Subject(s)
Multiple System Atrophy , Spinocerebellar Ataxias , Humans , Ataxia/pathology , Atrophy/pathology , Cerebellum/pathology , Cranial Nerves/pathology , Multiple System Atrophy/diagnosis , Spinocerebellar Ataxias/diagnosis
3.
Clin Neuropsychol ; 38(1): 235-246, 2024 01.
Article in English | MEDLINE | ID: mdl-37021325

ABSTRACT

Objective: Hypertrophic Olivary Degeneration is a rare condition causing transneuronal degeneration of the inferior olivary nucleus. Symptoms manifest as progressively worsening palatal tremor, ataxia, and eye movement disturbances that plateau after several months. Though rarely documented in the literature of this specific condition, disconnection of the inferior olivary nucleus from the cerebellum, and cerebellar atrophy represent a pathway to developing subsequent cerebellar cognitive affective syndrome. The presented case documents the neuropsychological sequelae of a 39-year-old female with a history of hypertrophic olivary degeneration and symptoms of palatal tremor, opsoclonus myoclonus, ataxia, and delusions. Method: Review of the patient's medical records, interviews with the patient and her father, and a neuropsychological assessment battery were used to collect data. Review of currently published literature lent to case conceptualization. Results: Neuropsychological testing revealed deficits in executive functioning, attention, and language. An anomalous, fixed persecutory delusion was revealed. Conclusion: Hypertrophic olivary degeneration creates disconnection syndromes between the inferior olivary nucleus, red nucleus, and cerebellum. Late stages of the disorder cause atrophy of the inferior olivary nucleus and adjacent structures. While the motor sequela is well documented, the neuropsychological and psychiatric impact is infrequently discussed in existing literature. We present the first case to detail the neuropsychological sequelae of hypertrophic olivary degeneration and propose a mechanism for the development of cognitive impairment and psychotic features within this condition.


Subject(s)
Olivary Degeneration , Tremor , Female , Humans , Adult , Tremor/diagnosis , Tremor/etiology , Tremor/pathology , Olivary Nucleus/pathology , Neuropsychological Tests , Ataxia/complications , Ataxia/pathology , Atrophy/complications , Atrophy/pathology , Cognition , Magnetic Resonance Imaging
4.
Zebrafish ; 21(1): 28-38, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37603286

ABSTRACT

Zebrafish (Danio rerio) is a widely used vertebrate animal for modeling genetic diseases by targeted editing strategies followed by gross phenotypic and biomarker characterization. While larval transparency permits microscopic detection of anatomical defects, histological adult screening for organ-level defects remains invasive, tedious, inefficient, and subject to technical artifact. Here, we describe a noninvasive magnetic resonance imaging (MRI) approach to systematically screen adult zebrafish for anatomical growth defects. An anatomical atlas of wild-type (WT) zebrafish at 5-31 months post-fertilization was created by ex vivo MRI with a 9.4 T magnet. Volumetric growth over time was measured of animals and major organs, including the brain, spinal cord, heart, eyes, optic nerve, ear, liver, kidneys, and swim bladder. Subsequently, surf1-/-, fbxl4-/-, and opa1+/- mitochondrial disease mutant adult zebrafish were quantitatively studied to compare organ volumes with age-matched WT zebrafish. Results demonstrated that MRI enabled noninvasive, high-resolution, rapid screening of mutant adult zebrafish for overall and organ-specific growth abnormalities. Detailed volumetric analyses of three mitochondrial disease mutants delineated specific organ differences, including significantly increased brain growth in surf1-/- and opa1+/-, and marginally significant decreased heart and spinal cord volumes in surf1-/- mutants. This is interesting as we know neurological involvement can be seen in SURF1-/- patients with ataxia, dystonia, and lesions in basal ganglia, as well as in OPA1+/- patients with spasticity, ataxia, and hyperreflexia indicative of neuropathology. Similarly, cardiomyopathy is a known sequelae of cardiac pathology in patients with SURF1-/--related disease. Future studies will define MRI signaling patterns of organ dysfunction to further delineate specific pathology.


Subject(s)
Mitochondrial Diseases , Zebrafish , Animals , Zebrafish/genetics , Brain/diagnostic imaging , Mitochondrial Diseases/pathology , Magnetic Resonance Imaging , Ataxia/pathology
5.
Cytoskeleton (Hoboken) ; 81(1): 66-70, 2024 01.
Article in English | MEDLINE | ID: mdl-37795931

ABSTRACT

Ageing-associated tauopathies like frontotemporal dementia (FTD), variants thereof (like progressive supranuclear palsy (PSP), pick diseases (PiD), corticobasal degeneration (CBD)), and of course the most prevalent form of dementia, Alzheimer Disease (AD), are widely recognized forms of tauopathies. The list of tauopathies is expanding. We now include: (i) tauopathies where the disease cause or trigger is clearly either physical, such as in Traumatic Brain Injury (TBI) or Chronic Traumatic Encephalopathy (CTE), and (ii) genetic diseases that result in tauopathy but have pathogenic genetic variants in genes not related to TAU. Examples of the latter are myotonic dystrophy Type 1 and Type 2 (DM1, DM2, due to pathogenic genetic variants in the genes DMPK and CNBP, respectively), Niemann-Pick Disease Type C (NPD, due to mutations in NPC1 or NPC2), Kufs Disease (CLN6), Christianson Syndrome (SLC9A6), familial forms of Parkinson Disease (PD), and many others. In terms of affected brain regions and cell types, intracellular distribution of TAU pathology/aggregates, age of disease onset, velocity of disease progression and spreading of TAU pathology, there is, however, little in common in most of these disease entities. Here, I reason that TAU/MAPT is causative for the minority of tauopathies (e.g., MAPT-related FTD/PSP and Vacuolar Tauopathy (VCP)) and a critical mediator for others, like shown by overwhelming evidence for AD. However, TAU may also be a mere bystander or even protective in other settings. Improved understanding of rare tauopathies is necessary to develop specific treatments, but also to improve our understanding of the pathomechanistic role of TAU and to identify diseases that may profit from TAU-based therapies.


Subject(s)
Frontotemporal Dementia , Tauopathies , Humans , tau Proteins/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , Brain/metabolism , Ataxia/metabolism , Ataxia/pathology , Membrane Proteins/metabolism
6.
J Child Neurol ; 38(13-14): 659-664, 2023 12.
Article in English | MEDLINE | ID: mdl-37981797

ABSTRACT

Introduction: We describe 5 children with GFAP astrocytopathy with the goal of further characterizing this rare form of meningoencephalomyelitis. Methods: Retrospective chart review of patients diagnosed with GFAP astrocytopathy between 2019 and 2021. Results: Patients were 8-17 years old, and all were male. Fever, headache, and vomiting were common presenting symptoms, and weakness, tremor, and ataxia were common initial examination findings. Initial magnetic resonance imaging (MRI) showed spinal cord abnormalities in 2 patients and leptomeningeal enhancement in 1. Most patients had cerebral spinal fluid pleocytosis, and all screened negative for malignancy. Three patients progressed to coma, and all were treated with immunosuppressant therapy. By discharge, all patients had improved over their clinical nadir, although none had returned to baseline. Discussion: GFAP astrocytopathy is a recently recognized cause of meningoencephalomyelitis in children. Here, we expand our understanding of this entity with the goal of aiding those treating children with GFAP astrocytopathy.


Subject(s)
Astrocytes , Magnetic Resonance Imaging , Adolescent , Child , Humans , Male , Astrocytes/metabolism , Astrocytes/pathology , Ataxia/pathology , Autoantibodies , Glial Fibrillary Acidic Protein , Retrospective Studies
7.
Cells ; 12(19)2023 09 26.
Article in English | MEDLINE | ID: mdl-37830578

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that appears in adult FMR1 premutation carriers. The neuropathological hallmark of FXTAS is an intranuclear inclusion in neurons and astrocytes. Nearly 200 different proteins have been identified in FXTAS inclusions, being the small ubiquitin-related modifier 2 (SUMO2), ubiquitin and p62 the most highly abundant. These proteins are components of the protein degradation machinery. This study aimed to characterize SUMO2/3 expression levels and autophagy process in human postmortem brain samples and skin fibroblast cultures from FXTAS patients. Results revealed that FXTAS postmortem brain samples are positive for SUMO2/3 conjugates and supported the idea that SUMO2/3 accumulation is involved in inclusion formation. Insights from RNA-sequencing data indicated that SUMOylation processes are significantly upregulated in FXTAS samples. In addition, the analysis of the autophagy flux showed the accumulation of p62 protein levels and autophagosomes in skin fibroblasts from FXTAS patients. Similarly, gene set analysis evidenced a significant downregulation in gene ontology terms related to autophagy in FXTAS samples. Overall, this study provides new evidence supporting the role of SUMOylation and autophagic processes in the pathogenic mechanisms underlying FXTAS.


Subject(s)
Fragile X Syndrome , Tremor , Adult , Humans , Tremor/genetics , Tremor/metabolism , Tremor/pathology , Ubiquitin/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/pathology , Ataxia/pathology , Autophagy , Small Ubiquitin-Related Modifier Proteins/metabolism
8.
Emerg Top Life Sci ; 7(3): 289-312, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-37668011

ABSTRACT

Spinocerebellar ataxias (SCAs) are a heterogenous group of neurodegenerative disorders which commonly inherited in an autosomal dominant manner. They cause muscle incoordination due to degeneration of the cerebellum and other parts of nervous system. Out of all the characterized (>50) SCAs, 14 SCAs are caused due to microsatellite repeat expansion mutations. Repeat expansions can result in toxic protein gain-of-function, protein loss-of-function, and/or RNA gain-of-function effects. The location and the nature of mutation modulate the underlying disease pathophysiology resulting in varying disease manifestations. Potential toxic effects of these mutations likely affect key major cellular processes such as transcriptional regulation, mitochondrial functioning, ion channel dysfunction and synaptic transmission. Involvement of several common pathways suggests interlinked function of genes implicated in the disease pathogenesis. A better understanding of the shared and distinct molecular pathogenic mechanisms in these diseases is required to develop targeted therapeutic tools and interventions for disease management. The prime focus of this review is to elaborate on how expanded 'CAG' repeats contribute to the common modes of neurotoxicity and their possible therapeutic targets in management of such devastating disorders.


Subject(s)
DNA Repeat Expansion , Spinocerebellar Ataxias , Humans , DNA Repeat Expansion/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Cerebellum/pathology , Ataxia/genetics , Ataxia/pathology , Proteins
9.
Glia ; 71(12): 2832-2849, 2023 12.
Article in English | MEDLINE | ID: mdl-37610133

ABSTRACT

Canavan disease (CD) is a recessively inherited pediatric leukodystrophy resulting from inactivating mutations to the oligodendroglial enzyme aspartoacylase (ASPA). ASPA is responsible for hydrolyzing the amino acid derivative N-acetyl-L-aspartate (NAA), and without it, brain NAA concentrations increase by 50% or more. Infants and children with CD present with progressive cognitive and motor delays, cytotoxic edema, astroglial vacuolation, and prominent spongiform brain degeneration. ASPA-deficient CD mice (Aspanur7/nur7 ) present similarly with elevated NAA, widespread astroglial dysfunction, ataxia, and Purkinje cell (PC) dendritic atrophy. Bergmann glia (BG), radial astrocytes essential for cerebellar development, are intimately intertwined with PCs, where they regulate synapse stability, functionality, and plasticity. BG damage is common to many neurodegenerative conditions and frequently associated with PC dysfunction and ataxia. Here, we report that, in CD mice, BG exhibit significant morphological alterations, decreased structural associations with PCs, loss of synaptic support proteins, and altered calcium dynamics. We also find that BG dysfunction predates cerebellar vacuolation and PC damage in CD mice. Previously, we developed an antisense oligonucleotide (ASO) therapy targeting Nat8l (N-acetyltransferase-8-like, "Nat8l ASO") that inhibits the production of NAA and reverses ataxia and PC atrophy in CD mice. Here, we show that Nat8l ASO administration in adult CD mice also leads to BG repair. Furthermore, blocking astroglial uptake of NAA is neuroprotective in astroglia-neuron cocultures exposed to elevated NAA. Our findings suggest that restoration of BG structural and functional integrity could be a mechanism for PC regeneration and improved motor function.


Subject(s)
Canavan Disease , Neurodegenerative Diseases , Humans , Child , Infant , Mice , Animals , Canavan Disease/genetics , Canavan Disease/metabolism , Canavan Disease/pathology , Calcium , Ataxia/pathology , Oligodendroglia/metabolism , Neurodegenerative Diseases/pathology , Aspartic Acid , Atrophy/complications , Atrophy/pathology
10.
Cells ; 12(10)2023 05 13.
Article in English | MEDLINE | ID: mdl-37408217

ABSTRACT

Dominantly inherited missense mutations of the KCNA1 gene, which encodes the KV1.1 potassium channel subunit, cause Episodic Ataxia type 1 (EA1). Although the cerebellar incoordination is thought to arise from abnormal Purkinje cell output, the underlying functional deficit remains unclear. Here we examine synaptic and non-synaptic inhibition of Purkinje cells by cerebellar basket cells in an adult mouse model of EA1. The synaptic function of basket cell terminals was unaffected, despite their intense enrichment for KV1.1-containing channels. In turn, the phase response curve quantifying the influence of basket cell input on Purkine cell output was maintained. However, ultra-fast non-synaptic ephaptic coupling, which occurs in the cerebellar 'pinceau' formation surrounding the axon initial segment of Purkinje cells, was profoundly reduced in EA1 mice in comparison with their wild type littermates. The altered temporal profile of basket cell inhibition of Purkinje cells underlines the importance of Kv1.1 channels for this form of signalling, and may contribute to the clinical phenotype of EA1.


Subject(s)
Ataxia , Kv1.1 Potassium Channel , Myokymia , Neural Inhibition , Purkinje Cells , Purkinje Cells/metabolism , Purkinje Cells/pathology , Animals , Mice , Disease Models, Animal , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Synapses/physiology , Cell Communication , Synaptic Transmission , Ataxia/genetics , Ataxia/pathology , Ataxia/physiopathology , Myokymia/genetics , Myokymia/pathology , Myokymia/physiopathology , Evoked Potentials , Mice, Inbred C57BL , Male , Female
11.
Mult Scler ; 29(9): 1195-1198, 2023 08.
Article in English | MEDLINE | ID: mdl-37401111

ABSTRACT

Cerebellar dysfunction is likely to cause severe and treatment-resistant disability in multiple sclerosis (MS). Certain spinocerebellar ataxia (SCA)-related alleles can increase MS susceptibility, and channel polymorphisms can impact disability measures. Following an index patient with the coexistence of MS and SCA Type-8 (SCA8) in the MS clinic, an institutional engine search for MS and hereditary ataxia coexistence was conducted but did not reveal any other cases. This extremely rare coexistence of MS and SCA8 in our index patient may be incidental; however, a yet-to-be-identified contribution of coexistent hereditary ataxia(s) to the susceptibility of a prominent progressive ataxia MS phenotype cannot be ruled out.


Subject(s)
Multiple Sclerosis , Spinocerebellar Degenerations , Multiple Sclerosis/complications , Multiple Sclerosis/diagnosis , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Spinocerebellar Degenerations/complications , Spinocerebellar Degenerations/diagnosis , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/pathology , Humans , Male , Adult , Ataxia/genetics , Ataxia/pathology , Brain/pathology , Spinal Cord
12.
Cells ; 12(14)2023 07 20.
Article in English | MEDLINE | ID: mdl-37508562

ABSTRACT

Brain changes at the end-stage of fragile X-associated tremor/ataxia syndrome (FXTAS) are largely unknown due to mobility impairment. We conducted a postmortem MRI study of FXTAS to quantify cerebrovascular disease, brain atrophy and iron content, and examined their relationships using principal component analysis (PCA). Intracranial hemorrhage (ICH) was observed in 4/17 FXTAS cases, among which one was confirmed by histologic staining. Compared with seven control brains, FXTAS cases showed higher ratings of T2-hyperintensities (indicating cerebral small vessel disease) in the cerebellum, globus pallidus and frontoparietal white matter, and significant atrophy in the cerebellar white matter, red nucleus and dentate nucleus. PCA of FXTAS cases revealed negative associations of T2-hyperintensity ratings with anatomic volumes and iron content in the white matter, hippocampus and amygdala, that were independent from a highly correlated number of regions with ICH and iron content in subcortical nuclei. Post-hoc analysis confirmed PCA findings and further revealed increased iron content in the white matter, hippocampus and amygdala in FXTAS cases compared to controls, after adjusting for T2-hyperintensity ratings. These findings indicate that both ischemic and hemorrhagic brain damage may occur in FXTAS, with the former being marked by demyelination/iron depletion and atrophy, and the latter by ICH and iron accumulation in basal ganglia.


Subject(s)
Cerebrovascular Disorders , Fragile X Syndrome , Humans , Tremor/diagnostic imaging , Tremor/pathology , Iron , Ataxia/diagnostic imaging , Ataxia/pathology , Fragile X Syndrome/diagnostic imaging , Fragile X Syndrome/pathology , Magnetic Resonance Imaging , Atrophy
13.
Brain Nerve ; 75(6): 737-747, 2023 Jun.
Article in Japanese | MEDLINE | ID: mdl-37287357

ABSTRACT

Autoimmune mechanisms affect the cerebellum leading to the development of cerebellar ataxias (CAs), which are termed immune-mediated cerebellar ataxias (IMCAs). IMCAs have diverse etiologies. Gluten ataxia (GA), post-infectious cerebellitis (PIC), paraneoplastic cerebellar degeneration (PCD), opsoclonus myoclonus syndrome (OMS), anti-glutamate decarboxylase 65 antibody-associated CA (anti-GAD ataxia), and primary autoimmune cerebellar ataxia (PACA). In addition to these well-established entities, CAs are associated with autoimmunity against ion channels and their related proteins, synaptic adhesion proteins, transmitter receptors, glial cells, and brainstem antigens. Cell-mediated mechanisms are assumed to be involved in PCD, whereas accumulating evidence shows that anti-GAD antibodies decrease gamma-aminobutyric acid (GABA) release to elicit functional synaptic deficits. The therapeutic benefits of immunotherapies vary depending on the etiology. Early intervention is recommended when the cerebellar reserve, abilities for compensation and restoration of pathologies are preserved.


Subject(s)
Cerebellar Ataxia , Humans , Cerebellum , Ataxia/pathology , Antibodies , Autoantibodies
14.
Optom Vis Sci ; 100(6): 419-421, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37200199

ABSTRACT

SIGNIFICANCE: Unilateral gaze-evoked nystagmus is a rare neurologic finding that is largely diagnosed in connection with ischemic stroke. Gazed-evoked nystagmus is also a rare initial presentation of multiple sclerosis. PURPOSE: This study aimed to report a rare presentation of gaze-evoked nystagmus in a patient with multiple sclerosis and review the mechanism underlying the gaze-evoked nystagmus. CASE REPORT: A 32-year-old man presented with a 1-week history of diplopia. Neurologic examination revealed right-sided gaze-evoked nystagmus and right-sided ataxia. Laboratory test revealed a positive result for oligoclonal bands. Contrast brain MRI revealed multiple hyperintense T2 lesions including a hyperintense patch at the right inferior cerebellar peduncle. A diagnosis of multiple sclerosis was made. The patient received methylprednisolone 500 mg intravenously for 14 days. The diplopia and gaze-evoked nystagmus resolved and remained stable 2 months later. CONCLUSIONS: Our case demonstrates that damage to the inferior cerebellar peduncle may result in ipsilesional gaze-evoked nystagmus and ipsilesional ataxia, in contrast to ipsilesional gaze-evoked nystagmus and contralesional ataxia.


Subject(s)
Multiple Sclerosis , Nystagmus, Pathologic , Male , Humans , Adult , Multiple Sclerosis/complications , Multiple Sclerosis/diagnosis , Diplopia/diagnosis , Diplopia/etiology , Nystagmus, Pathologic/diagnosis , Nystagmus, Pathologic/etiology , Cerebellum/pathology , Ataxia/pathology
15.
Proc Natl Acad Sci U S A ; 120(23): e2300052120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252957

ABSTRACT

Short trinucleotide expansions at the FMR1 locus are associated with the late-onset condition fragile X-associated tremor/ataxia syndrome (FXTAS), which shows very different clinical and pathological features from fragile X syndrome (associated with longer expansions), with no clear molecular explanation for these marked differences. One prevailing theory posits that the shorter, premutation expansion uniquely causes extreme neurotoxic increases in FMR1 mRNA (i.e., four to eightfold increases), but evidence to support this hypothesis is largely derived from analysis of peripheral blood. We applied single-nucleus RNA sequencing to postmortem frontal cortex and cerebellum from 7 individuals with premutation and matched controls (n = 6) to assess cell type-specific molecular neuropathology. We found only modest upregulation (~1.3-fold) of FMR1 in some glial populations associated with premutation expansions. In premutation cases, we also identified decreased astrocyte proportions in the cortex. Differential expression and gene ontology analysis demonstrated altered neuroregulatory roles of glia. Using network analyses, we identified cell type-specific and region-specific patterns of FMR1 protein target gene dysregulation unique to premutation cases, with notable network dysregulation in the cortical oligodendrocyte lineage. We used pseudotime trajectory analysis to determine how oligodendrocyte development was altered and identified differences in early gene expression in oligodendrocyte trajectories in premutation cases specifically, implicating early cortical glial developmental perturbations. These findings challenge dogma regarding extremely elevated FMR1 increases in FXTAS and implicate glial dysregulation as a critical facet of premutation pathophysiology, representing potential unique therapeutic targets directly derived from the human condition.


Subject(s)
Fragile X Syndrome , Humans , Fragile X Syndrome/pathology , Tremor/genetics , Trinucleotide Repeat Expansion , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Ataxia/genetics , Ataxia/pathology , Brain/metabolism , Astrocytes/metabolism
16.
Turk J Gastroenterol ; 34(3): 182-195, 2023 03.
Article in English | MEDLINE | ID: mdl-36919830

ABSTRACT

About one-third of chronically constipated patients have an evacuation disorder, and dyssynergic defecation is a common cause of the evacuation disorder. In dyssynergic defecation, the coordination between abdominal and pelvic floor muscles during defecation is disrupted and patients cannot produce a normal bowel movement. The etiology of dyssynergic defecation is still unknown. Although a detailed history taking and a careful examination including digital rectal examination could be useful, other modalities such as anorectal manometry and balloon expulsion test are necessary for the diagnosis. Biofeedback therapy is one of the most effective and safe treatments. Here, we provide an overview of dyssynergic defecation as well as how to diagnose and manage this condition.


Subject(s)
Anal Canal , Constipation , Defecation , Humans , Constipation/diagnosis , Constipation/etiology , Constipation/therapy , Manometry , Biofeedback, Psychology , Digital Rectal Examination , Ataxia/pathology
17.
Ann Clin Transl Neurol ; 10(5): 706-718, 2023 05.
Article in English | MEDLINE | ID: mdl-36932648

ABSTRACT

OBJECTIVE: The co-morbidity of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP)/autoimmune nodopathies with nephropathy has been gradually known in recent years. This study was intended to explore the clinical, serological and neuropathological features of seven patients with CIDP/autoimmune nodopathies and nephropathy. METHODS: Among 83 CIDP patients, seven were identified with nephropathy. Their clinical, electrophysiological and laboratory examination data were collected. The nodal/paranodal antibodies were tested. The sural biopsies were performed in all the patients, and renal biopsies were operated in 6 patients. RESULTS: Six patients had chronic onsets and one had an acute onset. Four patients exhibited peripheral neuropathy preceding nephropathy while two showed concurrent onset of neuropathy and nephropathy, and one started with nephropathy. All the patients showed demyelination in electrophysiological examination. Nerve biopsies showed mild to moderate mixed neuropathies including demyelinating and axonal changes in all patients. Renal biopsies showed membranous nephropathy in all 6 patients. Immunotherapy was effective in all patients, with two patients showing good response to corticosteroid treatment alone. Four of the patients were positive to anti-CNTN1 antibody. Compared with anti-CNTN1 antibody-negative patients, antibody-positive patients had a higher proportion of ataxia (3/4 vs. 1/3), autonomic dysfunction (3/4 vs. 1/3), less frequent antecedent infections (1/4 vs. 2/3), higher cerebrospinal fluid proteins (3.2 g/L vs. 1.69 g/L), more frequent conduction block on electrophysiological examination (3/4 vs. 1/3), higher myelinated nerve fiber density, and positive CNTN1 expression in the glomeruli of kidney tissues. CONCLUSION: Anti-CNTN1 antibody was the most frequent antibody in this group of patients with CIDP/autoimmune nodopathies and nephropathy. Our study suggested that there might be some clinical and pathological differences between the antibody positive and negative patients.


Subject(s)
Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Axons/pathology , Nerve Fibers, Myelinated/pathology , Adrenal Cortex Hormones , Ataxia/pathology
18.
Biol Pharm Bull ; 46(2): 139-146, 2023.
Article in English | MEDLINE | ID: mdl-36724941

ABSTRACT

Repeat-associated non-AUG (RAN) translation is a pathogenic mechanism in which repetitive sequences are translated into aggregation-prone proteins from multiple reading frames, even without a canonical AUG start codon. Since its discovery in spinocerebellar ataxia type 8 (SCA8) and myotonic dystrophy type 1 (DM1), RAN translation is now known to occur in the context of 12 disease-linked repeat expansions. This review discusses recent advances in understanding the regulatory mechanisms controlling RAN translation and its contribution to the pathophysiology of repeat expansion diseases. We discuss the key findings in the context of Fragile X Tremor Ataxia Syndrome (FXTAS), a neurodegenerative disorder caused by a CGG repeat expansion in the 5' untranslated region of FMR1.


Subject(s)
Fragile X Syndrome , Neurodegenerative Diseases , Humans , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Ataxia/metabolism , Ataxia/pathology , Tremor/genetics , Tremor/metabolism , Tremor/pathology
19.
Nihon Yakurigaku Zasshi ; 158(1): 30-33, 2023.
Article in Japanese | MEDLINE | ID: mdl-36596485

ABSTRACT

Repeat expansion diseases are caused by the aberrant repeat expansions within specific genes. RNAs derived from aberrant repeat sequences form non-canonical secondary structures, contributing to induce cell toxicity. In particular, RNA G-quadruplexes (G4RNAs) formed in guanine-rich repeat expanded RNAs trigger neurodegeneration. We have previously shown that the expanded CGG repeat-derived G4RNAs initiate aggregation of FMRpolyG, a neuropathogenic protein generated by repeat-associated non-AUG (RAN) translation in Fragile X-associated tremor/ataxia syndrome (FXTAS). In this review, we describe the neuropathological mechanism attributed to G4RNAs in guanine-rich repeat expansion diseases, including FXTAS.


Subject(s)
Fragile X Syndrome , Trinucleotide Repeat Expansion , Humans , Trinucleotide Repeat Expansion/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , RNA
20.
Cerebellum ; 22(6): 1137-1151, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36219306

ABSTRACT

Hom ozygous variants in the peptidyl-tRNA hydrolase 2 gene (PTRH2) cause infantile-onset multisystem neurologic, endocrine, and pancreatic disease. The objective is to delineate the mechanisms underlying the core cerebellar phenotype in this disease. For this, we generated constitutive (Ptrh2LoxPxhCMVCre, Ptrh2-/- mice) and Purkinje cell (PC) specific (Ptrh2LoxPxPcp2Cre, Ptrh2ΔPCmice) Ptrh2 mutant mouse models and investigated the effect of the loss of Ptrh2 on cerebellar development. We show that Ptrh2-/- knockout mice had severe postnatal runting and lethality by postnatal day 14. Ptrh2ΔPC PC specific knockout mice survived until adult age; however, they showed progressive cerebellar atrophy and functional cerebellar deficits with abnormal gait and ataxia. PCs of Ptrh2ΔPC mice had reduced cell size and density, stunted dendrites, and lower levels of ribosomal protein S6, a readout of the mammalian target of rapamycin pathway. By adulthood, there was a marked loss of PCs. Thus, we identify a cell autonomous requirement for PTRH2 in PC maturation and survival. Loss of PTRH2 in PCs leads to downregulation of the mTOR pathway and PC atrophy. This suggests a molecular mechanism underlying the ataxia and cerebellar atrophy seen in patients with PTRH2 mutations leading to infantile-onset multisystem neurologic, endocrine, and pancreatic disease.


Subject(s)
Cerebellar Ataxia , Pancreatic Diseases , Humans , Mice , Animals , Adult , Ataxia/pathology , Purkinje Cells/physiology , Mice, Knockout , Pancreatic Diseases/genetics , Pancreatic Diseases/metabolism , Pancreatic Diseases/pathology , Cell Differentiation , Atrophy/pathology , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL
...