Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.420
Filter
1.
Mol Genet Genomic Med ; 12(8): e2505, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39108195

ABSTRACT

BACKGROUND: Biallelic variants in the major facilitator superfamily domain containing 8 gene (MFSD8) are associated with distinct clinical presentations that range from typical late-infantile neuronal ceroid lipofuscinosis type 7 (CLN7 disease) to isolated adult-onset retinal dystrophy. Classic late-infantile CLN7 disease is a severe, rare neurological disorder with an age of onset typically between 2 and 6 years, presenting with seizures and/or cognitive regression. Its clinical course is progressive, leading to premature death, and often includes visual loss due to severe retinal dystrophy. In rare cases, pathogenic variants in MFSD8 can be associated with isolated non-syndromic macular dystrophy with variable age at onset, in which the disease process predominantly or exclusively affects the cones of the macula and where there are no neurological or neuropsychiatric manifestations. METHODS: Here we present longitudinal studies on four adult-onset patients who were biallelic for four MFSD8 variants. RESULTS: Two unrelated patients who presented with adult-onset ataxia and had macular dystrophy on examination were homozygous for a novel variant in MFSD8 NM_152778.4: c.935T>C p.(Ile312Thr). Two other patients presented in adulthood with visual symptoms, and one of these developed mild to moderate cerebellar ataxia years after the onset of visual symptoms. CONCLUSIONS: Our observations expand the knowledge on biallelic pathogenic MFSD8 variants and confirm that these are associated with a spectrum of more heterogeneous clinical phenotypes. In MFSD8-related disease, adult-onset recessive ataxia can be the presenting manifestation or may occur in combination with retinal dystrophy.


Subject(s)
Macular Degeneration , Humans , Adult , Male , Female , Macular Degeneration/genetics , Macular Degeneration/pathology , Age of Onset , Ataxia/genetics , Ataxia/pathology , Alleles , Middle Aged , Mutation , Membrane Transport Proteins/genetics , Phenotype
2.
Arq Neuropsiquiatr ; 82(8): 1-2, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39117345

Subject(s)
Ataxia , Humans , Brazil , Ataxia/etiology
3.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125677

ABSTRACT

In this study, the potential role and interaction of the APOε and KLOTHO genes on the penetrance of fragile X-associated tremor/ataxia syndrome (FXTAS) and on the IQ trajectory were investigated. FXTAS was diagnosed based on molecular, clinical and radiological criteria. Males with the premutation (PM) over 50 years, 165 with and 34 without an FXTAS diagnosis, were included in this study and were compared based on their APO (ε2-ε3-ε4) and KLOTHO variant (KL-VS) genotypes. The effect of APOε4 on FXTAS stage and on diagnosis did not differ significantly by KL-VS genotype with interaction effect p = 0.662 and p = 0.91, respectively. In the FXTAS individuals with an APOε2 allele, a marginal significance was observed towards a larger decline in verbal IQ (VIQ) in individuals with an APOε4 allele compared to those without an APOε4 allele (p = 0.071). In conclusion, our findings suggest that the APOε4 and KL-VS genotypes alone or through their interaction effect do not appear to predispose to either FXTAS diagnosis or stage in male carriers of the PM allele. A further study is needed to establish the trend of IQ decline in the FXTAS individuals who carry APOε4 with APOε2 compared to those without APOε4.


Subject(s)
Ataxia , Fragile X Syndrome , Glucuronidase , Klotho Proteins , Tremor , Humans , Male , Tremor/genetics , Fragile X Syndrome/genetics , Ataxia/genetics , Aged , Middle Aged , Glucuronidase/genetics , Apolipoproteins E/genetics , Penetrance , Genotype , Alleles , Aged, 80 and over , Genetic Predisposition to Disease
4.
BMC Med Genomics ; 17(1): 203, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123271

ABSTRACT

BACKGROUND: A comprehensive understanding of the genetic basis of rare diseases and their regulatory mechanisms is essential for human molecular genetics. However, the genetic mutant spectrum of pathogenic genes within the Chinese population remains underrepresented. Here, we reported previously unreported functional ABHD12 variants in two Chinese families and explored the correlation between genetic polymorphisms and phenotypes linked to PHARC syndrome. METHODS: Participants with biallelic pathogenic ABHD12 variants were recruited from the Chinese Deafness Genetics Cohort. These participants underwent whole-genome sequencing. Subsequently, a comprehensive literature review was conducted. RESULTS: Two Han Chinese families were identified, one with a compound heterozygous variant and the other with a novel homozygous variant in ABHD12. Among 65 PHARC patients, including 62 from the literature and 3 from this study, approximately 90% (57 out of 63) exhibited hearing loss, 82% (50 out of 61) had cataracts, 82% (46 out of 56) presented with retinitis pigmentosa, 79% (42 out of 53) experienced polyneuropathy, and 63% (36 out of 57) displayed ataxia. Seventeen different patterns were observed in the five main phenotypes of PHARC syndrome. A total of 33 pathogenic variants were identified in the ABHD12. Compared with other genotypes, individuals with biallelic truncating variants showed a higher incidence of polyneuropathy (p = 0.006), but no statistically significant differences were observed in the incidence of hearing loss, ataxia, retinitis pigmentosa and cataracts. CONCLUSIONS: The diagnosis of PHARC syndrome is challenging because of its genetic heterogeneity. Therefore, exploring novel variants and establishing genotype-phenotype correlations can significantly enhance gene diagnosis and genetic counseling for this complex disease.


Subject(s)
Ataxia , Cataract , Genetic Association Studies , Monoacylglycerol Lipases , Pedigree , Phenotype , Polyneuropathies , Retinitis Pigmentosa , Humans , Male , Female , Ataxia/genetics , Cataract/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Polyneuropathies/genetics , Monoacylglycerol Lipases/genetics , Mutation , Adult , Child , Adolescent , Genotype
5.
Medicine (Baltimore) ; 103(29): e38966, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39029081

ABSTRACT

RATIONALE: Neuromyelitis optica spectrum disorder (NMOSD) involves autoimmune and inflammatory responses in the central nervous system, primarily affecting the optic nerves and spinal cord. Atypical presentations such as ataxia and syncope complicate the diagnosis, and lesions in the medulla are easily mistaken for cerebral infarction. This case report emphasizes the need to recognize such manifestations to avoid misdiagnosis and ensure timely treatment. PATIENT CONCERNS: This case report presents an NMOSD female patient who experienced ataxia, syncope, and neuropathic pain during her illness. DIAGNOSIS: NMOSD. INTERVENTIONS: The patient managed her blood sugar with insulin, controlled neuropathic pain with pregabalin, and underwent 5 plasma exchanges. OUTCOMES: Significant improvement was noted 1 week post-plasma exchange, with complete resolution of neuropathic pain and no symptom recurrence reported at 6-month follow-up. LESSONS: Atypical manifestations of NMOSD, such as ataxia, syncope, and trigeminal neuralgia, increase diagnostic difficulty. Recognizing these symptoms is crucial to avoid misdiagnosis and ensure timely and appropriate treatment for patients.


Subject(s)
Ataxia , Neuralgia , Neuromyelitis Optica , Syncope , Humans , Female , Neuromyelitis Optica/complications , Neuromyelitis Optica/diagnosis , Ataxia/diagnosis , Ataxia/etiology , Syncope/etiology , Syncope/diagnosis , Neuralgia/etiology , Neuralgia/diagnosis , Disease Progression , Adult , Middle Aged , Plasma Exchange/methods
6.
Orphanet J Rare Dis ; 19(1): 264, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997701

ABSTRACT

BACKGROUND AND OBJECTIVES: Fragile X Syndrome (FXS) is the most common cause of inherited intellectual disability, caused by CGG-repeat expansions (> 200) in the FMR1 gene leading to lack of expression. Espansion between 55 and 200 triplets fall within the premutation range (PM) and can lead to different clinical conditions, including fragile X- primary ovarian insufficiency (FXPOI), fragile X-associated neuropsychiatric disorders (FXAND) and fragile X-associated tremor/ataxia syndrome (FXTAS). Although there is not a current cure for FXS and for the Fragile X-PM associated conditions (FXPAC), timely diagnosis as well as the implementation of treatment strategies, psychoeducation and behavioral intervention may improve the quality of life (QoL) of people with FXS or FXPAC. With the aim to investigate the main areas of concerns and the priorities of treatment in these populations, the Italian National Fragile X Association in collaboration with Bambino Gesù Children's Hospital, conducted a survey among Italian participants. METHOD: Here, we present a survey based on the previous study that Weber and colleagues conducted in 2019 and that aimed to investigate the main symptoms and challenges in American individuals with FXS. The survey has been translated into Italian language to explore FXS needs of treatment also among Italian individuals affected by FXS, family members, caretakers, and professionals. Furthermore, we added a section designated only to people with PM, to investigate the main symptoms, daily living challenges and treatment priorities. RESULTS: Anxiety, challenging behaviors, language difficulties and learning disabilities were considered the major areas of concern in FXS, while PM was reported as strongly associated to cognitive problems, social anxiety, and overthinking. Anxiety was reported as a treatment priority in both FXS and PM. CONCLUSION: FXS and PM can be associated with a range of cognitive, affective, and physical health complications. Taking a patient-first perspective may help clinicians to better characterize the cognitive-behavioral phenotype associated to these conditions, and eventually to implement tailored therapeutic approaches.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Fragile X Syndrome/genetics , Fragile X Syndrome/therapy , Humans , Fragile X Mental Retardation Protein/genetics , Female , Italy , Male , Surveys and Questionnaires , Adult , Quality of Life , Middle Aged , Ataxia/genetics , Ataxia/therapy , Young Adult , Adolescent , Tremor/genetics , Tremor/therapy , Child
7.
Elife ; 122024 Jul 29.
Article in English | MEDLINE | ID: mdl-39072369

ABSTRACT

The cerebellum contributes to a diverse array of motor conditions, including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single-neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.


Intentional movement is fundamental to achieving many goals, whether they are as complicated as driving a car or as routine as feeding ourselves with a spoon. The cerebellum is a key brain area for coordinating such movement. Damage to this region can cause various movement disorders: ataxia (uncoordinated movement); dystonia (uncontrolled muscle contractions); and tremor (involuntary and rhythmic shaking). While abnormal electrical activity in the brain associated with movement disorders has been recorded for decades, previous studies often explored one movement disorder at a time. Therefore, it remained unclear whether the underlying brain activity is similar across movement disorders. Van der Heijden and Brown et al. analyzed recordings of neuron activity in the cerebellum of mice with movement disorders to create an activity profile for each disorder. The researchers then used machine learning to generate a classifier that could separate profiles associated with manifestations of ataxia, dystonia, and tremor based on unique features of their neural activity. The ability of the model to separate the three types of movement disorders indicates that abnormal movements can be distinguished based on neural activity patterns. When additional manifestations of these abnormal movements were considered, multiple mouse models of dystonia and tremor tended to show similar profiles. Ataxia models had several different types of neural activity that were all distinct from the dystonia and tremor profiles. After identifying the activity associated with each movement disorder, Van der Heijden and Brown et al. induced the same activity in the cerebella of healthy mice, which then caused the corresponding abnormal movements. These findings lay an important groundwork for the development of treatments for neurological disorders involving ataxia, dystonia, and tremor. They identify the cerebellum, and specific patterns of activity within it, as potential therapeutic targets. While the different activity profiles of ataxia may require more consideration, the neural activity associated with dystonia and tremor appears to be generalizable across multiple manifestations, suggesting potential treatments could be broadly applicable for these disorders.


Subject(s)
Ataxia , Cerebellar Nuclei , Disease Models, Animal , Dystonia , Tremor , Animals , Tremor/physiopathology , Mice , Dystonia/physiopathology , Cerebellar Nuclei/physiopathology , Cerebellar Nuclei/physiology , Ataxia/physiopathology , Optogenetics , Action Potentials/physiology , Male , Female , Neurons/physiology
8.
J Neurogenet ; 38(2): 27-34, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38975939

ABSTRACT

Tropical ataxic neuropathy (TAN) is characterised by ataxic polyneuropathy, degeneration of the posterior columns and pyramidal tracts, optic atrophy, and sensorineural hearing loss. It has been attributed to nutritional/toxic etiologies, but evidence for the same has been equivocal. TAN shares common clinical features with inherited neuropathies and mitochondrial disorders, it may be hypothesised that genetic abnormalities may underlie the pathophysiology of TAN. This study aimed to establish evidence for mitochondrial dysfunction by adopting an integrated biochemical and multipronged genetic analysis. Patients (n = 65) with chronic progressive ataxic neuropathy with involvement of visual and/or auditory pathways underwent deep phenotyping, genetic studies including mitochondrial DNA (mtDNA) deletion analysis, mtDNA and clinical exome sequencing (CES), and respiratory chain complex (RCC) assay. The phenotypic characteristics included dysfunction of visual (n = 14), auditory (n = 12) and visual + auditory pathways (n = 29). Reduced RCC activity was present in 13 patients. Mitochondrial DNA deletions were noted in five patients. Sequencing of mtDNA (n = 45) identified a homoplasmic variant (MT-ND6) and a heteroplasmic variant (MT-COI) in one patient each. CES (n = 45) revealed 55 variants in nuclear genes that are associated with neuropathy (n = 27), deafness (n = 7), ataxia (n = 4), and mitochondrial phenotypes (n = 5) in 36 patients. This study provides preliminary evidence that TAN is associated with a spectrum of genetic abnormalities, including those associated with mitochondrial dysfunction, which is in contradistinction from the prevailing hypothesis that TAN is related to dietary toxins. Analysing the functional relevance of these genetic variants may improve the understanding of the pathogenesis of TAN.


Subject(s)
Ataxia , DNA, Mitochondrial , Humans , Male , Female , DNA, Mitochondrial/genetics , Adult , Middle Aged , Ataxia/genetics , Adolescent , Mitochondrial Diseases/genetics , Young Adult , Mitochondria/genetics , Child , Aged , Exome Sequencing , Phenotype
9.
Neurobiol Dis ; 199: 106600, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996985

ABSTRACT

Familial Dysautonomia (FD) is an autosomal recessive disorder caused by a splice site mutation in the gene ELP1, which disproportionally affects neurons. While classically characterized by deficits in sensory and autonomic neurons, neuronal defects in the central nervous system have also been described. Although ELP1 expression remains high in the normal developing and adult cerebellum, its role in cerebellar development is unknown. To explore the role of Elp1 in the cerebellum, we knocked out Elp1 in cerebellar granule cell progenitors (GCPs) and examined the outcome on animal behavior and cellular composition. We found that GCP-specific conditional knockout of Elp1 (Elp1cKO) resulted in ataxia by 8 weeks of age. Cellular characterization showed that the animals had smaller cerebella with fewer granule cells. This defect was already apparent as early as 7 days after birth, when Elp1cKO animals also had fewer mitotic GCPs and shorter Purkinje dendrites. Through molecular characterization, we found that loss of Elp1 was associated with an increase in apoptotic cell death and cell stress pathways in GCPs. Our study demonstrates the importance of ELP1 in the developing cerebellum, and suggests that loss of Elp1 in the GC lineage may also play a role in the progressive ataxia phenotypes of FD patients.


Subject(s)
Cerebellum , Dysautonomia, Familial , Mice, Knockout , Phenotype , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/pathology , Cerebellum/metabolism , Cerebellum/pathology , Mice , Disease Models, Animal , Ataxia/genetics , Ataxia/pathology , Ataxia/metabolism , Neural Stem Cells/metabolism , Apoptosis/physiology , Intracellular Signaling Peptides and Proteins
10.
Int J Mol Sci ; 25(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891946

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinal dystrophy caused by the loss of photoreceptors and retinal pigment epithelial atrophy, leading to severe visual impairment or blindness. RP can be classified as nonsyndromic or syndromic with complex clinical phenotypes. Three unrelated Polish probands affected with retinitis pigmentosa coexisting with cerebellar ataxia were recruited for this study. Clinical heterogeneity and delayed appearance of typical disease symptoms significantly prolonged the patients' diagnostic process. Therefore, many clinical and genetic tests have been performed in the past. Here, we provide detailed clinical and genetic analysis results of the patients. Whole-exome sequencing (WES) and targeted NGS analysis allow the identification of four novel and two previously reported variants in the following genes: ABHD12, FLVCR1, and PNPLA6. The use of next-generation sequencing (NGS) methods finally allowed for confirmation of the clinical diagnosis. Ultra-rare diseases such as PHARC, PCARP, and Oliver-McFarlane syndromes were diagnosed in patients, respectively. Our findings confirmed the importance of the application of next-generation sequencing methods, especially in ultra-rare genetic disorders with overlapping features.


Subject(s)
Exome Sequencing , Retinitis Pigmentosa , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/diagnosis , Male , Female , Pedigree , High-Throughput Nucleotide Sequencing , Adult , Cerebellar Ataxia/genetics , Cerebellar Ataxia/diagnosis , Membrane Transport Proteins/genetics , Monoacylglycerol Lipases/genetics , Mutation , Ataxia/genetics , Ataxia/diagnosis , Phenotype , Acyltransferases , Cataract , Phospholipases , Polyneuropathies
11.
Neurobiol Dis ; 199: 106555, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38844245

ABSTRACT

Progressive myoclonus ataxia (PMA) is a rare clinical syndrome characterized by the presence of progressive myoclonus and ataxia, and can be accompanied by mild cognitive impairment and infrequent epileptic seizures. This is the first study to describe the natural history of PMA and identify clinical, electrophysiological, and genetic features explaining the variability in disease progression. A Dutch cohort of consecutive patients meeting the criteria of the refined definition of PMA was included. The current phenotype was assessed during in-person consultation by movement disorders experts, and retrospective data was collected to describe disease presentation and progression, including brain imaging and therapy efficacy. Extensive genetic and electrophysiological tests were performed. The presence of cortical hyperexcitability was determined, by either the identification of a cortical correlate of myoclonic jerks with simultaneous electromyography-electroencephalography or a giant somatosensory evoked potential. We included 34 patients with PMA with a median disease duration of 15 years and a clear progressive course in most patients (76%). A molecular etiology was identified in 82% patients: ATM, CAMTA1, DHDDS, EBF3, GOSR2, ITPR1, KCNC3, NUS1, POLR1A, PRKCG, SEMA6B, SPTBN2, TPP1, ZMYND11, and a 12p13.32 deletion. The natural history is a rather homogenous onset of ataxia in the first two years of life followed by myoclonus in the first 5 years of life. Main accompanying neurological dysfunctions included cognitive impairment (62%), epilepsy (38%), autism spectrum disorder (27%), and behavioral problems (18%). Disease progression showed large variability ranging from an epilepsy free PMA phenotype (62%) to evolution towards a progressive myoclonus epilepsy (PME) phenotype (18%): the existence of a PMA-PME spectrum. Cortical hyperexcitability could be tested in 17 patients, and was present in 11 patients and supported cortical myoclonus. Interestingly, post-hoc analysis showed that an absence of cortical hyperexcitability, suggesting non-cortical myoclonus, was associated with the PMA-end of the spectrum with no epilepsy and milder myoclonus, independent of disease duration. An association between the underlying genetic defects and progression on the PMA-PME spectrum was observed. By describing the natural history of the largest cohort of published patients with PMA so far, we see a homogeneous onset with variable disease progression, in which phenotypic evolution to PME occurs in the minority. Genetic and electrophysiological features may be of prognostic value, especially the determination of cortical hyperexcitability. Furthermore, the identification of cortical and non-cortical myoclonus in PMA helps us gain insight in the underlying pathophysiology of myoclonus.


Subject(s)
Disease Progression , Humans , Female , Male , Adult , Middle Aged , Young Adult , Retrospective Studies , Electroencephalography/methods , Aged , Electromyography , Ataxia/genetics , Ataxia/physiopathology , Adolescent , Myoclonus/physiopathology , Myoclonus/genetics
12.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928282

ABSTRACT

Biotin (vitamin B7, or vitamin H) is a water-soluble B-vitamin that functions as a cofactor for carboxylases, i.e., enzymes involved in the cellular metabolism of fatty acids and amino acids and in gluconeogenesis; moreover, as reported, biotin may be involved in gene regulation. Biotin is not synthesized by human cells, but it is found in food and is also produced by intestinal bacteria. Biotin status/homeostasis in human individuals depends on several factors, including efficiency/deficiency of the enzymes involved in biotin recycling within the human organism (biotinidase, holocarboxylase synthetase), and/or effectiveness of intestinal uptake, which is mainly accomplished through the sodium-dependent multivitamin transporter. In the last years, administration of biotin at high/"pharmacological" doses has been proposed to treat specific defects/deficiencies and human disorders, exhibiting mainly neurological and/or dermatological symptoms and including biotinidase deficiency, holocarboxylase synthetase deficiency, and biotin-thiamine-responsive basal ganglia disease. On the other hand, according to warnings of the Food and Drug Administration, USA, high biotin levels can affect clinical biotin-(strept)avidin assays and thus lead to false results during quantification of critical biomarkers. In this review article, recent findings/advancements that may offer new insight in the abovementioned research fields concerning biotin will be presented and briefly discussed.


Subject(s)
Biotin , Biotinidase Deficiency , Biotinidase , Homeostasis , Humans , Biotin/metabolism , Biotinidase Deficiency/metabolism , Biotinidase Deficiency/diagnosis , Biotinidase Deficiency/genetics , Biotinidase Deficiency/drug therapy , Biotinidase/metabolism , Biotinidase/genetics , Holocarboxylase Synthetase Deficiency/metabolism , Carbon-Nitrogen Ligases/metabolism , Carbon-Nitrogen Ligases/genetics , Animals , Ataxia/metabolism , Ataxia/genetics , Basal Ganglia Diseases
13.
Neuropharmacology ; 257: 110035, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38876310

ABSTRACT

We previously showed that the PDE4 inhibitor apremilast reduces ethanol consumption in mice by protein kinase A (PKA) and GABAergic mechanisms. Preventing PKA phosphorylation of GABAA ß3 subunits partially blocked apremilast-mediated decreases in drinking. Here, we produced Gabrb1-S409A mice to render GABAA ß1 subunits resistant to PKA-mediated phosphorylation. Mass spectrometry confirmed the presence of the S409A mutation and lack of changes in ß1 subunit expression or phosphorylation at other residues. ß1-S409A male and female mice did not differ from wild-type C57BL/6J mice in expression of Gabrb1, Gabrb2, or Gabrb3 subunits or in behavioral characteristics. Apremilast prolonged recovery from ethanol ataxia to a greater extent in Gabrb1-S409A mice but prolonged recovery from zolpidem and propofol to a similar extent in both genotypes. Apremilast shortened recovery from diazepam ataxia in wild-type but prolonged recovery in Gabrb1-S409A mice. In wild-type mice, the PKA inhibitor H89 prevented apremilast modulation of ataxia by ethanol and diazepam, but not by zolpidem. In Gabrb1-S409A mice, inhibiting PKA or EPAC2 (exchange protein directly activated by cAMP) partially reversed apremilast potentiation of ethanol, diazepam, and zolpidem ataxia. Apremilast prevented acute tolerance to ethanol ataxia in both genotypes, but there were no genotype differences in ethanol consumption before or after apremilast. In contrast to results in Gabrb3-S408A/S409A mice, PKA phosphorylation of ß1-containing GABAA receptors is not required for apremilast's effects on acute tolerance or on ethanol consumption but is required for its ability to decrease diazepam intoxication. Besides PKA we identified EPAC2 as an additional cAMP-dependent mechanism by which apremilast regulates responses to GABAergic drugs.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Ethanol , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors , Receptors, GABA-A , Thalidomide , Animals , Thalidomide/pharmacology , Thalidomide/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Male , Female , Ethanol/pharmacology , Mice , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Gene Knock-In Techniques , Phosphorylation/drug effects , Ataxia/genetics , Alcohol Drinking/drug therapy , Alcohol Drinking/genetics , Mice, Transgenic , Diazepam/pharmacology
14.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928331

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder currently affecting the ageing population. Although the aetiology of PD has yet to be fully elucidated, environmental factors such as exposure to the naturally occurring neurotoxin rotenone has been associated with an increased risk of developing PD. Rotenone inhibits mitochondrial respiratory chain (MRC) complex I activity as well as induces dopaminergic neuronal death. The aim of the present study was to investigate the underlying mechanisms of rotenone-induced mitochondrial dysfunction and oxidative stress in an in vitro SH-SY5Y neuronal cell model of PD and to assess the ability of pre-treatment with Coenzyme Q10 (CoQ10) to ameliorate oxidative stress in this model. Spectrophotometric determination of the mitochondrial enzyme activities and fluorescence probe studies of reactive oxygen species (ROS) production was assessed. Significant inhibition of MRC complex I and II-III activities was observed, together with a significant loss of neuronal viability, CoQ10 status, and ATP synthesis. Additionally, significant increases were observed in intracellular and mitochondrial ROS production. Remarkably, CoQ10 supplementation was found to reduce ROS formation. These results have indicated mitochondrial dysfunction and increased oxidative stress in a rotenone-induced neuronal cell model of PD that was ameliorated by CoQ10 supplementation.


Subject(s)
Mitochondria , Neurons , Oxidative Stress , Rotenone , Ubiquinone , Humans , Ataxia , Cell Line, Tumor , Cell Survival/drug effects , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Diseases , Muscle Weakness/metabolism , Muscle Weakness/chemically induced , Muscle Weakness/pathology , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/etiology , Reactive Oxygen Species/metabolism , Rotenone/toxicity , Rotenone/adverse effects , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/deficiency
15.
J Am Coll Radiol ; 21(6S): S100-S125, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38823940

ABSTRACT

Diagnostic evaluation of a patient with dizziness or vertigo is complicated by a lack of standardized nomenclature, significant overlap in symptom descriptions, and the subjective nature of the patient's symptoms. Although dizziness is an imprecise term often used by patients to describe a feeling of being off-balance, in many cases dizziness can be subcategorized based on symptomatology as vertigo (false sense of motion or spinning), disequilibrium (imbalance with gait instability), presyncope (nearly fainting or blacking out), or lightheadedness (nonspecific). As such, current diagnostic paradigms focus on timing, triggers, and associated symptoms rather than subjective descriptions of dizziness type. Regardless, these factors complicate the selection of appropriate diagnostic imaging in patients presenting with dizziness or vertigo. This document serves to aid providers in this selection by using a framework of definable clinical variants. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision process support the systematic analysis of the medical literature from peer reviewed journals. Established methodology principles such as Grading of Recommendations Assessment, Development, and Evaluation or GRADE are adapted to evaluate the evidence. The RAND/UCLA Appropriateness Method User Manual provides the methodology to determine the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where peer reviewed literature is lacking or equivocal, experts may be the primary evidentiary source available to formulate a recommendation.


Subject(s)
Dizziness , Societies, Medical , Dizziness/diagnostic imaging , Humans , United States , Ataxia/diagnostic imaging , Evidence-Based Medicine , Diagnosis, Differential
16.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928470

ABSTRACT

Coenzyme Q10 (CoQ10) plays a key role in many aspects of cellular metabolism. For CoQ10 to function normally, continual interconversion between its oxidised (ubiquinone) and reduced (ubiquinol) forms is required. Given the central importance of this ubiquinone-ubiquinol redox cycle, this article reviews what is currently known about this process and the implications for clinical practice. In mitochondria, ubiquinone is reduced to ubiquinol by Complex I or II, Complex III (the Q cycle) re-oxidises ubiquinol to ubiquinone, and extra-mitochondrial oxidoreductase enzymes participate in the ubiquinone-ubiquinol redox cycle. In clinical terms, the outcome of deficiencies in various components associated with the ubiquinone-ubiquinol redox cycle is reviewed, with a particular focus on the potential clinical benefits of CoQ10 and selenium co-supplementation.


Subject(s)
Oxidation-Reduction , Ubiquinone , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Ubiquinone/deficiency , Humans , Mitochondria/metabolism , Animals , Selenium/metabolism , Ataxia , Muscle Weakness , Mitochondrial Diseases
18.
Orphanet J Rare Dis ; 19(1): 200, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755691

ABSTRACT

BACKGROUND: MT-ATP6 is a mitochondrial gene which encodes for the intramembrane subunit 6 (or A) of the mitochondrial ATP synthase, also known asl complex V, which is involved in the last step of oxidative phosphorylation to produce cellular ATP through aerobic metabolism. Although classically associated with the NARP syndrome, recent evidence highlights an important role of MT-ATP6 pathogenic variants in complicated adult-onset ataxias. METHODS: We describe two unrelated patients with adult-onset cerebellar ataxia associated with severe optic atrophy and mild cognitive impairment. Whole mitochondrial DNA sequencing was performed in both patients. We employed patients' primary fibroblasts and cytoplasmic hybrids (cybrids), generated from patients-derived cells, to assess the activity of respiratory chain complexes, oxygen consumption rate (OCR), ATP production and mitochondrial membrane potential. RESULTS: In both patients, we identified the same novel m.8777 T > C variant in MT-ATP6 with variable heteroplasmy level in different tissues. We identifed an additional heteroplasmic novel variant in MT-ATP6, m.8879G > T, in the patients with the most severe phenotype. A significant reduction in complex V activity, OCR and ATP production was observed in cybrid clones homoplasmic for the m.8777 T > C variant, while no functional defect was detected in m.8879G > T homoplasmic clones. In addition, fibroblasts with high heteroplasmic levelsof m.8777 T > C variant showed hyperpolarization of mitochondrial membranes. CONCLUSIONS: We describe a novel pathogenic mtDNA variant in MT-ATP6 associated with adult-onset ataxia, reinforcing the value of mtDNA screening within the diagnostic workflow of selected patients with late onset ataxias.


Subject(s)
Mitochondrial Proton-Translocating ATPases , Humans , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Male , Female , Middle Aged , Ataxia/genetics , Ataxia/pathology , Italy , DNA, Mitochondrial/genetics , Adult , Fibroblasts/metabolism , Fibroblasts/pathology
19.
Free Radic Biol Med ; 221: 257-260, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38754742

ABSTRACT

It has generally been accepted that vitamin E refers to a group of tocochromanols, α-, ß-, γ-, and δ-tocopherols and the corresponding four tocotrienols. Recently, Azzi and colleagues proposed to restrict the term vitamin E only to RRR-α-tocopherol, not to other tocopherols and tocotrienols (Azzi A et al. Free Radic Biol Med. 2023; 207:178-180. doi: 10.1016/j.freeradbiomed.2023.06.029). The aim of this paper is to express our opinion on the nomenclature of vitamin E based on available scientific data. In our opinion, it would be inappropriate to exclude all the tocochromanols other than RRR-α-tocopherol from the vitamin E group at this stage when the molecular mechanisms showing how vitamin E deficiency causes diseases such as ataxia and how vitamin E prevents/reverses such diseases are not elucidated. Understanding of whole functions of tocochromanols including underlying mechanisms and dynamics is essential before revision of currently accepted definition of vitamin E. The potential roles of γ-tocopherol and tocotrienols are discussed despite whether they are vitamin function should be clarified in the future studies.


Subject(s)
Terminology as Topic , Vitamin E Deficiency , Vitamin E , alpha-Tocopherol , Vitamin E/chemistry , Vitamin E/classification , Humans , alpha-Tocopherol/chemistry , Ataxia/classification , Tocotrienols/classification , Tocotrienols/chemistry , Antioxidants/chemistry , Animals
20.
Neurosci Biobehav Rev ; 162: 105731, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763180

ABSTRACT

Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Neuroglia , Humans , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Neuroglia/metabolism , Animals , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Fragile X Syndrome/pathology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Brain Diseases/genetics , Ataxia/metabolism , Ataxia/physiopathology , Ataxia/genetics , Tremor/metabolism , Tremor/physiopathology , Tremor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL