Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Neurol Sci ; 45(4): 1749-1753, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38252374

ABSTRACT

BACKGROUND: ATP13A2 is a monogenic causative gene of Parkinson's disease, whose biallelic mutations can result in Kufor-Rakeb syndrome. Biallelic mutations in ATP13A2 have also been reported in pure or complicated hereditary spastic paraplegia (HSP). Here, we report clinical, neuroimaging, and genetic findings from a patient with a novel homozygous mutation in ATP13A2 presenting with HSP plus parkinsonism. METHODS: Whole genome sequencing was performed on the patient, a 46-year-old Chinese woman from a consanguineous family, to identify the genetic cause. Furthermore, functional studies of the identified ATP13A2 mutation were conducted. RESULTS: The patient initially presented with abnormal gait because of lower-limb spasticity and recurrent seizures. Parkinsonism (presenting as bradykinesia and rigidity) and peripheral neuropathy in lower limbs further evolved and resulted in her eventual use of a wheelchair. Symmetrically decreased dopamine transporter density was detected within the bilateral putamen and caudate nucleus in dopamine transporter-positron emission tomography. Genetic analysis revealed a novel homozygous missense mutation in ATP13A2 (c.2780 T > C, p.Leu927Pro), which was heterozygous in the patient's parents and son. Functional studies suggested that this mutation results in the reduced expression and altered subcellular localization of ATP13A2. CONCLUSIONS: Our report broadens the genetic and phenotypic spectrum of ATP13A2-related HSP. Further research is needed to fully elucidate the mechanism linking ATP13A2 variants to HSP.


Subject(s)
Parkinson Disease , Parkinsonian Disorders , Spastic Paraplegia, Hereditary , Humans , Female , Middle Aged , Dopamine Plasma Membrane Transport Proteins , Spastic Paraplegia, Hereditary/diagnostic imaging , Spastic Paraplegia, Hereditary/genetics , Mutation/genetics , Parkinsonian Disorders/genetics , Phenotype , Pedigree , Proton-Translocating ATPases/genetics
2.
Epilepsia ; 63(5): 1093-1103, 2022 05.
Article in English | MEDLINE | ID: mdl-35247271

ABSTRACT

OBJECTIVE: Although previous imaging studies have reported cerebellar gray matter loss in patients with familial cortical myoclonic tremor with epilepsy (FCMTE), the corresponding white matter alterations remain unknown. We investigated white matter structural changes in FCMTE1 and compared them with clinical and electrophysiological features. METHODS: We enrolled 36 patients carrying heterozygous pathogenic intronic pentanucleotide insertions in the SAMD12 gene and 52 age- and sex-matched healthy controls. Diffusion tensor imaging-derived metrics, including fractional anisotropy, mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD), were calculated along with white matter voxel-based morphometry (VBM) analysis. We also examined correlations between magnetic resonance metrics and clinical and electrophysiological features. RESULTS: We detected widespread white matter reductions in MD, RD, and AD values in FCMTE1 patients, including in the commissural, projection, and association fibers. VBM analysis revealed that increases in white matter volume predominantly occurred in the right cerebellum and sagittal stratum. MD, RD, AD, and VBM analysis clearly indicated changes in the sagittal stratum. We found a positive correlation between VBM values in the right cerebellum and somatosensory-evoked potential P25-N33 amplitude. Decreased MD and AD values in the right sagittal stratum were detected in patients with versus without photophobia. SIGNIFICANCE: FCMTE is a network disorder involving a wide range of cortical and subcortical structures, including the cerebellum, thalamus, thalamocortical connections, and corticocortical connections. The right sagittal stratum is closely related with visual symptoms, especially photophobia. Our findings indicate that cerebellum and cortical hyperexcitability are closely linked, and emphasize the important role of the cerebellum in the pathophysiological mechanisms of cortical tremor.


Subject(s)
Epilepsy , White Matter , Brain/pathology , Diffusion Tensor Imaging/methods , Epilepsies, Myoclonic , Epilepsy/pathology , Gray Matter/pathology , Humans , Photophobia , Tremor/diagnostic imaging , Tremor/genetics , Tremor/pathology , White Matter/diagnostic imaging , White Matter/pathology
3.
Eur J Neurol ; 29(6): 1610-1618, 2022 06.
Article in English | MEDLINE | ID: mdl-35147270

ABSTRACT

BACKGROUND AND PURPOSE: Recently, the pathogenic and intermediate GGC repeat expansion in NOTCH2NLC was detected in Parkinson's disease (PD). However, detailed clinical, neuroimaging, and pathological information of clinically diagnosed PD patients with pathogenic GGC repeat expansion in NOTCH2NLC remains scarce. Thus, we aimed to elucidate the clinical, neuroimaging, and pathological characteristics of PD patients carrying the pathogenic GGC repeat expansion in NOTCH2NLC. METHODS: The NOTCH2NLC GGC repeat expansion was screened in 941 sporadic PD patients and 244 unrelated probands. Comprehensive assessments were performed in three PD patients with pathogenic GGC repeat expansion in NOTCH2NLC. The repeat expansion length was estimated using CRISPR/Cas9-based targeted long-read sequencing. RESULTS: The three patients (two PD patients from Family 1 and one sporadic PD) carrying the pathogenic NOTCH2NLC expansion were reconfirmed with a diagnosis of clinically established PD. Although they lacked the typical neuronal intranuclear inclusion disease (NIID) magnetic resonance imaging (MRI) feature, the typical PD pattern of striatal dopamine transporter loss was detected. Notably, all three patients presented with systemic areflexia, and other secondary causes of polyneuropathy were excluded. Skin biopsy showed intranuclear inclusions and an absence of phosphorylated alpha-synuclein deposition in the skin nerve fibers of all three patients. CONCLUSIONS: Although these clinically diagnosed PD patients with pathogenic GGC repeat expansion in NOTCH2NLC were hardly distinguishable from idiopathic PD based on clinical course and neuroimaging features, the pathological findings indicated that their phenotype was a PD phenocopy of NIID. Systemic areflexia may be an important and unique clinical clue suggesting further genetic testing and skin biopsy examination to confirm the diagnosis of NIID in patients presenting with a PD phenocopy.


Subject(s)
Parkinson Disease , Humans , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/pathology , Neurodegenerative Diseases , Neuroimaging , Parkinson Disease/diagnostic imaging , Parkinson Disease/genetics , Parkinson Disease/pathology , Trinucleotide Repeat Expansion
4.
Eur J Neurol ; 29(2): 450-458, 2022 02.
Article in English | MEDLINE | ID: mdl-34750918

ABSTRACT

BACKGROUND AND PURPOSE: The GGC repeat expansion in the NOTCH2NLC gene has been identified as the genetic cause of neuronal intranuclear inclusion disease (NIID). Recently, this repeat expansion was also reported to be associated with essential tremor (ET). However, some patients with this repeat expansion, initially diagnosed with ET, were eventually diagnosed with NIID. Therefore, controversy remains regarding the clinical diagnosis of these expansion-positive patients presenting with tremor-dominant symptoms. This study aimed to clarify the clinical phenotype in tremor-dominant patients who have the GGC repeat expansion in the NOTCH2NLC gene. METHODS: We screened for pathogenic GGC repeat expansions in 602 patients initially diagnosed with ET and systematically re-evaluated the clinical features of the expansion-positive probands and their family members. RESULTS: Pathogenic GGC repeat expansion in the NOTCH2NLC gene was detected in 10 probands (1.66%). Seven of these probands were re-evaluated and found to have systemic areflexia, cognitive impairment, and abnormal nerve conduction, which prompted a change of diagnosis from ET to NIID. Three of the probands had typical hyperintensity in the corticomedullary junction on diffusion-weighted imaging. Intranuclear inclusions were detected in all four probands who underwent skin biopsy. CONCLUSIONS: The NIID tremor-dominant subtype can be easily misdiagnosed as ET. We should take NIID into account for differential diagnosis of ET. Systemic areflexia could be an important clinical clue suggesting that cranial magnetic resonance imaging examination, or even further genetic testing and skin biopsy examination, should be used to confirm the diagnosis of NIID.


Subject(s)
Essential Tremor , Intranuclear Inclusion Bodies , Essential Tremor/diagnosis , Essential Tremor/genetics , Humans , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/pathology , Neurodegenerative Diseases , Tremor/diagnosis , Tremor/genetics , Trinucleotide Repeat Expansion/genetics
5.
Brain ; 143(2): 491-502, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31851307

ABSTRACT

Primary familial brain calcification is a monogenic disease characterized by bilateral calcifications in the basal ganglia and other brain regions, and commonly presents motor, psychiatric, and cognitive symptoms. Currently, four autosomal dominant (SLC20A2, PDGFRB, PDGFB, XPR1) and one autosomal recessive (MYORG) causative genes have been identified. Compared with patients with autosomal dominant primary familial brain calcification, patients with the recessive form of the disease present with more severe clinical and imaging phenotypes, and deserve more clinical and research attention. Biallelic mutations in MYORG cannot explain all autosomal recessive primary familial brain calcification cases, indicating the existence of novel autosomal recessive genes. Using homozygosity mapping and whole genome sequencing, we detected a homozygous frameshift mutation (c.140delT, p.L48*) in the JAM2 gene in a consanguineous family with two affected siblings diagnosed with primary familial brain calcification. Further genetic screening in a cohort of 398 probands detected a homozygous start codon mutation (c.1A>G, p.M1?) and compound heterozygous mutations [c.504G>C, p.W168C and c.(67+1_68-1)_(394+1_395-1), p.Y23_V131delinsL], respectively, in two unrelated families. The clinical phenotypes of the four patients included parkinsonism (3/4), dysarthria (3/4), seizures (1/4), and probable asymptomatic (1/4), with diverse onset ages. All patients presented with severe calcifications in the cortex in addition to extensive calcifications in multiple brain areas (lenticular nuclei, caudate nuclei, thalamus, cerebellar hemispheres, ± brainstem; total calcification scores: 43-77). JAM2 encodes junctional adhesion molecule 2, which is highly expressed in neurovascular unit-related cell types (endothelial cells and astrocytes) and is predominantly localized on the plasma membrane. It may be important in cell-cell adhesion and maintaining homeostasis in the CNS. In Chinese hamster ovary cells, truncated His-tagged JAM2 proteins were detected by western blot following transfection of p.Y23_V131delinsL mutant plasmid, while no protein was detected following transfection of p.L48* or p.1M? mutant plasmids. In immunofluorescence experiments, the p.W168C mutant JAM2 protein failed to translocate to the plasma membrane. We speculated that mutant JAM2 protein resulted in impaired cell-cell adhesion functions and reduced integrity of the neurovascular unit. This is similar to the mechanisms of other causative genes for primary familial brain calcification or brain calcification syndromes (e.g. PDGFRB, PDGFB, MYORG, JAM3, and OCLN), all of which are highly expressed and functionally important in the neurovascular unit. Our study identifies a novel causative gene for primary familial brain calcification, whose vital function and high expression in the neurovascular unit further supports impairment of the neurovascular unit as the root of primary familial brain calcification pathogenesis.


Subject(s)
Brain Diseases/genetics , Brain/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Adult , Brain/pathology , Brain Diseases/metabolism , Calcinosis/genetics , Female , Genes, Recessive/genetics , Humans , Male , Middle Aged , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Pedigree , Phenotype , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Xenotropic and Polytropic Retrovirus Receptor
6.
Int J Neurosci ; 131(6): 521-526, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32942935

ABSTRACT

INTRODUCTION: Lateral trunk flexion (LTF) is a common postural deformity in Parkinson's disease (PD). Postural control is known to depend on visual, vestibular, and somatosensory information. OBJECTIVES: This study aimed to investigate the relationship between vestibular dysfunction and postural abnormalities in PD patients with LTF. METHODS: We enrolled a total of 19 PD patients with LTF (PD-LTF+) and 19 age- and sex-matched PD patients without LTF (PD-LTF-). All patients underwent vestibular tests, including spontaneous nystagmus, gaze-evoked nystagmus, ocular movements, optokinetic eye test, fast positioning maneuvers, and the bithermal caloric test. RESULTS: Most of the PD-LTF + patients had abnormal vestibular function (11/19), while there were fewer vestibular function injuries in the control group (3/19). In PD-LTF + group, there were 5 patients (5/11, 45.5%) of peripheral vestibular dysfunction, 2 patients (2/11, 18.2%) of central vestibular damage, and 4 patients (4/11, 36.4%) of mixed injuries. The peripheral vestibular deficiencies could be either bilateral (4/9, 44.4%) or unilateral (5/9, 55.6%). The unilateral vestibular dysfunction was ipsilateral to the leaning side in 2 patients and contralateral to the leaning side in the other 3 patients. CONCLUSION: Vestibular dysfunction may be an independent risk factor for LTF in PD patients.


Subject(s)
Parkinson Disease/physiopathology , Postural Balance/physiology , Torso/physiopathology , Vestibular Diseases/physiopathology , Aged , Female , Humans , Male , Middle Aged , Risk Factors , Vestibular Diseases/diagnosis , Vestibular Function Tests
7.
Mov Disord ; 35(6): 1012-1020, 2020 06.
Article in English | MEDLINE | ID: mdl-32129927

ABSTRACT

BACKGROUND: Intronic pentanucleotide insertion in the sterile alpha motif domain-containing 12 gene was recently identified as the genetic cause of familial cortical myoclonic tremor with epilepsy type 1. OBJECTIVES: We thereafter conducted a multimodal MRI research to further understand familial cortical myoclonic tremor with epilepsy type 1. METHODS: We enrolled 31 patients carrying heterozygous pathogenic intronic pentanucleotide insertion in the sterile alpha motif domain-containing 12 gene and 31 age- and sex-matched healthy controls. We compared multimodal MRI metrics, including voxel-based morphometry, fractional anisotropy of diffuse tensor imaging, frequency-dependent percent amplitude fluctuation, and seed-based functional connectivity of resting-state functional MRI. RESULTS: Significant decreased gray matter volume was found in the cerebellum. Percent amplitude fluctuation analysis showed significant interaction effect of "Frequency by Group" in three regions, including the vermis VIII, left cerebellar lobule VIII, and left precentral gyrus. Specifically, the lowest-frequency band exhibited significant increased percent amplitude fluctuation in patients in the two cerebellar subregions, whereas the highest-frequency band exhibited decreased percent amplitude fluctuation in the precentral gyrus in patients. Discriminative analysis by support vector machine showed a mean accuracy of 82% (P = 1.0-5 ). An increased functional connectivity between vermis VIII and the left precentral gyrus was found in patients with familial cortical myoclonic tremor with epilepsy type 1. A positive correlation between the percent amplitude fluctuation in the left cerebellar lobule VIII and duration of cortical tremor was also found. CONCLUSION: The cerebellum showed both structural and functional damages. The distinct change of spontaneous brain activity, that is, increased ultra-low-frequency amplitude in the cerebellum and the decreased higher-frequency amplitude in the motor cortex, might be a pathophysiological feature of familial cortical myoclonic tremor with epilepsy type 1. © 2020 International Parkinson and Movement Disorder Society.


Subject(s)
Epilepsies, Myoclonic , Epilepsy , Cerebellum , Epilepsies, Myoclonic/diagnostic imaging , Epilepsies, Myoclonic/genetics , Humans , Magnetic Resonance Imaging , Tremor/diagnostic imaging , Tremor/genetics
8.
Mov Disord ; 35(4): 679-686, 2020 04.
Article in English | MEDLINE | ID: mdl-31951047

ABSTRACT

BACKGROUND: Biallelic mutations in the MYORG gene were first identified as the cause of recessively inherited primary familial brain calcification. Interestingly, some heterozygous carriers also exhibited brain calcifications. OBJECTIVES: To further investigate the role of single heterozygous MYORG mutations in the development of brain calcifications. METHODS: A nation-wide cohort of Chinese primary familial brain calcification probands was enrolled from March 2016 through September 2019. Mutational analysis of MYORG was performed in 435 primary familial brain calcification probands who were negative for mutations in the other four known primary familial brain calcification-causative genes (SLC20A2, PDGFRB, PDGFB, and XPR1). RESULTS: Biallelic MYORG mutations were identified in 14 primary familial brain calcification patients from 10 unrelated families. Interestingly, 12 heterozygous carriers from seven of these families also exhibited mild-to-moderate brain calcifications. Moreover, single heterozygous mutations were detected in an additional 9 probands and in 7 of their family members affected with brain calcifications. In our cohort, clinical and imaging penetrance of individuals with biallelic mutations were 100%, whereas among individuals with heterozygous mutations, penetrance of imaging phenotype was reduced to 73.7% (28 of 38) and clinical penetrance was much lower. Most (34 of 38) remained asymptomatic whereas 4 carriers had symptoms of uncertain clinical significance (nonspecific depression, epilepsy and late-onset parkinsonism). Compared with individuals with biallelic MYORG mutations, individuals with heterozygous mutations had brain calcifications with much lower calcification scores (P < 2e-16). CONCLUSIONS: Presence of brain calcifications in individuals with heterozygous MYORG mutations suggested a semidominant inheritance pattern with incomplete penetrance. This finding further expanded the genotype-phenotype correlations of MYORG-related primary familial brain calcification. © 2020 International Parkinson and Movement Disorder Society.


Subject(s)
Brain Diseases , Glycoside Hydrolases/genetics , Brain/diagnostic imaging , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Heterozygote , Humans , Mutation/genetics , Pedigree , Xenotropic and Polytropic Retrovirus Receptor
9.
Mov Disord ; 34(6): 912-916, 2019 06.
Article in English | MEDLINE | ID: mdl-30964957

ABSTRACT

BACKGROUND: Recently, the LRP10 gene has been identified as a novel genetic cause in individuals affected by Parkinson's disease (PD), Parkinson's disease dementia, or dementia with Lewy bodies. OBJECTIVE: We investigated the involvement of LRP10 mutations in Chinese patients with familial PD and reviewed previous studies of LRP10 mutations in patients with PD. METHODS: A mutation analysis of the LRP10 gene was performed in a cohort of 205 unrelated Chinese patients with familial PD. Burden analysis was conducted using data from the Genome Aggregation Database and 5 genetic studies of LRP10 in patients with PD (including our cohort). RESULTS: A total of 3 novel potentially pathogenic variants, c.32T>A (p.L11H), c.1184G>A (p.R395H), and c.1333G>A (p.A445T), were detected in 3 probands of our cohort. However, burden analysis argued against an overrepresentation of variant alleles in patients with PD. CONCLUSIONS: Genetic screening of the LRP10 gene in our cohort may provide independent, albeit limited, evidence for the pathogenicity of LRP10 in familial PD. Burden analysis using data from current studies failed to support the association between LRP10 and PD in general. Thus, more robust replication studies are warranted to determine the involvement of LRP10 in the pathogenesis of PD. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
LDL-Receptor Related Proteins/genetics , Mutation , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Aged , China , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Male , Middle Aged , Pedigree
10.
Mov Disord ; 34(2): 291-297, 2019 02.
Article in English | MEDLINE | ID: mdl-30589467

ABSTRACT

BACKGROUND: Very recently, the MYORG gene was identified as a novel causative gene for autosomal-recessive primary familial brain calcification. OBJECTIVE: To investigate the clinical, genetic, and neuroradiological characteristics of primary familial brain calcification patients with biallelic MYORG mutations in China. METHODS: We collected clinical and neuroradiological data of 169 Chinese patients with primary familial brain calcification, including 151 sporadic patients and 18 patients from 13 families compatible with an autosomal-recessive mode of inheritance. Mutational analysis of MYORG was performed in the cohort. RESULTS: We identified four, including three novel, MYORG mutations segregating in four families with 5 patients: one nonsense mutation (c.1431C>A, p.Y477*), one missense mutation (c.687G>T, p.W229C), and two nonframeshift indels (c.348_349insCTGGCCTTCCGC, p.116_117insLAFR; c. 428_442delTGCACTTCTTCATCC, p.143_147delLHFFI). The 12-base-pair insertion, c.348_349insCTGGCCTTCCGC, was found in either homozygous or heterozygous state in 2 probands of our cohort and another Chinese primary familial brain calcification patient previously reported on in the literature. Haplotype analysis of our patients harboring the insertion indicated a founder effect in the ethnic Han Chinese population. To date, biallelic MYORG mutations have been reported in 17 patients (including our cohort). Most patients were symptomatic (13 of 17; 76.5%), and the most recurrent symptoms were movement disorders (10 of 17; 58.8%), cognitive decline (7 of 17; 41.2%), and cerebellar symptoms (6 of 17; 35.3%). All patients had calcifications on comprehensive cranial CT, most frequently located in the basal ganglia (17 of 17; 100%), cerebellum (17 of 17; 100%), subcortical white matter (14 of 17; 82.4%), and thalamus (13 of 17; 76.5%). CONCLUSIONS: We confirmed MYORG as a novel causative gene for primary familial brain calcification and further expanded the mutational and phenotypic spectrum of MYORG-related primary familial brain calcification. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Brain Diseases/genetics , Calcinosis/genetics , Glycoside Hydrolases/genetics , Mutation/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Adult , Basal Ganglia/pathology , China , Cohort Studies , DNA Mutational Analysis/methods , Female , Heterozygote , Humans , Male , Neurodegenerative Diseases/genetics , Pedigree
11.
Mov Disord ; 34(10): 1571-1576, 2019 10.
Article in English | MEDLINE | ID: mdl-31483537

ABSTRACT

BACKGROUND: Intronic (TTTCA)n insertions in the SAMD12, TNRC6A, and RAPGEF2 genes have been identified as causes of familial cortical myoclonic tremor with epilepsy. OBJECTIVE: To identify the cause of familial cortical myoclonic tremor with epilepsy pedigrees without (TTTCA)n insertions in SAMD12, TNRC6A, and RAPGEF2. METHODS: Repeat-primed polymerase chain reaction, long-range polymerase chain reaction, and Sanger sequencing were performed to identify the existence of a novel (TTTGA)n insertion. Targeted long-read sequencing was performed to confirm the accurate structure of the (TTTGA)n insertion. RESULTS: We identified a novel expanded intronic (TTTGA)n insertion at the same site as the previously reported (TTTCA)n insertion in SAMD12. This insertion cosegregated with familial cortical myoclonic tremor with epilepsy in 1 Chinese pedigree with no (TTTCA)n insertion. In the targeted long-read sequencing of 2 patients and 1 asymptomatic carrier in this pedigree, with 1 previously reported (TTTCA)n -insertion-carrying patient as a positive control, a respective total of 302, 159, 207, and 50 on-target subreads (predicated accuracy: ≥90%) spanning the target repeat expansion region were generated. These sequencing data revealed the accurate repeat expansion structures as (TTTTA)114-123 (TTTGA)108-116 in the pedigree and (TTTTA)38 (TTTCA)479 in (TTTCA)n -insertion-carrying patient. CONCLUSION: The targeted long-read sequencing helped us to elucidate the accurate structures of the (TTTGA)n and (TTTCA)n insertions. Our finding offers a novel possible cause for familial cortical myoclonic tremor with epilepsy and might shed light on the identification of genetic causes of this disease in pedigrees with no detected (TTTCA)n insertion in the reported causative genes. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Epilepsies, Myoclonic/genetics , Nerve Tissue Proteins/genetics , Tremor/genetics , Adult , Asian People , Epilepsies, Myoclonic/complications , Humans , Introns/physiology , Male , Pedigree , Tremor/complications
12.
Brain ; 141(8): 2280-2288, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29939203

ABSTRACT

Familial cortical myoclonic tremor with epilepsy is an autosomal dominant neurodegenerative disease, characterized by cortical tremor and epileptic seizures. Although four subtypes (types 1-4) mapped on different chromosomes (8q24, 2p11.1-q12.2, 5p15.31-p15.1 and 3q26.32-3q28) have been reported, the causative gene has not yet been identified. Here, we report the genetic study in a cohort of 20 Chinese pedigrees with familial cortical myoclonic tremor with epilepsy. Linkage and haplotype analysis in 11 pedigrees revealed maximum two-point logarithm of the odds (LOD) scores from 1.64 to 3.77 (LOD scores in five pedigrees were >3.0) in chromosomal region 8q24 and narrowed the candidate region to an interval of 4.9 Mb. Using whole-genome sequencing, long-range polymerase chain reaction and repeat-primed polymerase chain reaction, we identified an intronic pentanucleotide (TTTCA)n insertion in the SAMD12 gene as the cause, which co-segregated with the disease among the 11 pedigrees mapped on 8q24 and additional seven unmapped pedigrees. Only two pedigrees did not contain the (TTTCA)n insertion. Repeat-primed polymerase chain reaction revealed that the sizes of (TTTCA)n insertion in all affected members were larger than 105 repeats. The same pentanucleotide insertion (ATTTCATTTC)58 has been reported to form RNA foci resulting in neurotoxicity in spinocerebellar ataxia type 37, which suggests the similar pathogenic process in familial cortical myoclonic tremor with epilepsy type 1.


Subject(s)
Epilepsies, Myoclonic/genetics , Microsatellite Repeats/genetics , Nerve Tissue Proteins/genetics , Adult , Aged , Asian People , China , Chromosome Mapping , Epilepsies, Myoclonic/physiopathology , Epilepsy/genetics , Ethnicity/genetics , Female , Genetic Linkage , Haplotypes , Humans , Introns/genetics , Male , Middle Aged , Mutagenesis, Insertional/genetics , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/genetics , Pedigree , Tremor/genetics
13.
Dysphagia ; 34(4): 600-607, 2019 08.
Article in English | MEDLINE | ID: mdl-31037327

ABSTRACT

Dysphagia is a common non-primary symptom of patients with Parkinson's disease. The aim of this study is to investigate the underlying alterations of brain functional connectivity in Parkinson's disease patients with dysphagia by resting-state functional magnetic resonance imaging. We recruited 13 Parkinson's disease patients with dysphagia and ten patients without dysphagia, diagnosed by videofluoroscopic study of swallowing. Another 13 age and sex-matched healthy subjects were recruited. Eigenvector centrality mapping was computed to identify functional connectivity alterations among these groups. Parkinson's disease patients with dysphagia had significantly increased functional connectivity in the cerebellum, left premotor cortex, the supplementary motor area, the primary motor cortex, right temporal pole of superior temporal gyrus, inferior frontal gyrus, anterior cingulate cortex and insula, compared with patients without dysphagia. This study suggests that functional connectivity changes in swallowing-related cortexes might contribute to the occurrence of dysphagia in Parkinson's disease patients.


Subject(s)
Brain/physiopathology , Deglutition Disorders/etiology , Nerve Net/physiopathology , Parkinson Disease/complications , Parkinson Disease/etiology , Brain/diagnostic imaging , Case-Control Studies , Deglutition Disorders/diagnostic imaging , Deglutition Disorders/physiopathology , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Net/diagnostic imaging , Parkinson Disease/diagnostic imaging , Parkinson Disease/physiopathology
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(11): 1104-1106, 2019 Nov 10.
Article in Zh | MEDLINE | ID: mdl-31703136

ABSTRACT

OBJECTIVE: To explore the genetic basis for a pedigree affected with X-linked adrenoleukodystrophy presenting as spastic paraplegia of the lower limbs. METHODS: Genomic DNA was extracted from peripheral blood samples of the patient and his mother. Potential variant was detected with a panel for genes associated with spastic paraplegia. Candidate variant was verified by PCR and Sanger sequencing. RESULTS: Both the proband and his mother presented with walking difficulty. A previously known variant, c.623T to A (p.V208E), was identified in the ABCD1 gene mapped on chromosome X in both. CONCLUSION: X-link adrenoleukodystrophy should be taken into account as a possible diagnosis for this pedigree.


Subject(s)
ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Adrenoleukodystrophy/genetics , Female , Genetic Testing , Humans , Male , Pedigree , Spastic Paraplegia, Hereditary
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(10): 957-960, 2019 Oct 10.
Article in Zh | MEDLINE | ID: mdl-31598935

ABSTRACT

OBJECTIVE: To explore the genetic basis of a patient with early-onset Parkinson disease from a consanguineous family. METHODS: Homozygosity mapping and Sanger sequencing of cDNA were used to identify the causative mutation. RESULTS: A homozygous missense variation (c.56C>G, p.Thr19Arg) in the PARK7 gene was identified in the patient. In silico analysis suggested the c.56C>G variation to be pathogenic. CONCLUSION: Homozygous c.56C>G variation of the PARK7 gene was the disease-causing variation in this family.


Subject(s)
Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Consanguinity , Homozygote , Humans , Mutation, Missense , Pedigree
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(6): 815-818, 2018 Dec 10.
Article in Zh | MEDLINE | ID: mdl-30512153

ABSTRACT

OBJECTIVE: To explore the genetic basis for a Chinese pedigree where three siblings were affected with Parkinson's disease. METHODS: Multiple ligation-dependent probe amplification (MLPA) and next-generation sequencing (NGS) were employed to detect the causative mutation. Sanger sequencing of cDNA was also used for verify the effect of mutation on the transcription of RNA. RESULTS: Heterozygous deletion of exon 3 of the PARK2 gene was detected by MLPA, while a heterozygous splice site variant c.619-3G>C was detected by NGS. Both mutations were shown to result in aberrant transcripts of the PARK2 gene (loss of exons 3 and 6, respectively) by Sanger sequencing of cDNA. Both mutations have co-segregated with the disease in the pedigree. CONCLUSION: Compound heterozygous mutations of the PARK2 gene probably underlie the disease in this pedigree. Identification of the splice site variant c.619-3G>C has expanded the mutation spectrum of the PARK2 gene.


Subject(s)
Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , Asian People , China , DNA Mutational Analysis , Exons , Heterozygote , Humans , Mutation , Pedigree
17.
J Magn Reson Imaging ; 46(3): 697-703, 2017 09.
Article in English | MEDLINE | ID: mdl-27880010

ABSTRACT

PURPOSE: To investigate the differences in spontaneous brain activity between Parkinson's disease (PD) patients with rapid eye movement sleep behavior disorder (RBD), PD patients without RBD, and normal controls, which may shed new light on the neural mechanism of RBD. MATERIALS AND METHODS: Eighteen PD patients with RBD, 16 patients without RBD, and 19 age- and gender-matched normal controls underwent clinical assessment and functional magnetic resonance imaging (fMRI) with a 3.0T scanner. Resting-state fMRI scans were collected using an echo planar imaging sequence. Amplitude of low-frequency fluctuations (ALFF) were calculated to measure spontaneous brain activity in each subject. RESULTS: Compared with PD patients without RBD, patients with RBD exhibited significantly decreased ALFF values (P < 0.001, cluster level) in primary motor cortex extending to premotor cortex. Compared with normal controls, PD patients exhibited decreased ALFF values (P < 0.001, cluster level) in caudate and putamen (P < 0.001, cluster level), and increased ALFF values (P = 0.03, cluster level) in prefrontal cortex. CONCLUSION: The altered spontaneous brain activity in motor cortex may contribute to the pathogenesis of RBD in PD patients, which further supports the idea that the pathophysiology of RBD involves not only midbrain dysfunction but also cerebral cortex abnormalities. Our findings provide additional insight into the neural mechanism of RBD and may drive future research to develop better treatment. LEVEL OF EVIDENCE: 3 Technical Efficacy: Stage 3 J. MAGN. RESON. IMAGING 2017;46:697-703.


Subject(s)
Brain/physiopathology , Magnetic Resonance Imaging/methods , Parkinson Disease/physiopathology , REM Sleep Behavior Disorder/physiopathology , Brain/diagnostic imaging , Female , Humans , Male , Middle Aged , Parkinson Disease/complications , REM Sleep Behavior Disorder/complications , Severity of Illness Index
18.
Eur Spine J ; 26(7): 1862-1870, 2017 07.
Article in English | MEDLINE | ID: mdl-28281005

ABSTRACT

BACKGROUND: Postural deformities in the coronal plane were frequent and disabling complications of PD, which reduces the quality of life of patients. This study aimed to garner greater attention to the Parkinson disease (PD)-related postural trunk deviations in the coronal plane by exploring a method for diagnosis because of the lack of any uniform diagnostic criteria and epidemiological studies. It also aimed to provide correlation data in the Chinese PD patients. METHODS: In this cross-sectional study, 503 consecutive outpatients with PD were enrolled who underwent standardized clinical evaluation. The study recruited 83 PD patients diagnosed with Pisa syndrome (PS). Scoliosis and coronal imbalance were diagnosed accurately by radiographic data. The PD patients were compared based on the Cobb angle and coronal balance for several demographic and clinical variables. RESULTS: PD patients with PS had a prevalence of 16.5%. The prevalence of coronal imbalance and scoliosis was 10.34 and 7.75%, respectively. PD patients with PS were older and had a more severe disease, significantly longer disease duration and treatment duration, and reduced quality of life. The most important finding was that the different morphology of the spinal level had an effect on the severity of coronal balance or Cobb angle. CONCLUSIONS: The present study indicated that the postural deformities in the coronal plane were related to the morphology of the spinal level, especially the position of the Cobb angle. To benefit the PD patients with PS, the full-length standing spine radiographs should be performed as early as possible.


Subject(s)
Parkinson Disease/complications , Scoliosis/diagnosis , Scoliosis/etiology , Aged , China , Cross-Sectional Studies , Female , Humans , Linear Models , Male , Middle Aged , Prevalence , Prospective Studies , Radiography , Risk Factors , Scoliosis/epidemiology
19.
Mov Disord ; 31(11): 1704-1710, 2016 11.
Article in English | MEDLINE | ID: mdl-27613677

ABSTRACT

OBJECTIVE: Familial cortical myoclonic tremor with epilepsy is a rare epilepsy syndrome. Herein, we report on nine Chinese familial cortical myoclonic tremor with epilepsy pedigrees to delineate its clinical and neurophysiological features. METHODS: Detailed clinical and neurophysiological data were obtained. Somatosensory evoked potential amplitudes and clinical profile were analyzed using multilevel statistical models. Age-at-onset anticipation was analyzed using Kaplan-Meier survival analysis. RESULTS: Fifty-five patients were interviewed directly, whose mean age at onset of cortical tremor and generalized tonic-clonic seizures were 31.0 ± 8.3 and 36.0 ± 7.9 years. Giant somatosensory evoked potential was detected in 87.5% (28 of 32) of patients, and long-latency cortical reflex was detected in 93.5% (29 of 31). Cortical tremor severity was significantly higher in patients with longer disease duration of cortical tremor (P = 0.0061). Somatosensory evoked potential amplitudes were significant higher in patients with higher level of cortical tremor severity (P = 0.0003) and those using antiepileptic drugs (P = 0.0150). Age-at-onset anticipation of cortical tremor with paternal transmission was found with statistical significance (P = 0.022). CONCLUSION: We provided the clinical and neurophysiological features of familial cortical myoclonic tremor with epilepsy patients. This study is reported for the presentation of this rare disease in a Chinese population with the largest single report on familial cortical myoclonic tremor with epilepsy worldwide. Age-at-onset anticipation of cortical tremor with paternal transmission was statistically significant, which further confirmed a possibility of unstable expanding repeat in the genetic mechanism of familial cortical myoclonic tremor with epilepsy. © 2016 International Parkinson and Movement Disorder Society.


Subject(s)
Anticipation, Genetic , Cerebral Cortex/physiopathology , Epilepsies, Myoclonic/physiopathology , Evoked Potentials, Somatosensory/physiology , Adolescent , Adult , Age of Onset , Aged , China , Epilepsies, Myoclonic/genetics , Female , Humans , Male , Middle Aged , Pedigree , Young Adult
20.
Mov Disord ; 30(13): 1777-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26180026

ABSTRACT

BACKGROUND: Depression is a relatively common and serious nonmotor symptom of Parkinson's disease (PD), which reduces the quality of patients' life. Although disturbances in some related brain networks have been reported, the pathophysiology of depression in PD is still unclear. Here, we aim to investigate whole-brain functional connectivity patterns in depressed PD patients. METHODS: We recruited 17 PD patients diagnosed with major depressive disorder, 17 PD patients without depression, and 17 healthy control subjects. Resting-state functional MRI and eigenvector centrality mapping were used to identify functional connectivity alterations among these groups. RESULTS: Results showed that depressed PD patients had decreased functional connectivity in the left dorsolateral prefrontal cortex and right superior temporal gyrus and increased functional connectivity in the right posterior cingulate cortex, compared to nondepressed patients. In addition, there was a significant negative correlation between functional connectivity and depression scores in the posterior cingulate cortex. CONCLUSIONS: This study suggests that functional connectivity changes in certain nodes of brain networks might contribute to depression in patients with PD.


Subject(s)
Brain/physiopathology , Depressive Disorder, Major/etiology , Depressive Disorder, Major/pathology , Neural Pathways/physiology , Parkinson Disease/complications , Aged , Analysis of Variance , Brain/blood supply , Case-Control Studies , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Neurologic Examination , Oxygen/blood , Psychiatric Status Rating Scales
SELECTION OF CITATIONS
SEARCH DETAIL