302 results on '"Chloride Channels genetics"'
Search Results
2. Mutations in CLCN6 as a Novel Genetic Cause of Neuronal Ceroid Lipofuscinosis in Patients and a Murine Model.
- Author
-
He H, Cao X, He F, Zhang W, Wang X, Peng P, Xie C, Yin F, Li D, Li J, Wang M, Klüssendorf M, Jentsch TJ, Stauber T, and Peng J
- Subjects
- Animals, Mice, Female, Humans, Autophagy genetics, Exome Sequencing, Membrane Proteins, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses pathology, Chloride Channels genetics, Disease Models, Animal, Mutation genetics
- Abstract
Objective: The aim of this study was to explore the pathogenesis of CLCN6-related disease and to assess whether its Cl
- /H+ -exchange activity is crucial for the biological role of ClC-6., Methods: We performed whole-exome sequencing on a girl with development delay, intractable epilepsy, behavioral abnormities, retinal dysfunction, progressive brain atrophy, suggestive of neuronal ceroid lipofuscinoses (NCLs). We generated and analyzed the first knock-in mouse model of a patient variant (p.E200A) and compared it with a Clcn6-/- mouse model. Additional functional tests were performed with heterologous expression of mutant ClC-6., Results: We identified a de novo heterozygous p.E200A variant in the proband. Expression of disease-causing ClC-6E200A or ClC-6Y553C mutants blocked autophagic flux and activated transcription factors EB (TFEB) and E3 (TFE3), leading to autophagic vesicle and cholesterol accumulation. Such alterations were absent with a transport-deficient ClC-6E267A mutant. Clcn6E200A/+ mice developed severe neurodegeneration with typical features of NCLs. Mutant ClC-6E200A , but not loss of ClC-6 in Clcn6-/- mice, increased lysosomal biogenesis by suppressing mTORC1-TFEB signaling, blocked autophagic flux through impairing lysosomal function, and increased apoptosis. Carbohydrate and lipid deposits accumulated in Clcn6E200A/+ brain, while only lipid storage was found in Clcn6-/- brain. Lysosome dysfunction, autophagy defects, and gliosis were early pathogenic events preceding neuron loss., Interpretation: CLCN6 is a novel genetic cause of NCLs, highlighting the importance of considering CLCN6 mutations in the diagnostic workup for molecularly undefined forms of NCLs. Uncoupling of Cl- transport from H+ countertransport in the E200A mutant has a dominant effect on the autophagic/lysosomal pathway. ANN NEUROL 2024;96:608-624., (© 2024 The Author(s). Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)- Published
- 2024
- Full Text
- View/download PDF
3. Clinical and molecular characterization of myotonia congenita using whole-exome sequencing in Egyptian patients.
- Author
-
Elaraby NM, Ahmed HA, Dawoud H, Ashaat NA, Azmy A, Galal ER, Elhusseny Y, Awady HE, Metwally AM, and Ashaat EA
- Subjects
- Humans, Female, Male, Egypt, Child, Adolescent, Child, Preschool, Young Adult, Infant, NAV1.4 Voltage-Gated Sodium Channel genetics, Adult, Pedigree, Electromyography, Myotonia Congenita genetics, Myotonia Congenita diagnosis, Exome Sequencing methods, Chloride Channels genetics, Mutation genetics
- Abstract
Background: Myotonia Congenita (MC) is a rare disease classified into two major forms; Thomsen and Becker disease caused by mutations in the CLCN1 gene, which affects muscle excitability and encodes voltage-gated chloride channels (CLC-1). While, there are no data regarding the clinical and molecular characterization of myotonia in Egyptian patients., Methods: Herein, we report seven Egyptian MC patients from six unrelated families. Following the clinical diagnosis, whole-exome sequencing (WES) was performed for genetic diagnosis. Various in silico prediction tools were utilized to interpret variant pathogenicity. The candidate variants were then validated using Sanger sequencing technique., Results: In total, seven cases were recruited. The ages at the examination were ranged from eight months to nineteen years. Clinical manifestations included warm-up phenomenon, hand grip, and percussion myotonia. Electromyography was performed in all patients and revealed myotonic discharges. Molecular genetic analysis revealed five different variants. Of them, we identified two novel variants in the CLCN1 gene ( c.1583G > C; p.Gly528Ala and c.2203_2216del;p.Thr735ValfsTer57) and three known variants in the CLCN1 and SCN4A gene. According to in silico tools, the identified novel variants were predicted to have deleterious effects., Conclusions: As the first study to apply WES among Egyptian MC patients, our findings reported two novel heterozygous variants that expand the CLCN1 mutational spectrum for MC diagnosis. These results further confirm that genetic testing is essential for early diagnosis of MC, which affects follow-up treatment and prognostic assessment in clinical practice., (© 2024. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2024
- Full Text
- View/download PDF
4. [A pedigree of myotonia congenita with a novel mutation p.F343C of the CLCN1 gene].
- Author
-
Nakamura Y, Sato H, Kakiuchi K, Miyano Y, Hosokawa T, and Arawaka S
- Subjects
- Humans, Female, Adult, Amino Acid Substitution, Male, Myotonia Congenita genetics, Chloride Channels genetics, Pedigree, Mutation
- Abstract
A Japanese woman experienced slowness of movement in her early teens and difficulty in opening her hands during pregnancy. On admission to our hospital at 42 years of age, she showed grip myotonia with warm-up phenomenon. However, she had neither muscle weakness, muscle atrophy, cold-induced symptomatic worsening nor episodes of transient weakness of the extremities. Needle electromyography of the first dorsal interosseous and anterior tibial muscles demonstrated myotonic discharges. Whole exome sequencing of the patient revealed a heterozygous single-base substitution in the CLCN1 gene (c.1028T>G, p.F343C). The same substitution was identified in affected members of her family (mother and brother) by Sanger sequencing, but not in healthy family members (father and a different brother). We diagnosed myotonia congenita (Thomsen disease) with a novel CLCN1 mutation in this pedigree. This mutation causes a single amino acid substitution in the I-J extracellular loop region of CLCN1. Amino acid changes in the I-J loop region are rare in an autosomal-dominantly inherited form of myotonia congenita. We think that this pedigree is precious to understand the pathogenesis of myotonia congenita.
- Published
- 2024
- Full Text
- View/download PDF
5. Retinal pigment epithelium-specific CLIC4 mutant is a mouse model of dry age-related macular degeneration.
- Author
-
Chuang JZ, Yang N, Nakajima N, Otsu W, Fu C, Yang HH, Lee MP, Akbar AF, Badea TC, Guo Z, Nuruzzaman A, Hsu KS, Dunaief JL, and Sung CH
- Subjects
- Animals, Cell Death, Chloride Channels deficiency, Disease Models, Animal, Fundus Oculi, Homeostasis, Lipid Metabolism, Macular Degeneration diagnostic imaging, Macular Degeneration physiopathology, Mice, Inbred C57BL, Mice, Knockout, Mitochondrial Proteins deficiency, Organ Specificity genetics, Retinal Drusen complications, Retinal Drusen diagnostic imaging, Retinal Drusen pathology, Retinal Pigment Epithelium diagnostic imaging, Retinal Pigment Epithelium physiopathology, Retinal Pigment Epithelium ultrastructure, Risk Factors, Transcription, Genetic, Vision, Ocular physiology, Mice, Chloride Channels genetics, Macular Degeneration genetics, Mitochondrial Proteins genetics, Mutation genetics, Retinal Pigment Epithelium metabolism
- Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Dry AMD has unclear etiology and no treatment. Lipid-rich drusen are the hallmark of dry AMD. An AMD mouse model and insights into drusenogenesis are keys to better understanding of this disease. Chloride intracellular channel 4 (CLIC4) is a pleomorphic protein regulating diverse biological functions. Here we show that retinal pigment epithelium (RPE)-specific Clic4 knockout mice exhibit a full spectrum of functional and pathological hallmarks of dry AMD. Multidisciplinary longitudinal studies of disease progression in these mice support a mechanistic model that links RPE cell-autonomous aberrant lipid metabolism and transport to drusen formation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
6. Association of Three Different Mutations in the CLCN1 Gene Modulating the Phenotype in a Consanguineous Family with Myotonia Congenita.
- Author
-
Souza LS, Calyjur P, Ribeiro AF Jr, Gurgel-Giannetti J, Pavanello RCM, Zatz M, and Vainzof M
- Subjects
- Adult, Chloride Channels metabolism, Consanguinity, Female, Humans, Male, Muscle, Skeletal metabolism, Myotonia Congenita pathology, Pedigree, Chloride Channels genetics, Mutation, Myotonia Congenita genetics, Phenotype
- Abstract
Myotonia congenita is a genetic disease caused by mutations in the CLCN1 gene, which encodes for the major chloride skeletal channel ClC-1, involved in the normal repolarization of muscle action potentials and consequent relaxation of the muscle after contraction. Two allelic forms are recognized, depending on the phenotype and the inheritance pattern: the autosomal dominant Thomsen disease with milder symptoms and the autosomal recessive Becker disorder with a severe phenotype. Before the recent advances of molecular testing, the diagnosis and genetic counseling of families was a challenge due to the large number of mutations in the CLCN1 gene, found both in homozygous or in heterozygous state. Here, we studied a consanguineous family in which three members presented a variable phenotype of myotonia, associated to a combination of three different mutations in the CLCN1 gene. A pathogenic splicing site mutation which causes the skipping of exon 17 was present in homozygosis in one very severely affected son. This mutation was present in compound heterozygosis in the consanguineous parents, but interestingly it was associated to a different second variant in the other allele: c.1453 A > G in the mother and c.1842 G > C in the father. Both displayed variable, but less severe phenotypes than their homozygous son. These results highlight the importance of analyzing the combination of different variants in the same gene in particular in families with patients displaying different phenotypes. This approach may improve the diagnosis, prognosis, and genetic counseling of the involved families., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
7. Unique variants in CLCN3, encoding an endosomal anion/proton exchanger, underlie a spectrum of neurodevelopmental disorders.
- Author
-
Duncan AR, Polovitskaya MM, Gaitán-Peñas H, Bertelli S, VanNoy GE, Grant PE, O'Donnell-Luria A, Valivullah Z, Lovgren AK, England EM, Agolini E, Madden JA, Schmitz-Abe K, Kritzer A, Hawley P, Novelli A, Alfieri P, Colafati GS, Wieczorek D, Platzer K, Luppe J, Koch-Hogrebe M, Abou Jamra R, Neira-Fresneda J, Lehman A, Boerkoel CF, Seath K, Clarke L, van Ierland Y, Argilli E, Sherr EH, Maiorana A, Diel T, Hempel M, Bierhals T, Estévez R, Jentsch TJ, Pusch M, and Agrawal PB
- Subjects
- Adolescent, Animals, Child, Child, Preschool, Female, Homozygote, Humans, Infant, Infant, Newborn, Male, Mice, Mice, Knockout, Neurodevelopmental Disorders etiology, Neurodevelopmental Disorders metabolism, Chloride Channels genetics, Disease Models, Animal, Ion Channels physiology, Mutation, Neurodevelopmental Disorders pathology, Phenotype
- Abstract
The genetic causes of global developmental delay (GDD) and intellectual disability (ID) are diverse and include variants in numerous ion channels and transporters. Loss-of-function variants in all five endosomal/lysosomal members of the CLC family of Cl
- channels and Cl- /H+ exchangers lead to pathology in mice, humans, or both. We have identified nine variants in CLCN3, the gene encoding CIC-3, in 11 individuals with GDD/ID and neurodevelopmental disorders of varying severity. In addition to a homozygous frameshift variant in two siblings, we identified eight different heterozygous de novo missense variants. All have GDD/ID, mood or behavioral disorders, and dysmorphic features; 9/11 have structural brain abnormalities; and 6/11 have seizures. The homozygous variants are predicted to cause loss of ClC-3 function, resulting in severe neurological disease similar to the phenotype observed in Clcn3-/- mice. Their MRIs show possible neurodegeneration with thin corpora callosa and decreased white matter volumes. Individuals with heterozygous variants had a range of neurodevelopmental anomalies including agenesis of the corpus callosum, pons hypoplasia, and increased gyral folding. To characterize the altered function of the exchanger, electrophysiological analyses were performed in Xenopus oocytes and mammalian cells. Two variants, p.Ile607Thr and p.Thr570Ile, had increased currents at negative cytoplasmic voltages and loss of inhibition by luminal acidic pH. In contrast, two other variants showed no significant difference in the current properties. Overall, our work establishes a role for CLCN3 in human neurodevelopment and shows that both homozygous loss of ClC-3 and heterozygous variants can lead to GDD/ID and neuroanatomical abnormalities., (Copyright © 2021 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
8. West Syndrome Caused By a Chloride/Proton Exchange-Uncoupling CLCN6 Mutation Related to Autophagic-Lysosomal Dysfunction.
- Author
-
He H, Cao X, Yin F, Wu T, Stauber T, and Peng J
- Subjects
- Amino Acid Sequence, Base Sequence, Child, Child, Preschool, Chloride Channels chemistry, Computer Simulation, HEK293 Cells, HeLa Cells, Humans, Infant, Infant, Newborn, Male, Subcellular Fractions metabolism, Autophagy genetics, Chloride Channels genetics, Chlorides metabolism, Lysosomes metabolism, Mutation genetics, Protons, Spasms, Infantile genetics
- Abstract
Vesicular chloride/proton exchangers of the CLC family are critically involved in the function of the endosomal-lysosomal pathway. Their dysfunction leads to severe disorders including intellectual disability and epilepsy for ClC-4, Dent's disease for ClC-5, and lysosomal storage disease and osteopetrosis for ClC-7. Here, we report a de novo variant p.Glu200Ala (p.E200A; c.599A>C) of the late endosomal ClC-6, encoded by CLCN6, in a patient with West syndrome (WS), severe developmental delay, autism, movement disorder, microcephaly, facial dysmorphism, and visual impairment. Mutation of this conserved glutamate uncouples chloride transport from proton antiport by ClC-6. This affects organellar ion homeostasis and was shown to be deleterious for other CLCs. In this study, we found that upon heterologous expression, the ClC-6 E200A variant caused autophagosome accumulation and impaired the clearance of autophagosomes by blocking autophagosome-lysosome fusion. Our study provides clinical and functional support for an association between CLCN6 variants and WS. Our findings also provide novel insights into the molecular mechanisms underlying the pathogenesis of WS, suggesting an involvement of autophagic-lysosomal dysfunction.
- Published
- 2021
- Full Text
- View/download PDF
9. Functional and Structural Characterization of ClC-1 and Na v 1.4 Channels Resulting from CLCN1 and SCN4A Mutations Identified Alone and Coexisting in Myotonic Patients.
- Author
-
Brenes O, Barbieri R, Vásquez M, Vindas-Smith R, Roig J, Romero A, Valle GD, Bermúdez-Guzmán L, Bertelli S, Pusch M, and Morales F
- Subjects
- Female, Genetic Testing methods, Humans, Male, Middle Aged, Myotonia Congenita metabolism, NAV1.4 Voltage-Gated Sodium Channel metabolism, Pedigree, Chloride Channels genetics, Mutation genetics, Myotonia genetics, Myotonia Congenita genetics, NAV1.4 Voltage-Gated Sodium Channel genetics
- Abstract
Non-dystrophic myotonias have been linked to loss-of-function mutations in the ClC-1 chloride channel or gain-of-function mutations in the Na
v 1.4 sodium channel. Here, we describe a family with members diagnosed with Thomsen's disease. One novel mutation (p.W322*) in CLCN1 and one undescribed mutation (p.R1463H) in SCN4A are segregating in this family. The CLCN1 -p.W322* was also found in an unrelated family, in compound heterozygosity with the known CLCN1 -p.G355R mutation. One reported mutation, SCN4A -p.T1313M, was found in a third family. Both CLCN1 mutations exhibited loss-of-function: CLCN1 -p.W322* probably leads to a non-viable truncated protein; for CLCN1 -p.G355R, we predict structural damage, triggering important steric clashes. The SCN4A -p.R1463H produced a positive shift in the steady-state inactivation increasing window currents and a faster recovery from inactivation. These gain-of-function effects are probably due to a disruption of interaction R1463-D1356, which destabilizes the voltage sensor domain (VSD) IV and increases the flexibility of the S4-S5 linker. Finally, modelling suggested that the p.T1313M induces a strong decrease in protein flexibility on the III-IV linker. This study demonstrates that CLCN1 -p.W322* and SCN4A -p.R1463H mutations can act alone or in combination as inducers of myotonia. Their co-segregation highlights the necessity for carrying out deep genetic analysis to provide accurate genetic counseling and management of patients.- Published
- 2021
- Full Text
- View/download PDF
10. G551D mutation impairs PKA-dependent activation of CFTR channel that can be restored by novel GOF mutations.
- Author
-
Wang W, Fu L, Liu Z, Wen H, Rab A, Hong JS, Kirk KL, and Rowe SM
- Subjects
- Adenosine Triphosphate metabolism, Animals, Chloride Channels drug effects, Chloride Channels genetics, Chloride Channels metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Cystic Fibrosis genetics, Humans, Ion Channel Gating drug effects, Ion Channel Gating genetics, Mutation drug effects, Signal Transduction drug effects, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Mutation genetics
- Abstract
G551D is a major disease-associated gating mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) protein, an ATP- and phosphorylation-dependent chloride channel. G551D causes severe cystic fibrosis (CF) disease by disrupting ATP-dependent channel opening; however, whether G551D affects phosphorylation-dependent channel activation is unclear. Here, we use macropatch recording and Ussing chamber approaches to demonstrate that G551D impacts on phosphorylation-dependent activation of CFTR, and PKA-mediated phosphorylation regulates the interaction between the x-loop in nucleotide-binding domain 2 (NBD2) and cytosolic loop (CL) 1. We show that G551D not only disrupts ATP-dependent channel opening but also impairs phosphorylation-dependent channel activation by largely reducing PKA sensitivity consistent with the reciprocal relationship between channel opening/gating, ligand binding, and phosphorylation. Furthermore, we identified two novel GOF mutations: D1341R in the x-loop near the ATP-binding cassette signature motif in NBD2 and D173R in CL1, each of which strongly increased PKA sensitivity both in the wild-type (WT) background and when introduced into G551D-CFTR. When D1341R was combined with a second GOF mutation (e.g., K978C in CL3), we find that the double GOF mutation maximally increased G551D channel activity such that VX-770 had no further effect. We further show that a double charge-reversal mutation of D1341R/D173R-CFTR exhibited similar PKA sensitivity when compared with WT-CFTR. Together, our results suggest that charge repulsion between D173 and D1341 of WT-CFTR normally inhibits channel activation at low PKA activity by reducing PKA sensitivity, and negative allostery by the G551D is coupled to reduced PKA sensitivity of CFTR that can be restored by second GOF mutations.
- Published
- 2020
- Full Text
- View/download PDF
11. Two novel mutations in the CLCNKB gene leading to classic Bartter syndrome presenting as syncope and hypertension in a 13-year-old boy.
- Author
-
Le BT, Duong CM, Nguyen TQ, and Bui CB
- Subjects
- Adolescent, Bartter Syndrome complications, Bartter Syndrome diagnosis, Glomerulosclerosis, Focal Segmental complications, Humans, Hypertension etiology, Male, Syncope etiology, Bartter Syndrome genetics, Chloride Channels genetics, Mutation genetics
- Abstract
Classic Bartter syndrome is a rare condition caused by mutations in the CLCNKB gene and characterised by metabolic alkalosis, hypokalaemia, hyper-reninaemia and hyperaldosteronism. Early signs and symptoms usually occur before a child's sixth birthday and include polyuria and developmental delay. We treated a 13-year-old Vietnamese boy with this syndrome presenting with atypical presentations including syncope and hypertension, but normal growth and development. All common causes of hypertension were ruled out. Genetic testing found two novel mutations in the CLCNKB gene, that is, Ser12Ala (exon 2) and Glu192Ter (exon 6). His estimated glomerular filtration rate was 61 mL/min/1.73 m
2 and a kidney biopsy showed focal segmental glomerulosclerosis. He was well managed with long-term enalapril therapy instead of non-steroidalanti-inflammatory drugs which are recommended in managing the increased prostaglandin E2 production in Bartter syndrome. Paediatricians should be alerted with the variability in its presentation. To preserve the kidney function, treatment must include preventing factors damaging the kidneys., Competing Interests: Competing interests: None declared., (© BMJ Publishing Group Limited 2020. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
12. The chloride intracellular channel protein CLIC4 inhibits filopodium formation induced by constitutively active mutants of formin mDia2.
- Author
-
Argenzio E and Innocenti M
- Subjects
- Actins metabolism, Chloride Channels deficiency, Chloride Channels genetics, Formins chemistry, HeLa Cells, Humans, Chloride Channels metabolism, Formins genetics, Formins metabolism, Mutation, Pseudopodia metabolism
- Abstract
Chloride intracellular channel 4 (CLIC4) functions in diverse actin-dependent processes. Upon Rho activation, CLIC4 reversibly translocates from the cytosol to the plasma membrane to regulate cell adhesion and migration. At the plasma membrane, CLIC4 counters the formation of filopodia, which requires actin assembly by the formin mammalian Diaphanous (mDia)2. To this end, mDia2 must be activated through conversion from the closed to the open conformation. Thus, CLIC4 could harness the activation or the open conformation of mDia2 to inhibit filopodium formation. Here, we find that CLIC4 silencing enhances the filopodia induced by two constitutively active mDia2 mutants. Furthermore, we report that CLIC4 binds the actin-regulatory region of mDia2 in vitro. These results suggest that CLIC4 modulates the activity of the open conformation of mDia2, shedding new light into how cells may control filopodia., (© 2020 The Authors. FEBS Letters published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2020
- Full Text
- View/download PDF
13. Analysis of CLCNKB mutations at dimer-interface, calcium-binding site, and pore reveals a variety of functional alterations in ClC-Kb channel leading to Bartter syndrome.
- Author
-
Bignon Y, Sakhi I, Bitam S, Bakouh N, Keck M, Frachon N, Paulais M, Planelles G, Teulon J, and Andrini O
- Subjects
- Animals, Bartter Syndrome metabolism, Cell Line, Humans, Oocytes metabolism, Protein Binding, Protein Transport, Xenopus, Bartter Syndrome genetics, Binding Sites, Calcium metabolism, Chloride Channels chemistry, Chloride Channels genetics, Mutation, Protein Multimerization
- Abstract
Pathological missense mutations in CLCNKB gene give a wide spectrum of clinical phenotypes in Bartter syndrome type III patients. Molecular analysis of the mutated ClC-Kb channels can be helpful to classify the mutations according to their functional alteration. We investigated the functional consequences of nine mutations in the CLCNKB gene causing Bartter syndrome. We first established that all tested mutations lead to decreased ClC-Kb currents. Combining electrophysiological and biochemical methods in Xenopus laevis oocytes and in MDCKII cells, we identified three classes of mutations. One class is characterized by altered channel trafficking. p.A210V, p.P216L, p.G424R, and p.G437R are totally or partially retained in the endoplasmic reticulum. p.S218N is characterized by reduced channel insertion at the plasma membrane and altered pH-sensitivity; thus, it falls in the second class of mutations. Finally, we found a novel class of functionally inactivated mutants normally present at the plasma membrane. Indeed, we found that p.A204T alters the pH-sensitivity, p.A254V abolishes the calcium-sensitivity. p.G219C and p.G465R are probably partially inactive at the plasma membrane. In conclusion, most pathogenic mutants accumulate partly or totally in intracellular compartments, but some mutants are normally present at the membrane surface and simultaneously show a large range of altered channel gating properties., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
14. Genetic Analyses in Dent Disease and Characterization of CLCN5 Mutations in Kidney Biopsies.
- Author
-
Gianesello L, Ceol M, Bertoldi L, Terrin L, Priante G, Murer L, Peruzzi L, Giordano M, Paglialonga F, Cantaluppi V, Musetti C, Valle G, Del Prete D, Anglani F, and Network DDI
- Subjects
- Biomarkers, Biopsy, DNA Mutational Analysis, Genetic Association Studies, Humans, Immunohistochemistry, Exome Sequencing, Chloride Channels genetics, Dent Disease genetics, Dent Disease pathology, Genetic Predisposition to Disease, Kidney Diseases genetics, Kidney Diseases pathology, Mutation
- Abstract
Dent disease (DD), an X-linked renal tubulopathy, is mainly caused by loss-of-function mutations in CLCN5 (DD1) and OCRL genes. CLCN5 encodes the ClC-5 antiporter that in proximal tubules (PT) participates in the receptor-mediated endocytosis of low molecular weight proteins. Few studies have analyzed the PT expression of ClC-5 and of megalin and cubilin receptors in DD1 kidney biopsies. About 25% of DD cases lack mutations in either CLCN5 or OCRL genes (DD3), and no other disease genes have been discovered so far. Sanger sequencing was used for CLCN5 gene analysis in 158 unrelated males clinically suspected of having DD. The tubular expression of ClC-5, megalin, and cubilin was assessed by immunolabeling in 10 DD1 kidney biopsies. Whole exome sequencing (WES) was performed in eight DD3 patients. Twenty-three novel CLCN5 mutations were identified. ClC-5, megalin, and cubilin were significantly lower in DD1 than in control biopsies. The tubular expression of ClC-5 when detected was irrespective of the type of mutation. In four DD3 patients, WES revealed 12 potentially pathogenic variants in three novel genes (SLC17A1, SLC9A3, and PDZK1), and in three genes known to be associated with monogenic forms of renal proximal tubulopathies (SLC3A, LRP2, and CUBN). The supposed third Dent disease-causing gene was not discovered., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
15. Co-Existence of Congenital Adrenal Hyperplasia and Bartter Syndrome due to Maternal Uniparental Isodisomy of HSD3B2 and CLCNKB Mutations.
- Author
-
Giri D, Bockenhauer D, Deshpande C, Achermann JC, Taylor NF, Rumsby G, Morgan H, Senniappan S, and Ajzensztejn M
- Subjects
- Adrenal Hyperplasia, Congenital genetics, Bartter Syndrome genetics, Female, Humans, Infant, Infant, Newborn, Adrenal Hyperplasia, Congenital complications, Bartter Syndrome complications, Chloride Channels genetics, Mutation, Progesterone Reductase genetics, Uniparental Disomy
- Abstract
Introduction: We present a patient with co-existence of 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2) deficiency and Bartter syndrome, a unique dual combination of opposing pathologies that has not been reported previously in the literature., Case: A female infant (46,XX) born at 34/40 weeks' gestation, weighing 2.67 kg (-1.54 standard deviation score) to non-consanguineous parents presented on day 4 of life with significant weight loss. Subsequent investigations revealed hyponatraemia, hypochloraemia, metabolic alkalosis, elevated 17-hydroxyprogesterone, ACTH, and renin. Urine steroid profile suggested HSD3B2 deficiency, which was confirmed by the identification of a homozygous HSD3B2 mutation. Due to the persistence of the hypochlo-raemic and hypokalemic alkalosis, an underlying renal tubulopathy was suspected. Sequence analysis of a targeted tubulopathy gene panel revealed a homozygous deletion in CLCNKB, consistent with Bartter syndrome type 3. The mother was found to be heterozygous for both mutations in -HSD3B2 and CLCNKB, and the father was negative for both. Single-nucleotide polymorphism microarray analysis confirmed 2 segments of homozygosity on chromosome 1 of maternal ancestry, encompassing both HSD3B2 and CLCKNB., Discussion: Identification of a homozygous rare mutation in an offspring of non-consanguineous parents should raise suspicion of uniparental disomy, especially if the phenotype is unusual, potentially encompassing more than one disorder. The persistence of hypokalemic alkalosis, the biochemical fingerprint of hyperaldosteronism in a child with a form of CAH in which aldosterone production is severely impaired, challenges our current understanding of mineralocorticoid-mediated effects in the collecting duct., (© 2020 S. Karger AG, Basel.)
- Published
- 2020
- Full Text
- View/download PDF
16. A Case of Autosomal Dominant Osteopetrosis Type 2 with a CLCN7 Gene Mutation
- Author
-
Kang S, Kang YK, Lee JA, Kim DH, and Lim JS
- Subjects
- Adolescent, Genetic Predisposition to Disease, Heterozygote, Humans, Male, Osteopetrosis diagnostic imaging, Osteopetrosis physiopathology, Pelvic Bones diagnostic imaging, Phenotype, Skull Base diagnostic imaging, Spine diagnostic imaging, Chloride Channels genetics, Mutation, Osteogenesis genetics, Osteopetrosis genetics, Pelvic Bones physiopathology, Skull Base physiopathology, Spine physiopathology
- Abstract
Osteopetrosis is a rare genetic disease characterized by increased bone density and bone fractures due to defective osteoclast function. Autosomal dominant osteopetrosis type 2 (ADO-2), Albers-Schonberg disease, is characterized by the sclerosis of bones, predominantly involving the spine, pelvis and the base of the skull. Here, we report a typical case of osteopetrosis in a 17.7-year-old male who carries a heterozygous c.746C>T mutation in exon 9 in the chloride voltage-gated channel 7 (CLCN7) gene. The patient’s spine showed multiple sclerotic changes including sandwich vertebra. His father had the same mutation but his skeletal radiographs were normal. This is the first reported case of ADO-2, confirmed by genetic testing in a Korean patient.
- Published
- 2019
- Full Text
- View/download PDF
17. Elevated aldosterone and blood pressure in a mouse model of familial hyperaldosteronism with ClC-2 mutation.
- Author
-
Schewe J, Seidel E, Forslund S, Marko L, Peters J, Muller DN, Fahlke C, Stölting G, and Scholl U
- Subjects
- Adrenal Glands pathology, Amino Acid Sequence, Animals, Base Sequence, CLC-2 Chloride Channels, Chloride Channels chemistry, Chlorides urine, Cytochrome P-450 CYP11B2 metabolism, Disease Models, Animal, Female, Heterozygote, Hyperaldosteronism urine, Male, Mice, Inbred C57BL, Phenotype, Renin blood, Sodium urine, Aldosterone blood, Blood Pressure, Chloride Channels genetics, Hyperaldosteronism blood, Hyperaldosteronism physiopathology, Mutation genetics
- Abstract
Gain-of-function mutations in the chloride channel ClC-2 were recently described as a cause of familial hyperaldosteronism type II (FH-II). Here, we report the generation of a mouse model carrying a missense mutation homologous to the most common FH-II-associated CLCN2 mutation. In these Clcn2
R180Q/+ mice, adrenal morphology is normal, but Cyp11b2 expression and plasma aldosterone levels are elevated. Male Clcn2R180Q/+ mice have increased aldosterone:renin ratios as well as elevated blood pressure levels. The counterpart knockout model (Clcn2-/- ), in contrast, requires elevated renin levels to maintain normal aldosterone levels. Adrenal slices of Clcn2R180Q/+ mice show increased calcium oscillatory activity. Together, our work provides a knockin mouse model with a mild form of primary aldosteronism, likely due to increased chloride efflux and depolarization. We demonstrate a role of ClC-2 in normal aldosterone production beyond the observed pathophysiology.- Published
- 2019
- Full Text
- View/download PDF
18. A somatic mutation in CLCN2 identified in a sporadic aldosterone-producing adenoma.
- Author
-
Dutta RK, Arnesen T, Heie A, Walz M, Alesina P, Söderkvist P, and Gimm O
- Subjects
- Adenoma surgery, Adult, CLC-2 Chloride Channels, Gene Frequency, Humans, Male, Norway epidemiology, Pituitary Neoplasms surgery, Transcriptome genetics, Adenoma genetics, Adenoma metabolism, Aldosterone metabolism, Chloride Channels genetics, Mutation genetics, Pituitary Neoplasms genetics, Pituitary Neoplasms metabolism
- Abstract
Objective: To screen for CLCN2 mutations in apparently sporadic cases of aldosterone-producing adenomas (APAs)., Description: Recently, CLCN2, encoding for the voltage-gated chloride channel protein 2 (ClC-2), was identified to be mutated in familial hyperaldosteronism II (FH II). So far, somatic mutations in CLCN2 have not been reported in sporadic cases of APAs. We screened 80 apparently sporadic APAs for mutations in CLCN2. One somatic mutation was identified at p.Gly24Asp in CLCN2. The male patient had a small adenoma in size but high aldosterone levels preoperatively. Postoperatively, the patient had normal aldosterone levels and was clinically cured., Conclusion: In this study, we identified a CLCN2 mutation in a sporadic APA comprising about 1% of all APAs investigated. This mutation was complementary to mutations in other susceptibility genes for sporadic APAs and may thus be a driving mutation in APA formation.
- Published
- 2019
- Full Text
- View/download PDF
19. Genotyping, generation and proteomic profiling of the first human autosomal dominant osteopetrosis type II-specific induced pluripotent stem cells.
- Author
-
Ou M, Li C, Tang D, Xue W, Xu Y, Zhu P, Li B, Xie J, Chen J, Sui W, Yin L, and Dai Y
- Subjects
- Adult, Animals, Apoptosis, Cell Proliferation, Female, Gene Expression Profiling, Gene Expression Regulation, Genotype, Humans, Induced Pluripotent Stem Cells metabolism, Kruppel-Like Factor 4, Male, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasms genetics, Neoplasms metabolism, Osteopetrosis genetics, Osteopetrosis metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers analysis, Chloride Channels genetics, Induced Pluripotent Stem Cells pathology, Mutation, Neoplasms pathology, Osteopetrosis pathology, Proteome analysis
- Abstract
Background: Autosomal dominant osteopetrosis type II (ADO2) is a rare human genetic disease that has been broadly studied as an important osteopetrosis model; however, there are no disease-specific induced pluripotent stem cells (ADO2-iPSCs) that may be valuable for understanding the pathogenesis and may be a potential source of cells for autologous cell-based therapies., Methods: To generate the first human ADO2-iPSCs from a Chinese family with ADO2 and to identify their characteristics, blood samples were collected from the proband and his parents and were used for genotyping by whole-exome sequencing (WES); the urine-derived cells of the proband were reprogrammed with episomal plasmids that contained transcription factors, such as KLF4, OCT4, c-MYC, and SOX2. The proteome-wide protein quantification and lysine 2-hydroxyisobutyrylation detection of the ADO2-iPSCs and normal control iPSCs (NC-iPSCs) were performed by high-resolution LC-MS/MS and bioinformatics analysis., Results: WES with filtering strategies identified a mutation in CLCN7 (R286W) in the proband and his father, which was absent in the proband's mother and the healthy controls; this was confirmed by Sanger sequencing. The ADO2-iPSCs were successfully generated, which carried a normal male karyotype (46, XY) and the mutation of CLCN7 (R286W); the ADO2-iPSCs positively expressed alkaline phosphatase and other surface markers; and no vector and transgene were detected. The ADO2-iPSCs could differentiate into all three germ cell layers, both in vitro and in vivo. The proteomic profiling revealed similar expression of pluripotency markers in the two cell lines and identified 7405 proteins and 3664 2-hydroxyisobutyrylated peptides in 1036 proteins in the ADO2-iPSCs., Conclusions: Our data indicated that the mutation CLCN7 (R286W) may be a cause of the osteopetrosis family. The generated vector-free and transgene-free ADO2-iPSCs with known proteomic characteristics may be valuable for personalized and cell-based regenerative medicine in the future.
- Published
- 2019
- Full Text
- View/download PDF
20. A novel CLCNKB mutation in a Chinese girl with classic Bartter syndrome: a case report.
- Author
-
Zhu B, Jiang H, Cao M, Zhao X, and Jiang H
- Subjects
- Adolescent, Bartter Syndrome pathology, Dwarfism, Pituitary genetics, Female, Genetic Association Studies, Heart Septal Defects, Atrial, Heterozygote, Humans, Juxtaglomerular Apparatus, Pedigree, Renal Insufficiency, Chronic, Asian People genetics, Bartter Syndrome genetics, Chloride Channels genetics, Genetic Predisposition to Disease genetics, Mutation
- Abstract
Background: Bartter syndrome (BS) is a rare autosomal recessive disorder of salt reabsorption at the thick ascending limb of the Henle loop, characterized by hypokalemia, salt loss, metabolic alkalosis, hyperreninemic hyperaldosteronism with normal blood pressure. BS type III, often known as classic BS (CBS), is caused by loss-of-function mutations in CLCNKB (chloride voltage-gated channel Kb) encoding basolateral ClC-Kb., Case Presentation: We reported a 15-year-old CBS patient with a compound heterozygous mutation of CLCNKB gene. She first presented with vomiting, hypokalemic metabolic alkalosis at the age of 4 months, and was clinically diagnosed as CBS. Indomethacin, spironolactone and oral potassium were started from then. During follow-up, the serum electrolyte levels were generally normal, but the patient showed failure to thrive and growth hormone (GH) deficiency was diagnosed. The recombinant human GH therapy was performed, and the growth velocity was improved. When she was 14, severe proteinuria and chronic kidney disease (CKD) were developed. Renal biopsy showed focal segmental glomerulosclerosis (FSGS) with juxtaglomerular apparatus cell hyperplasia, and genetic testing revealed a point deletion of c.1696delG (p. Glu566fs) and a fragment deletion of exon 2-3 deletions in CLCNKB gene. Apart from the CBS, ostium secundum atrial septal defect (ASD) was diagnosed by echocardiography., Conclusions: This is the first report of this compound heterozygous of CLCNKB gene in BS Children. Our findings contribute to a growing list of CLCNKB mutations associated with CBS. Some recessive mutations can induce CBS in combination with other mutations.
- Published
- 2019
- Full Text
- View/download PDF
21. Digenetic inheritance of SLC12A3 and CLCNKB genes in a Chinese girl with Gitelman syndrome.
- Author
-
Kong Y, Xu K, Yuan K, Zhu J, Gu W, Liang L, and Wang C
- Subjects
- Child, China, Chloride Channels metabolism, DNA Mutational Analysis, Female, Gitelman Syndrome diagnosis, Gitelman Syndrome metabolism, Humans, Pedigree, Phenotype, Solute Carrier Family 12, Member 3 genetics, Solute Carrier Family 12, Member 3 metabolism, Chloride Channels genetics, DNA genetics, Genetic Predisposition to Disease, Gitelman Syndrome genetics, Mutation
- Abstract
Background: Gitelman syndrome (GS) is an autosomal recessive disorder and mild variant of classic Bartter syndrome. The latter is caused by defects in the genes CLCNKB and/or CLCNKA (chloride voltage-gated channel Ka and Kb). Patients with GS usually have loss-of-function mutations in SLC12A3. No patient has been reported with compound heterozygous mutations in these genes. We report a girl with GS with a paternally inherited heterozygous mutation in SLC12A3, and maternally inherited heterozygous variants in both CLCNKB and CLCNKA., Case Presentation: In this report, we reported a female patient (8 y and 10 mo) who had growth retardation (111.8 cm, - 1.62 standard deviation height for age) and normal blood pressure, with persistent hypokalemia, hypomagnesemia, hypocalciuria, hypochloremic alkalosis, and elevated levels of plasma renin and aldosterone. Her younger brother, father, and paternal grandmother all had histories of mild low levels of plasma potassium (3.0-3.5 mmol/L), which were rectified by potassium-rich foods. The genomic DNA of the patient, younger brother, parents, and grandparents were screened for gene variations and pedigree analysis using trio whole exome sequencing (WES). The candidate variants were validated by Sanger sequencing. Protein-protein interaction analysis utilized the following databases: Biogrid, MINT, HPRD, STRING, IntAct, iRefIndex, and ppiTrim. The trio WES screening showed that the patient has paternally inherited SLC12A3 p.N359K, and maternally inherited CLCNKB p.L94I. The paternal grandmother and younger brother are both carriers of SLC12A3 p.N359K. According to the STRING database, SLC12A3 and CLCNKB proteins may interact or coexpress with proteins associated with GS., Conclusions: Based on clinical phenotypes, genetic evidence of the pedigree, and previous reported studies, this case of GS indicates a digenetic inheritance of SLC12A3 and CLCNKB that resulted in renal tubular dysfunction perhaps, due to a genetic double-hit mechanism. The putative pathogenicity of the CLCNKB p.L94I variant requires confirmation.
- Published
- 2019
- Full Text
- View/download PDF
22. Generation of a human induced pluripotent stem cell line (BIHi002-A) from a patient with CLCN7-related infantile malignant autosomal recessive osteopetrosis.
- Author
-
Hennig AF, Rössler U, Boiti F, von der Hagen M, Gossen M, Kornak U, and Stachelscheid H
- Subjects
- Humans, Infant, Male, Cell Line, Chloride Channels genetics, Chloride Channels metabolism, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Leukocytes, Mononuclear metabolism, Leukocytes, Mononuclear pathology, Mutation, Osteopetrosis genetics, Osteopetrosis metabolism, Osteopetrosis pathology
- Abstract
Autosomal recessive osteopetrosis (ARO) is a genetic bone disease that can be caused by mutations in the CLCN7 gene preventing osteoclast-mediated bone resorption. We generated a human induced pluripotent stem cell (hiPSC) line, BIHi002-A, from peripheral blood mononuclear cells of an ARO patient carrying the CLCN7 mutations c.875G>A and c.1208G>A using Sendai viral vectors. The pluripotent identity of the BIHi002-A line was confirmed by their expression of typical markers for undifferentiated hiPSCs, their capacity to differentiate into cells of the three germ layers and by PluriTest analysis. The BIHi002-A line provides a tool for disease modelling and therapy development., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
23. CLCN7 and TCIRG1 mutations in a single family: Evidence for digenic inheritance of osteopetrosis.
- Author
-
Yang Y, Ye W, Guo J, Zhao L, Tu M, Zheng Y, and Li L
- Subjects
- Adolescent, Case-Control Studies, Child, DNA Mutational Analysis, Ethnicity, Female, Genetic Predisposition to Disease, Humans, Male, Osteopetrosis pathology, Pedigree, Exome Sequencing, Chloride Channels genetics, Multifactorial Inheritance, Mutation, Osteopetrosis genetics, Vacuolar Proton-Translocating ATPases genetics
- Abstract
Osteopetrosis is a monogenic condition with various inheritance patterns, including autosomal dominant, autosomal recessive and X‑linked. Several disease‑causing genes have been identified and three distinguished types of osteopetrosis have been reported. In the present study, a family with osteopetrosis was investigated. Two novel mutations in chloride voltage‑gated channel 7 (CLCN7) and T cell immune regulator 1 (TCIRG1) were identified by exome sequencing, Sanger sequencing and microsatellite marker analysis. The CLCN7 mutation occurred in amino acid R286, the same position as previously reported. The TCIRG1 mutation occurred on a splicing site of exon 15, thereby leading to a truncated transcript. These two mutations were undetected in 496 ethnic‑matched controls. To the best of our knowledge, this is the first report of human osteopetrosis involving digenic inheritance in a single family, which has important implications for decisions on clinical therapeutic regimen, prognosis evaluation and antenatal diagnosis.
- Published
- 2019
- Full Text
- View/download PDF
24. Becker's myotonia: novel mutations and clinical variability in patients born to consanguineous parents.
- Author
-
Sahin I, Erdem HB, Tan H, and Tatar A
- Subjects
- Adolescent, Child, Consanguinity, Genetic Association Studies, Humans, Male, Muscle, Skeletal pathology, Myotonia Congenita pathology, Turkey, Chloride Channels genetics, Mutation, Myotonia Congenita genetics
- Abstract
Myotonia congenita is an inherited muscle disease present from childhood that is characterized by impaired muscle relaxation after contraction resulting in muscle stiffness; moreover, skeletal striated muscle groups may be involved. Myotonia congenita occurs due to chloride (Cl) channel mutations that reduce the stabilizing Cl conductance, and it is caused by mutations in the CLCN1 gene. This paper describes four patients from two different healthy consanguineous Turkish families with muscle stiffness and easy fatigability. A genetic investigation was performed. Mutation analyses showed a homozygous p.Tyr150* (c.450C > A) mutation in patients 1, 2 and 3 and a homozygous p.Leu159Cysfs*11 (c.475delC) mutation in patient 4 in the CLCN1 gene. These mutations have never been reported before and in silico analyses showed that the mutations were disease causing. They may be predicted to cause nonsense-mediated mRNA decay. Our data expand the spectrum of CLCN1 mutations and provide insights for genotype-phenotype correlations of myotonia congenita.
- Published
- 2018
- Full Text
- View/download PDF
25. Novel compound heterozygous CLCNKB gene mutations (c.1755A>G/c.848_850delTCT) cause classic Bartter syndrome.
- Author
-
Wang C, Chen Y, Zheng B, Zhu M, Fan J, Wang J, Jia Z, Huang S, and Zhang A
- Subjects
- Bartter Syndrome diagnosis, Humans, Male, Phenotype, Protein Transport genetics, Bartter Syndrome genetics, Chloride Channels genetics, Heterozygote, Mutation genetics
- Abstract
Inactivated variants in CLCNKB gene encoding the basolateral chloride channel ClC-Kb cause classic Bartter syndrome characterized by hypokalemic metabolic alkalosis and hyperreninemic hyperaldosteronism. Here, we identified two cBS siblings presenting hypokalemia in a Chinese family due to novel compound heterozygous CLCNKB mutations (c.848_850delTCT/c.1755A>G). Compound heterozygosity was confirmed by amplifying and sequencing the patient's genomic DNA. The synonymous mutation c.1755A>G (Thr585Thr) was located at +2 bp from the 5' splice donor site in exon 15. Further transcript analysis demonstrated that this single nucleotide mutation causes exclusion of exon 15 in the cDNA from the proband and his mother. Furthermore, we investigated the expression and protein trafficking change of c.848_850delTCT (ΔTCT) and exon 15 deletion (ΔE15) mutation in vitro. The ΔE15 mutation markedly decreased the expression of ClC-Kb and resulted in a low-molecular-weight band (~55 kDa) trapping in the endoplasmic reticulum, while the ΔTCT mutant only decreased the total and plasma membrane ClC-Kb protein expression but did not affect the subcellular localization. Finally, we studied the physiological functions of mutations by using whole cell patch-clamp and found that the ΔE15 or ΔTCT mutation decreased the current of the ClC-Kb/barttin channel. These results suggested that the compound defective mutations of the CLCNKB gene are the molecular mechanism of the two cBS siblings.
- Published
- 2018
- Full Text
- View/download PDF
26. The analysis of myotonia congenita mutations discloses functional clusters of amino acids within the CBS2 domain and the C-terminal peptide of the ClC-1 channel.
- Author
-
Altamura C, Lucchiari S, Sahbani D, Ulzi G, Comi GP, D'Ambrosio P, Petillo R, Politano L, Vercelli L, Mongini T, Dotti MT, Cardani R, Meola G, Lo Monaco M, Matthews E, Hanna MG, Carratù MR, Conte D, Imbrici P, and Desaphy JF
- Subjects
- Adolescent, Adult, Amino Acids genetics, Female, Humans, Ion Channel Gating genetics, Male, Middle Aged, Myotonia Congenita drug therapy, Myotonia Congenita physiopathology, Patch-Clamp Techniques, Peptides genetics, Protein Domains genetics, Chloride Channels genetics, DNA Mutational Analysis, Mutation genetics, Myotonia Congenita genetics
- Abstract
Myotonia congenita (MC) is a skeletal-muscle hyperexcitability disorder caused by loss-of-function mutations in the ClC-1 chloride channel. Mutations are scattered over the entire sequence of the channel protein, with more than 30 mutations located in the poorly characterized cytosolic C-terminal domain. In this study, we characterized, through patch clamp, seven ClC-1 mutations identified in patients affected by MC of various severities and located in the C-terminal region. The p.Val829Met, p.Thr832Ile, p.Val851Met, p.Gly859Val, and p.Leu861Pro mutations reside in the CBS2 domain, while p.Pro883Thr and p.Val947Glu are in the C-terminal peptide. We showed that the functional properties of mutant channels correlated with the clinical phenotypes of affected individuals. In addition, we defined clusters of ClC-1 mutations within CBS2 and C-terminal peptide subdomains that share the same functional defect: mutations between 829 and 835 residues and in residue 883 induced an alteration of voltage dependence, mutations between 851 and 859 residues, and in residue 947 induced a reduction of chloride currents, whereas mutations on 861 residue showed no obvious change in ClC-1 function. This study improves our understanding of the mechanisms underlying MC, sheds light on the role of the C-terminal region in ClC-1 function, and provides information to develop new antimyotonic drugs., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
27. Novel Lys215Asn mutation in an Italian family with Thomsen myotonia.
- Author
-
Mantero V, Lucchiari S, Balgera R, Comi GP, Salmaggi A, and Rigamonti A
- Subjects
- Adult, Asparagine genetics, DNA Mutational Analysis, Electromyography, Humans, Italy, Lysine genetics, Male, Middle Aged, Chloride Channels genetics, Family Health, Mutation genetics, Myotonia Congenita genetics
- Published
- 2018
- Full Text
- View/download PDF
28. A novel CLCN5 pathogenic mutation supports Dent disease with normal endosomal acidification.
- Author
-
Bignon Y, Alekov A, Frachon N, Lahuna O, Jean-Baptiste Doh-Egueli C, Deschênes G, Vargas-Poussou R, and Lourdel S
- Subjects
- Animals, Dent Disease genetics, Dent Disease metabolism, Dent Disease pathology, Endocytosis genetics, Endocytosis physiology, HEK293 Cells, Humans, Xenopus laevis, Chloride Channels genetics, Chloride Channels metabolism, Endosomes metabolism, Endosomes pathology, Mutation genetics
- Abstract
Dent disease is an X-linked recessive renal tubular disorder characterized by low-molecular-weight proteinuria, hypercalciuria, nephrolithiasis, nephrocalcinosis, and progressive renal failure. Inactivating mutations of CLCN5, the gene encoding the 2Cl
- /H+ exchanger ClC-5, have been reported in patients with Dent disease 1. In vivo studies in mice harboring an artificial mutation in the "gating glutamate" of ClC-5 (c.632A > C, p.Glu211Ala) and mathematical modeling suggest that endosomal chloride concentration could be an important parameter in endocytosis, rather than acidification as earlier hypothesized. Here, we described a novel pathogenic mutation affecting the "gating glutamate" of ClC-5 (c.632A>G, p.Glu211Gly) and investigated its molecular consequences. In HEK293T cells, the p.Glu211Gly ClC-5 mutant displayed unaltered N-glycosylation and normal plasma membrane and early endosomes localizations. In Xenopus laevis oocytes and HEK293T cells, we found that contrasting with wild-type ClC-5, the mutation abolished the outward rectification, the sensitivity to extracellular H+ and converted ClC-5 into a Cl- channel. Investigation of endosomal acidification in HEK293T cells using the pH-sensitive pHluorin2 probe showed that the luminal pH of cells expressing a wild-type or p.Glu211Gly ClC-5 was not significantly different. Our study further confirms that impaired acidification of endosomes is not the only parameter leading to defective endocytosis in Dent disease 1., (© 2018 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
29. Activation of renal ClC-K chloride channels depends on an intact N terminus of their accessory subunit barttin.
- Author
-
Wojciechowski D, Thiemann S, Schaal C, Rahtz A, de la Roche J, Begemann B, Becher T, and Fischer M
- Subjects
- Biological Transport, Cells, Cultured, Chloride Channels genetics, HEK293 Cells, Humans, Protein Domains, Cell Membrane metabolism, Chloride Channels metabolism, Ion Channel Gating physiology, Kidney metabolism, Mutation
- Abstract
ClC-K channels belong to the CLC family of chloride channels and chloride/proton antiporters. They contribute to sodium chloride reabsorption in Henle's loop of the kidney and to potassium secretion into the endolymph by the stria vascularis of the inner ear. Their accessory subunit barttin stabilizes the ClC-K/barttin complex, promotes its insertion into the surface membrane, and turns the pore-forming subunits into a conductive state. Barttin mutations cause Bartter syndrome type IV, a salt-wasting nephropathy with sensorineural deafness. Here, studying ClC-K/barttin channels heterologously expressed in MDCK-II and HEK293T cells with confocal imaging and patch-clamp recordings, we demonstrate that the eight-amino-acids-long barttin N terminus is required for channel trafficking and activation. Deletion of the complete N terminus (Δ2-8 barttin) retained barttin and human hClC-Ka channels in intracellular compartments. Partial N-terminal deletions did not compromise subcellular hClC-Ka trafficking but drastically reduced current amplitudes. Sequence deletions encompassing Thr-6, Phe-7, or Arg-8 in barttin completely failed to activate hClC-Ka. Analyses of protein expression and whole-cell current noise revealed that inactive channels reside in the plasma membrane. Substituting the deleted N terminus with a polyalanine sequence was insufficient for recovering chloride currents, and single amino acid substitutions highlighted that the correct sequence is required for proper function. Fast and slow gate activation curves obtained from rat V166E rClC-K1/barttin channels indicated that mutant barttin fails to constitutively open the slow gate. Increasing expression of barttin over that of ClC-K partially recovered this insufficiency, indicating that N-terminal modifications of barttin alter both binding affinities and gating properties., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
30. Prevalence and mutation spectrum of skeletal muscle channelopathies in the Netherlands.
- Author
-
Stunnenberg BC, Raaphorst J, Deenen JCW, Links TP, Wilde AA, Verbove DJ, Kamsteeg EJ, van den Wijngaard A, Faber CG, van der Wilt GJ, van Engelen BGM, Drost G, and Ginjaar HB
- Subjects
- Adult, Aged, Andersen Syndrome genetics, Calcium Channels genetics, Calcium Channels, L-Type, Channelopathies genetics, Chloride Channels genetics, Female, Humans, Hypokalemic Periodic Paralysis genetics, Male, Middle Aged, Myotonia genetics, Myotonic Disorders genetics, NAV1.4 Voltage-Gated Sodium Channel genetics, Netherlands epidemiology, Pedigree, Potassium Channels, Inwardly Rectifying genetics, Prevalence, Young Adult, Andersen Syndrome epidemiology, Channelopathies epidemiology, Hypokalemic Periodic Paralysis epidemiology, Mutation, Myotonia epidemiology, Myotonic Disorders epidemiology
- Abstract
Few reliable data exist on the prevalence of skeletal muscle channelopathies. We determined the minimum point prevalence of genetically-defined skeletal muscle channelopathies in the Netherlands and report their mutation spectrum. Minimum point prevalence rates were calculated as number of genetically-confirmed skeletal muscle channelopathy patients (CLCN1, SCN4A, CACNA1S and KCNJ2 gene mutations) in the Netherlands (1990-2015) divided by the total number of at-risk individuals. Rates were expressed as cases/100.000 and 95% confidence intervals were calculated based on Poisson distribution. Results of standardized genetic diagnostic procedures were used to analyze mutation spectra. We identified 405 patients from 234 unrelated pedigrees, resulting in a minimum point prevalence of 2.38/100.000 (95% CI 2.16-2.63) for skeletal muscle channelopathies in the Netherlands. Minimum point prevalence rates for the disease groups, non-dystrophic myotonia and periodic paralysis, were 1.70/100.000 and 0.69/100.000 respectively. Sixty-one different CLCN1 mutations (including 12 novel mutations) were detected in myotonia congenita. Twenty-eight different SCN4A missense mutations (including three novel mutations) were identified in paramyotonia congenita/sodium channel myotonia, hypokalemic periodic paralysis and hyperkalemic periodic paralysis. Four different CACNA1S missense mutations were detected in hypokalemic periodic paralysis and five KCNJ2 missense mutations in Andersen-Tawil syndrome. The minimum point prevalence rates for genetically-defined skeletal muscle channelopathies confirm their rare disease status in the Netherlands. Rates are almost twice as high as in the UK and more in line with pre-genetic prevalence estimates in parts of Scandinavia. Future diagnostic and therapeutic studies may benefit from knowledge of the mutation spectrum of skeletal muscle channelopathies., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. CLCN2 chloride channel mutations in familial hyperaldosteronism type II.
- Author
-
Scholl UI, Stölting G, Schewe J, Thiel A, Tan H, Nelson-Williams C, Vichot AA, Jin SC, Loring E, Untiet V, Yoo T, Choi J, Xu S, Wu A, Kirchner M, Mertins P, Rump LC, Onder AM, Gamble C, McKenney D, Lash RW, Jones DP, Chune G, Gagliardi P, Choi M, Gordon R, Stowasser M, Fahlke C, and Lifton RP
- Subjects
- Adolescent, Adrenal Glands metabolism, Adrenal Glands pathology, Adult, Amino Acid Sequence, CLC-2 Chloride Channels, Child, Cohort Studies, DNA Mutational Analysis, Female, Humans, Hyperaldosteronism pathology, Infant, Male, Pedigree, Young Adult, Chloride Channels genetics, Hyperaldosteronism genetics, Mutation
- Abstract
Primary aldosteronism, a common cause of severe hypertension
1 , features constitutive production of the adrenal steroid aldosterone. We analyzed a multiplex family with familial hyperaldosteronism type II (FH-II)2 and 80 additional probands with unsolved early-onset primary aldosteronism. Eight probands had novel heterozygous variants in CLCN2, including two de novo mutations and four independent occurrences of a mutation encoding an identical p.Arg172Gln substitution; all relatives with early-onset primary aldosteronism carried the CLCN2 variant found in the proband. CLCN2 encodes a voltage-gated chloride channel expressed in adrenal glomerulosa that opens at hyperpolarized membrane potentials. Channel opening depolarizes glomerulosa cells and induces expression of aldosterone synthase, the rate-limiting enzyme for aldosterone biosynthesis. Mutant channels show gain of function, with higher open probabilities at the glomerulosa resting potential. These findings for the first time demonstrate a role of anion channels in glomerulosa membrane potential determination, aldosterone production and hypertension. They establish the cause of a substantial fraction of early-onset primary aldosteronism.- Published
- 2018
- Full Text
- View/download PDF
32. Probing the changes in gene expression due to α-crystallin mutations in mouse models of hereditary human cataract.
- Author
-
Andley UP, Tycksen E, McGlasson-Naumann BN, and Hamilton PD
- Subjects
- Animals, Carboxypeptidases genetics, Carboxypeptidases metabolism, Cataract metabolism, Chloride Channels genetics, Chloride Channels metabolism, Disease Models, Animal, Gene Expression, Gene Knock-In Techniques, Histones genetics, Histones metabolism, Humans, Lens, Crystalline metabolism, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mice, Transgenic, Proteins genetics, Proteins metabolism, Proteomics, Repressor Proteins genetics, Repressor Proteins metabolism, alpha-Crystallin A Chain metabolism, alpha-Crystallin B Chain metabolism, Cataract genetics, Mutation, alpha-Crystallin A Chain genetics, alpha-Crystallin B Chain genetics
- Abstract
The mammalian eye lens expresses a high concentration of crystallins (α, β and γ-crystallins) to maintain the refractive index essential for lens transparency. Crystallins are long-lived proteins that do not turnover throughout life. The structural destabilization of crystallins by UV exposure, glycation, oxidative stress and mutations in crystallin genes leads to protein aggregation and development of cataracts. Several destabilizing mutations in crystallin genes are linked with human autosomal dominant hereditary cataracts. To investigate the mechanism by which the α-crystallin mutations Cryaa-R49C and Cryab-R120G lead to cataract formation, we determined whether these mutations cause an altered expression of specific transcripts in the lens at an early postnatal age by RNA-seq analysis. Using knock-in mouse models previously generated in our laboratory, in the present work, we identified genes that exhibited altered abundance in the mutant lenses, including decreased transcripts for Clic5, an intracellular water channel in Cryaa-R49C heterozygous mutant lenses, and increased transcripts for Eno1b in Cryab-R120G heterozygous mutant lenses. In addition, RNA-seq analysis revealed increased histones H2B, H2A, and H4 gene expression in Cryaa-R49C mutant lenses, suggesting that the αA-crystallin mutation regulates histone expression via a transcriptional mechanism. Additionally, these studies confirmed the increased expression of histones H2B, H2A, and H4 by proteomic analysis of Cryaa-R49C knock-in and Cryaa;Cryab gene knockout lenses reported previously. Taken together, these findings offer additional insight into the early transcriptional changes caused by Cryaa and Cryab mutations associated with autosomal dominant human cataracts, and indicate that the transcript levels of certain genes are affected by the expression of mutant α-crystallin in vivo.
- Published
- 2018
- Full Text
- View/download PDF
33. Genetic Analysis of Dent's Disease and Functional Research of CLCN5 Mutations.
- Author
-
Zhang Y, Fang X, Xu H, and Shen Q
- Subjects
- Child, Child, Preschool, Chloride Channels metabolism, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Dent Disease diagnosis, Dent Disease metabolism, Female, Genetic Diseases, X-Linked diagnosis, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked metabolism, HEK293 Cells, Humans, Male, Mutant Proteins genetics, Mutant Proteins metabolism, Nephrolithiasis diagnosis, Nephrolithiasis genetics, Nephrolithiasis metabolism, Phenotype, Phosphoric Monoester Hydrolases genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Subcellular Fractions metabolism, Chloride Channels genetics, Dent Disease genetics, Mutation
- Abstract
Dent's disease is an X-linked inherited renal disease. Patients with Dent's disease often carry mutations in genes encoding the Cl
- /H+ exchanger ClC-5 and/or inositol polyphosphate 5-phosphatase (OCRL1). However, the mutations involved and the biochemical effects of these mutations are not fully understood. To characterize genetic changes in Dent's disease patients, in this study, samples from nine Chinese patients were subjected to genetic analysis. Among the nine patients, six were classified as having Dent-1 disease, one had Dent-2 disease, and two could not be classified. Expression of ClC-5 carrying Dent's disease-associated mutations in HEK293 cells had varying effects: (1) no detectable expression of mutant protein; (2) retention of a truncated protein in the endoplasmic reticulum; or (3) diminished protein expression with normal distribution in early endosomes. Dent's disease patients showed genetic heterogeneity and over 20% of patients did not have CLCN5 or OCRL1 mutations, suggesting the existence of other genetic factors. Using next-generation sequencing, we identified possible modifier genes that have not been previously reported in Dent's disease patients. Heterozygous variants in CFTR, SCNN1A, and SCNN1B genes associated with cystic fibrosis (CF) or CF-like disease were detected in four of our nine patients. These results may form the basis for future characterization of Dent's disease and genetic counseling approaches.- Published
- 2017
- Full Text
- View/download PDF
34. Coexistence of CLCN1 and SCN4A mutations in one family suffering from myotonia.
- Author
-
Maggi L, Ravaglia S, Farinato A, Brugnoni R, Altamura C, Imbrici P, Camerino DC, Padovani A, Mantegazza R, Bernasconi P, Desaphy JF, and Filosto M
- Subjects
- Adult, Female, Genetic Association Studies methods, Heterozygote, Humans, Myotonia diagnosis, Pedigree, Phenotype, Chloride Channels genetics, Mutation genetics, Myotonia genetics, NAV1.4 Voltage-Gated Sodium Channel genetics
- Abstract
Non-dystrophic myotonias are characterized by clinical overlap making it challenging to establish genotype-phenotype correlations. We report clinical and electrophysiological findings in a girl and her father concomitantly harbouring single heterozygous mutations in SCN4A and CLCN1 genes. Functional characterization of N1297S hNav1.4 mutant was performed by patch clamp. The patients displayed a mild phenotype, mostly resembling a sodium channel myotonia. The CLCN1 c.501C>G (p.F167L) mutation has been already described in recessive pedigrees, whereas the SCN4A c.3890A>G (p.N1297S) variation is novel. Patch clamp experiments showed impairment of fast and slow inactivation of the mutated Nav1.4 sodium channel. The present findings suggest that analysis of both SCN4A and CLCN1 genes should be considered in myotonic patients with atypical clinical and neurophysiological features.
- Published
- 2017
- Full Text
- View/download PDF
35. Leukoencephalopathy-causing CLCN2 mutations are associated with impaired Cl - channel function and trafficking.
- Author
-
Gaitán-Peñas H, Apaja PM, Arnedo T, Castellanos A, Elorza-Vidal X, Soto D, Gasull X, Lukacs GL, and Estévez R
- Subjects
- Animals, CLC-2 Chloride Channels, Cell Membrane metabolism, Cells, Cultured, Chloride Channels genetics, Chlorides metabolism, HEK293 Cells, HeLa Cells, Humans, Neuroglia metabolism, Protein Stability, Protein Transport, Rats, Rats, Sprague-Dawley, Tight Junctions metabolism, Xenopus, Chloride Channels metabolism, Ion Channel Gating, Leukoencephalopathies genetics, Mutation
- Abstract
Key Points: Characterisation of most mutations found in CLCN2 in patients with CC2L leukodystrophy show that they cause a reduction in function of the chloride channel ClC-2. GlialCAM, a regulatory subunit of ClC-2 in glial cells and involved in the leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC), increases the activity of a ClC-2 mutant by affecting ClC-2 gating and by stabilising the mutant at the plasma membrane. The stabilisation of ClC-2 at the plasma membrane by GlialCAM depends on its localisation at cell-cell junctions. The membrane protein MLC1, which is defective in MLC, also contributes to the stabilisation of ClC-2 at the plasma membrane, providing further support for the view that GlialCAM, MLC1 and ClC-2 form a protein complex in glial cells., Abstract: Mutations in CLCN2 have been recently identified in patients suffering from a type of leukoencephalopathy involving intramyelinic oedema. Here, we characterised most of these mutations that reduce the function of the chloride channel ClC-2 and impair its plasma membrane (PM) expression. Detailed biochemical and electrophysiological analyses of the Ala500Val mutation revealed that defective gating and increased cellular and PM turnover contributed to defective A500V-ClC-2 functional expression. Co-expression of the adhesion molecule GlialCAM, which forms a tertiary complex with ClC-2 and megalencephalic leukoencephalopathy with subcortical cysts 1 (MLC1), rescued the functional expression of the mutant by modifying its gating properties. GlialCAM also restored the PM levels of the channel by impeding its turnover at the PM. This rescue required ClC-2 localisation to cell-cell junctions, since a GlialCAM mutant with compromised junctional localisation failed to rescue the impaired stability of mutant ClC-2 at the PM. Wild-type, but not mutant, ClC-2 was also stabilised by MLC1 overexpression. We suggest that leukodystrophy-causing CLCN2 mutations reduce the functional expression of ClC-2, which is partly counteracted by GlialCAM/MLC1-mediated increase in the gating and stability of the channel., (© 2017 The Authors. The Journal of Physiology © 2017 The Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
36. An L319F mutation in transmembrane region 3 (TM3) selectively reduces sensitivity to okaramine B of the Bombyx mori l-glutamate-gated chloride channel.
- Author
-
Furutani S, Okuhara D, Hashimoto A, Ihara M, Kai K, Hayashi H, Sattelle DB, and Matsuda K
- Subjects
- Amino Acid Sequence, Animals, Bombyx metabolism, Chloride Channels genetics, Dose-Response Relationship, Drug, Drug Interactions, Glutamic Acid pharmacology, Insect Proteins chemistry, Insect Proteins genetics, Insect Proteins metabolism, Ivermectin pharmacology, Models, Molecular, Protein Conformation, Sequence Alignment, Azetidines pharmacology, Azocines pharmacology, Bombyx drug effects, Bombyx genetics, Cell Membrane metabolism, Chloride Channels chemistry, Chloride Channels metabolism, Indole Alkaloids pharmacology, Mutation
- Abstract
Okaramines produced by Penicillium simplicissimum AK-40 activate l-glutamate-gated chloride channels (GluCls) and thus paralyze insects. However, the okaramine binding site on insect GluCls is poorly understood. Sequence alignment shows that the equivalent of residue Leucine319 of the okaramine B sensitive Bombyx mori (B. mori) GluCl is a phenylalanine in the okaramine B insensitive B. mori γ-aminobutyric acid-gated chloride channel of the same species. This residue is located in the third transmembrane (TM3) region, a location which in a nematode GluCl is close to the ivermectin binding site. The B. mori GluCl containing the L319F mutation retained its sensitivity to l-glutamate, but responses to ivermectin were reduced and those to okaramine B were completely blocked.
- Published
- 2017
- Full Text
- View/download PDF
37. Screening of BEST1 Gene in a Chinese Cohort With Best Vitelliform Macular Dystrophy or Autosomal Recessive Bestrophinopathy.
- Author
-
Tian L, Sun T, Xu K, Zhang X, Peng X, and Li Y
- Subjects
- Adult, Bestrophins, China, DNA Mutational Analysis, Eye Diseases, Hereditary diagnosis, Humans, Male, Mass Screening methods, Middle Aged, Phenotype, Polymerase Chain Reaction, Retinal Diseases diagnosis, Vitelliform Macular Dystrophy diagnosis, Asian People genetics, Chloride Channels genetics, Eye Diseases, Hereditary genetics, Eye Proteins genetics, Mutation, Retinal Diseases genetics, Vitelliform Macular Dystrophy genetics
- Abstract
Purpose: Mutations in the BEST1 gene can cause Best vitelliform macular dystrophy (BVMD) and autosomal recessive bestrophinopathy (ARB). The aim of the current study was to establish the BEST1 mutation spectrum in Chinese patients with BVMD and ARB and to describe the phenotypic characteristics of patients carrying BEST1 mutations., Methods: A total of 37 probands with a clinical diagnosis of BVMD (17 patients) or ARB (20 patients) were recruited for genetic analysis; of these, only 5 probands had a family history. All probands underwent detailed ophthalmic examinations. All coding exons and exon-intron boundaries of the BEST1 gene were screened by PCR-based DNA sequencing. In silico programs were used to analyze the pathogenicity of all the variants. Genomic DNA rearrangements of the BEST1 gene were identified by real-time quantitative PCR (RQ-PCR)., Results: For patients with BVMD, single heterozygous BEST1 mutations were identified in 13 patients and compound heterozygous mutations were found in 3 patients. For patients with ARB, biallelic mutations were found in 13 probands and single mutant alleles in six patients. Overall, 36 disease-causing variants (20 novel mutations) of the BEST1 gene were identified, including 28 (77.8%) missense, 3 (8.3%) nonsense, 4 (11.1%) splicing effect, and 1 (2.8%) frameshift small duplication mutations., Conclusions: The mutation spectrum of the BEST1 gene in Chinese patients differed from those of Caucasian patients. Mutations that cause ARB differ from those that cause BVMD. BEST1 screening is important for precise diagnosis of BVMD or ARB.
- Published
- 2017
- Full Text
- View/download PDF
38. Clinical exome sequencing in early-onset generalized dystonia and large-scale resequencing follow-up.
- Author
-
Zech M, Boesch S, Jochim A, Weber S, Meindl T, Schormair B, Wieland T, Lunetta C, Sansone V, Messner M, Mueller J, Ceballos-Baumann A, Strom TM, Colombo R, Poewe W, Haslinger B, and Winkelmann J
- Subjects
- Adenylyl Cyclases genetics, Adult, Anoctamins, Apoptosis Regulatory Proteins genetics, Chloride Channels genetics, DNA Mutational Analysis, DNA-Binding Proteins genetics, Family Health, Female, Follow-Up Studies, GTP Cyclohydrolase genetics, Humans, Male, Middle Aged, Models, Molecular, Molecular Chaperones genetics, Nuclear Proteins genetics, Young Adult, Dystonia genetics, Exome genetics, Mutation genetics
- Abstract
Background: Dystonia is clinically and genetically heterogeneous. Despite being a first-line testing tool for heterogeneous inherited disorders, whole-exome sequencing has not yet been evaluated in dystonia diagnostics. We set up a pilot study to address the yield of whole-exome sequencing for early-onset generalized dystonia, a disease subtype enriched for monogenic causation., Methods: Clinical whole-exome sequencing coupled with bioinformatics analysis and detailed phenotyping of mutation carriers was performed on 16 consecutive cases with genetically undefined early-onset generalized dystonia. Candidate pathogenic variants were validated and tested for cosegregation. The whole-exome approach was complemented by analyzing 2 mutated yet unestablished causative genes in another 590 dystonia cases., Results: Whole-exome sequencing detected clinically relevant mutations of known dystonia-related genes in 6 generalized dystonia cases (37.5%), among whom 3 had novel variants. Reflecting locus heterogeneity, identified unique variants were distributed over 5 genes (GCH1, THAP1, TOR1A, ANO3, ADCY5), of which only 1 (ANO3) was mutated recurrently. Three genes (GCH1, THAP1, TOR1A) were associated with isolated generalized dystonia, whereas 2 (ANO3, ADCY5) gave rise to combined dystonia-myoclonus phenotypes. Follow-up screening of ANO3 and ADCY5 revealed a set of distinct variants of interest, the pathogenicity of which was supported by bioinformatics testing and cosegregation work., Conclusions: Our study identified whole-exome sequencing as an effective strategy for molecular diagnosis of early-onset generalized dystonia and offers insights into the heterogeneous genetic architecture of this condition. Furthermore, it provides confirmatory evidence for a dystonia-relevant role of ANO3 and ADCY5, both of which likely associate with a broader spectrum of dystonic expressions than previously thought. © 2016 International Parkinson and Movement Disorder Society., (© 2016 International Parkinson and Movement Disorder Society.)
- Published
- 2017
- Full Text
- View/download PDF
39. Malignant Infantile Osteopetrosis.
- Author
-
Gillani S and Abbas Z
- Subjects
- Biopsy, Chloride Channels metabolism, DNA Mutational Analysis, Fatal Outcome, Humans, Infant, Newborn, Male, Osteopetrosis diagnosis, Radiography, Vacuolar Proton-Translocating ATPases metabolism, Bone Marrow pathology, Chloride Channels genetics, DNA, Neoplasm genetics, Mutation, Osteopetrosis genetics, Vacuolar Proton-Translocating ATPases genetics
- Abstract
Two main forms of osteopetrosis are recognized, a severe autosomal recessive form (MIM 259700) with an incidence of approximately 1 in 250,000 births and a mild autosomal dominant form (MIM166600) with an incidence of 1 in 20,000 births. Intrinsic disturbances of osteoclastic function due to mutations in genes encoding osteoclast-specific subunits of the vacuolar proton pump (TCIRG1, CLCN7) are found in most patients with recessive form. Mutations of CLCN7 are observed in dominant form of osteopetrosis .The recessive form of ostreopetrosis, i.e., malignant infantile osteopetrosis (MIOP) presents early in life with extreme sclerosis of the skeleton and reduction of marrow space. Signs/symptoms of MIOP appear as early as neonatal age. As there is defect in bone marrow children present with deficiency of red blood cells, white blood cells and platelets. There is extramedullary haemopoiesis, cranial nerve compressions and severe growth failure. The condition also presents with early and late onset neonatal sepsis and is often lethal in the first decade of life due to secondary infections. Treatment is mainly supportive. The only curative treatment is stem cell transplantation. This is a case report of a new-born who was admitted in nursery of Ayub Teaching Hospital initially with complains of neonatal jaundice and sepsis , and a second time with lower respiratory tract infection. Death was eventually due to sepsis. Workup led to diagnosis of Malignant infantile osteopetrosis.
- Published
- 2017
40. Digenic mutations involving both the BSND and GJB2 genes detected in Bartter syndrome type IV.
- Author
-
Wang HH, Feng Y, Li HB, Wu H, Mei LY, Wang XW, Jiang L, and He CF
- Subjects
- Bartter Syndrome complications, Child, Preschool, Connexin 26, Exons, Female, Homozygote, Humans, Infant, Infant, Newborn, Bartter Syndrome genetics, Chloride Channels genetics, Connexins genetics, Hearing Loss, Sensorineural genetics, Mutation
- Abstract
Bartter syndrome type IV, characterized by salt-losing nephropathies and sensorineural deafness, is caused by mutations of BSND or simultaneous mutations of both CLCNKA and CLCNKB. GJB2 is the primary causative gene for non-syndromic sensorineural deafness and associated with several syndromic sensorineural deafness. Owing to the rarity of Bartter syndrome, only a few mutations have been reported in the abovementioned causative genes. To investigate the underlying mutations in a Chinese patient with Bartter syndrome type IV, genetic analysis of BSND, CLCNKA, CLCNKB and GJB2 were performed by polymerase chain reaction and direct sequencing. Finally, double homozygous mutations c.22C > T (p.Arg8Trp) and c.127G > A (Val43Ile) were detected in exon 1 of BSND. Intriguingly, compound heterozygous mutations c.235delC (p.Leu79CysfsX3) and c.109G > A (p.Val37Ile) were also revealed in exon 2 of GJB2 in the same patient. No pathogenic mutations were found in CLCNKA and CLCNKB. Our results indicated that the homozygous mutation c.22C > T was the key genetic reason for the proband, and a digenic effect of BSND and GJB2 might contributed to sensorineural deafness. To our knowledge, it was the first report showing that the GJB2 gene mutations were detected in Bartter syndrome., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
41. Salt-losing nephropathy in mice with a null mutation of the Clcnk2 gene.
- Author
-
Grill A, Schießl IM, Gess B, Fremter K, Hammer A, and Castrop H
- Subjects
- Animals, Bartter Syndrome metabolism, Bartter Syndrome physiopathology, Chloride Channels metabolism, Dinoprostone metabolism, Glomerular Filtration Rate genetics, Kidney metabolism, Mice, Mice, Knockout, Phenotype, Bartter Syndrome genetics, Chloride Channels genetics, Kidney physiopathology, Mutation, Renin-Angiotensin System genetics
- Abstract
Aim: The basolateral chloride channel ClC-Kb facilitates Cl reabsorption in the distal nephron of the human kidney. Functional mutations in CLCNKB are associated with Bartter's syndrome type 3, a hereditary salt-losing nephropathy. To address the function of ClC-K2 in vivo, we generated ClC-K2-deficient mice., Methods: ClC-K2-deficient mice were generated using TALEN technology., Results: ClC-K2-deficient mice were viable and born in a Mendelian ratio. ClC-K2-/- mice showed no gross anatomical abnormalities, but they were growth retarded. The 24-h urine volume was increased in ClC-K2-/- mice (4.4 ± 0.6 compared with 0.9 ± 0.2 mL per 24 h in wild-type littermates; P = 0.001). Accordingly, ambient urine osmolarity was markedly reduced (590 ± 39 vs. 2216 ± 132 mosmol L
-1 in wild types; P < 0.0001). During water restriction (24 h), urinary osmolarity increased to 1633 ± 153 and 3769 ± 129 mosmol L-1 in ClC-K2-/- and wild-type mice (n = 12; P < 0.0001), accompanied by a loss of body weight of 12 ± 0.4 and 8 ± 0.2% respectively (P < 0.0001). ClC-K2-/- mice showed an increased renal sodium excretion and compromised salt conservation during a salt-restricted diet. The salt-losing phenotype of ClC-K2-/- mice was associated with a reduced plasma volume, hypotension, a slightly reduced glomerular filtration rate, an increased renal prostaglandin E2 generation and a massively stimulated renin-angiotensin system. Clckb-/- mice showed a reduced sensitivity to furosemide and were completely resistant to thiazides., Conclusion: Loss of ClC-K2 compromises TAL function and abolishes salt reabsorption in the distal convoluted tubule. Our data suggest that ClC-K2 is crucial for renal salt reabsorption and concentrating ability. ClC-K2-deficient mice in most aspects mimic patients with Bartter's syndrome type 3., (© 2016 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)- Published
- 2016
- Full Text
- View/download PDF
42. A case of non-dystrophic myotonia with concomitant mutations in the SCN4A and CLCN1 genes.
- Author
-
Kato H, Kokunai Y, Dalle C, Kubota T, Madokoro Y, Yuasa H, Uchida Y, Ikeda T, Mochizuki H, Nicole S, Fontaine B, Takahashi MP, and Mitake S
- Subjects
- Adult, DNA Mutational Analysis, Electromyography, Evoked Potentials, Motor genetics, Exercise Test, Humans, Male, Myotonia physiopathology, Chloride Channels genetics, Mutation genetics, Myotonia genetics, NAV1.4 Voltage-Gated Sodium Channel genetics
- Abstract
Non-dystrophic myotonias are caused by mutations of either the skeletal muscle chloride (CLCN1) or sodium channel (SCN4A) gene. They exhibit several distinct phenotypes, including myotonia congenita, paramyotonia congenita and sodium channel myotonia, and a genotype-phenotype correlation has been established. However, there are atypical cases that do not fit with the standard classification. We report a case of 27-year-old male who had non-dystrophic myotonia with periodic paralysis and two heterozygous mutations, E950K in CLCN1 and F1290L in SCN4A. His mother, who exhibited myotonia without paralytic attack, only harbored E950K, and no mutations were identified in his asymptomatic father. Therefore, the E950K mutation was presumed to be pathogenic, although it was reported as an extremely rare genetic variant. The proband experienced paralytic attacks that lasted for weeks and were less likely to be caused by CLCN1 mutation alone. Functional analysis of the F1290L mutant channel heterologously expressed in cultured cells revealed enhanced activation inducing membrane hyperexcitability. We therefore propose that the two mutations had additive effects on membrane excitability that resulted in more prominent myotonia in the proband. Our case stresses the value of performing genetic analysis of both CLCN1 and SCN4A genes for myotonic patients with an atypical phenotype., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. Multidisciplinary study of a new ClC-1 mutation causing myotonia congenita: a paradigm to understand and treat ion channelopathies.
- Author
-
Imbrici P, Altamura C, Camerino GM, Mangiatordi GF, Conte E, Maggi L, Brugnoni R, Musaraj K, Caloiero R, Alberga D, Marsano RM, Ricci G, Siciliano G, Nicolotti O, Mora M, Bernasconi P, Desaphy JF, Mantegazza R, and Camerino DC
- Subjects
- Humans, Ion Channel Gating genetics, Ion Channel Gating physiology, Muscle, Skeletal metabolism, Patch-Clamp Techniques methods, Phenotype, Channelopathies metabolism, Chloride Channels genetics, Chloride Channels metabolism, Electrophysiological Phenomena genetics, Mutation genetics, Myotonia Congenita metabolism
- Abstract
Myotonia congenita is an inherited disease that is characterized by impaired muscle relaxation after contraction caused by loss-of-function mutations in the skeletal muscle ClC-1 channel. We report a novel ClC-1 mutation, T335N, that is associated with a mild phenotype in 1 patient, located in the extracellular I-J loop. The purpose of this study was to provide a solid correlation between T335N dysfunction and clinical symptoms in the affected patient as well as to offer hints for drug development. Our multidisciplinary approach includes patch-clamp electrophysiology on T335N and ClC-1 wild-type channels expressed in tsA201 cells, Western blot and quantitative PCR analyses on muscle biopsies from patient and unaffected individuals, and molecular dynamics simulations using a homology model of the ClC-1 dimer. T335N channels display reduced chloride currents as a result of gating alterations rather than altered surface expression. Molecular dynamics simulations suggest that the I-J loop might be involved in conformational changes that occur at the dimer interface, thus affecting gating. Finally, the gene expression profile of T335N carrier showed a diverse expression of K
+ channel genes, compared with control individuals, as potentially contributing to the phenotype. This experimental paradigm satisfactorily explained myotonia in the patient. Furthermore, it could be relevant to the study and therapy of any channelopathy.-Imbrici, P., Altamura, C., Camerino, G. M., Mangiatordi, G. F., Conte, E., Maggi, L., Brugnoni, R., Musaraj, K., Caloiero, R., Alberga, D., Marsano, R. M., Ricci, G., Siciliano, G., Nicolotti, O., Mora, M., Bernasconi, P., Desaphy, J.-F., Mantegazza, R., Camerino, D. C. Multidisciplinary study of a new ClC-1 mutation causing myotonia congenita: a paradigm to understand and treat ion channelopathies., (© The Author(s).)- Published
- 2016
- Full Text
- View/download PDF
44. Myotonia congenita type Becker in Bulgaria: First genetically proven cases and mutation screening of two presumable endemic regions.
- Author
-
Tincheva S, Georgieva B, Todorov T, Savov A, Tsaneva S, Litvinenko I, Mitev V, and Todorova A
- Subjects
- Adult, Bulgaria epidemiology, Child, Child, Preschool, Endemic Diseases, Family, Female, Genetic Testing, Humans, Male, Myotonia Congenita epidemiology, Chloride Channels genetics, Mutation, Myotonia Congenita genetics
- Abstract
Myotonia congenita type Becker is an autosomal recessive nondystrophic skeletal muscle disorder, caused by mutations in the CLCN1 gene. The disease is characterized by muscle stiffness and an inability of the muscle to relax after voluntary contraction. Here we report the results from molecular genetic testing of 6 families, referred for sequencing of the CLCN1 gene. The disease causing mutations were detected in 5 of the cases, representing diverse type of nucleotide changes: nonsense (p.Arg894*), splice-site (c.1471+1G>A), missense (p.Val273Met; p.Tyr524Cys). Two additional changes were detected in an asymptomatic individual (c.2284+5C>T and p.Phe167Leu). Two of the detected mutations are interesting from population point of view. The novel missense mutation p.Tyr524Cys was found in a large Bulgarian family with affected individuals in both vertical and horizontal pedigree directions, all of them carrying the mutation in homozygous form. They populate a village located in the northwest part of the country. Endogamous marriages are very unusual for the Bulgarian population, supposing a high carrier frequency in this subpopulation. Screening of 154 residents of the corresponding region showed a significant carrier frequency for the p.Tyr524Cys mutation of about 0.65% (1/154). The second interesting region in the context of Myotonia congenita type Becker is the southwest part of the country, where we found a large family of Bulgarian Turkish origin. The disease causing missense mutation p.Val273Met was again present in homozygous state. Surprisingly, the genetic testing of newborns from southwest Bulgaria showed an even higher carrier status of about 2.6% (3/116), disproving our initial hypothesis of endogamous marriages (traditionally common in this subpopulation) being the cause of the disease in these patients. However the probability of consanguineous marriages being the cause for further exaggeration of the anyway very high carrier frequency cannot be excluded., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Unilateral BEST1-Associated Retinopathy.
- Author
-
Arora R, Khan K, Kasilian ML, Strauss RW, Holder GE, Robson AG, Thompson DA, Moore AT, and Michaelides M
- Subjects
- Adolescent, Adult, Bestrophins, Child, Electrooculography, Electroretinography, Exons genetics, Female, Fluorescein Angiography, Humans, Male, Polymerase Chain Reaction, Retrospective Studies, Tomography, Optical Coherence, Visual Acuity physiology, Vitelliform Macular Dystrophy diagnosis, Chloride Channels genetics, Eye Proteins genetics, Mutation, Vitelliform Macular Dystrophy genetics
- Abstract
Purpose: To describe a series of patients with molecularly confirmed mutation in BEST1 causing Best disease but with unilateral clinical manifestation., Design: Retrospective observational case series., Setting: Moorfields Eye Hospital and Great Ormond Street Hospital, London (United Kingdom)., Patients: Five patients (10 eyes) with uniocular manifestation of BEST1 mutation causing Best disease were ascertained retrospectively from the clinical and genetic databases., Main Outcome Measures: Patients had full ophthalmologic examination, color fundus photography, fundus autofluorescence imaging, spectral-domain optical coherence tomography, and detailed electrophysiological assessment. Genetic testing was performed., Results: All cases had a clinical appearance typical of and consistent with Best disease at various stages, except that the presentation was unilateral. The reduced electrooculogram light rise was bilateral and in the context of normal electroretinograms therefore indicates generalized dysfunction at the level of the retinal pigment epithelium., Conclusions: Mutation in BEST1 has variable penetrance and expressivity, and can be uniocular. The clinical and electrophysiological features described assist targeted mutational screening and alert to the potential diagnosis even when there is an atypical unilateral presentation., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
46. Dent Disease in Chinese Children and Findings from Heterozygous Mothers: Phenotypic Heterogeneity, Fetal Growth, and 10 Novel Mutations.
- Author
-
Li F, Yue Z, Xu T, Chen M, Zhong L, Liu T, Jing X, Deng J, Hu B, Liu Y, Wang H, Lai KN, Sun L, Liu J, Maxwell PH, and Wang Y
- Subjects
- Adolescent, Adult, Child, Child, Preschool, China, Female, Fetal Development genetics, Heterozygote, Humans, Male, Mothers, Phenotype, Asian People genetics, Chloride Channels genetics, Dent Disease diagnosis, Dent Disease genetics, Mutation genetics, Phosphoric Monoester Hydrolases genetics
- Abstract
Objective: To characterize the phenotypes of Dent disease in Chinese children and their heterozygous mothers and to establish genetic diagnoses., Study Design: Using a modified protocol, we screened 1288 individuals with proteinuria. A diagnosis of Dent disease was established in 19 boys from 16 families by the presence of loss of function/deleterious mutations in CLCN5 or OCRL1. We also analyzed 16 available patients' mothers and examined their pregnancy records., Results: We detected 14 loss of function/deleterious mutations of CLCN5 in 15 boys and 2 mutations of OCRL1 in 4 boys. Of the patients, 16 of 19 had been wrongly diagnosed with other diseases and 11 of 19 had incorrect or unnecessary treatment. None of the patients, but 6 of 14 mothers, had nephrocalcinosis or nephrolithiasis at diagnosis. Of the patients, 8 of 14 with Dent disease 1 were large for gestational age (>90th percentile); 8 of 15 (53.3%) had rickets. We also present predicted structural changes for 4 mutant proteins., Conclusions: Pediatric Dent disease often is misdiagnosed; genetic testing achieves a correct diagnosis. Nephrocalcinosis or nephrolithiasis may not be sensitive diagnostic criteria. We identified 10 novel mutations in CLCN5 and OCRL1. The possibility that altered CLCN5 function could affect fetal growth and a possible link between a high rate of rickets and low calcium intake are discussed., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. A pure chloride channel mutant of CLC-5 causes Dent's disease via insufficient V-ATPase activation.
- Author
-
Satoh N, Yamada H, Yamazaki O, Suzuki M, Nakamura M, Suzuki A, Ashida A, Yamamoto D, Kaku Y, Sekine T, Seki G, and Horita S
- Subjects
- Animals, Cell Line, Cell Membrane metabolism, Child, Dent Disease genetics, Endocytosis physiology, Female, HEK293 Cells, Homeostasis physiology, Humans, Ion Transport physiology, Kidney Tubules, Proximal metabolism, Male, Oocytes metabolism, Xenopus laevis metabolism, Chloride Channels genetics, Chloride Channels metabolism, Dent Disease metabolism, Mutation genetics, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
Dent's disease is characterized by defective endocytosis in renal proximal tubules (PTs) and caused by mutations in the 2Cl(-)/H(+) exchanger, CLC-5. However, the pathological role of endosomal acidification in endocytosis has recently come into question. To clarify the mechanism of pathogenesis for Dent's disease, we examined the effects of a novel gating glutamate mutation, E211Q, on CLC-5 functions and endosomal acidification. In Xenopus oocytes, wild-type (WT) CLC-5 showed outward-rectifying currents that were inhibited by extracellular acidosis, but E211Q and an artificial pure Cl(-) channel mutant, E211A, showed linear currents that were insensitive to extracellular acidosis. Moreover, depolarizing pulse trains induced a robust reduction in the surface pH of oocytes expressing WT CLC-5 but not E211Q or E211A, indicating that the E211Q mutant functions as a pure Cl(-) channel similar to E211A. In HEK293 cells, E211A and E211Q stimulated endosomal acidification and hypotonicity-inducible vacuolar-type H(+)-ATPase (V-ATPase) activation at the plasma membrane. However, the stimulatory effects of these mutants were reduced compared with WT CLC-5. Furthermore, gene silencing experiments confirmed the functional coupling between V-ATPase and CLC-5 at the plasma membrane of isolated mouse PTs. These results reveal for the first time that the conversion of CLC-5 from a 2Cl(-)/H(+) exchanger into a Cl(-) channel induces Dent's disease in humans. In addition, defective endosomal acidification as a result of insufficient V-ATPase activation may still be important in the pathogenesis of Dent's disease.
- Published
- 2016
- Full Text
- View/download PDF
48. Novel CLCN7 mutation identified in a Han Chinese family with autosomal dominant osteopetrosis-2.
- Author
-
Deng H, He D, Rong P, Xu H, Yuan L, Li L, Lu Q, and Guo Y
- Subjects
- Adolescent, Adult, Aged, Amino Acid Sequence, Chloride Channels chemistry, Conserved Sequence genetics, DNA Mutational Analysis, Exome genetics, Family, Female, Humans, Male, Middle Aged, Osteopetrosis diagnostic imaging, Pedigree, Asian People genetics, Chloride Channels genetics, Ethnicity genetics, Genes, Dominant, Mutation genetics, Osteopetrosis genetics
- Abstract
Osteopetrosis is a heritable bone condition featuring increased bone density due to defective osteoclastic bone resorption. Exome sequencing and Sanger sequencing were conducted in Han Chinese family members, some of whom had typical osteopetrosis, and a novel missense variant c.2350A>T (p.R784W) in the chloride channel 7 gene (CLCN7) was identified. This variant cosegregated with the disorder in the family but was not observed in 800 controls. The data indicate that exome sequencing is a powerful and effective molecular diagnostic tool for detecting mutations in osteopetrosis, which is a genetically and clinically heterogeneous disorder. This discovery broadens the CLCN7 gene mutation spectrum and has important implications for clinical therapeutic regimen decisions, prognosis evaluations, and antenatal diagnoses., (© The Author(s) 2016.)
- Published
- 2016
- Full Text
- View/download PDF
49. Focal seizures in a patient with myotonic disorder type 2 co-segregating with a chloride voltage-gated channel 1 gene mutation: a case report.
- Author
-
Peddareddygari LR, Grewal AS, and Grewal RP
- Subjects
- Humans, Male, Middle Aged, Myotonic Dystrophy genetics, Seizures genetics, Chloride Channels genetics, Mutation genetics, Myotonic Dystrophy complications, Seizures complications
- Abstract
Background: Myotonic dystrophy type 2 is a multisystem disorder with both neurological and non-neurological signs and symptoms. Seizures are not a commonly associated neurological feature of this disorder., Case Presentation: A 57-year-old white American man presented with a long history of clinical and electrophysiological features of a myotonic disorder. He also developed multiple episodes of focal seizures and underwent a series of investigations which showed no structural or metabolic etiology. Genetic testing revealed that he had an expansion mutation in CCHC-type zinc finger, nucleic acid binding protein gene confirming the diagnosis of myotonic disorder type 2 and carried a mutation in the chloride voltage-gated channel 1 gene., Conclusions: We report a rare association between myotonic dystrophy type 2 and a seizure disorder. The pathophysiology of a possible relationship between these two neurological conditions is discussed.
- Published
- 2016
- Full Text
- View/download PDF
50. Mutation Analysis of MR-1, SLC2A1, and CLCN1 in 28 PRRT2-negative Paroxysmal Kinesigenic Dyskinesia Patients.
- Author
-
Wang HX, Li HF, Liu GL, Wen XD, and Wu ZY
- Subjects
- Adolescent, Adult, Child, Chorea genetics, Dystonia diagnosis, Female, Humans, Male, Myotonia Congenita genetics, Chloride Channels genetics, Dystonia genetics, Glucose Transporter Type 1 genetics, Membrane Proteins genetics, Muscle Proteins genetics, Mutation, Nerve Tissue Proteins genetics
- Abstract
Background: Paroxysmal kinesigenic dyskinesia (PKD) is the most common subtype of paroxysmal dyskinesias and is caused by mutations in PRRT2 gene. The majority of familial PKD was identified to harbor PRRT2 mutations. However, over two-third of sporadic PKD patients did not carry anyPRRT2 mutation, suggesting an existence of additional genetic mutations or possible misdiagnosis due to clinical overlap., Methods: A cohort of 28 Chinese patients clinically diagnosed with sporadic PKD and excluded PRRT2 mutations were recruited. Clinical features were evaluated, and all subjects were screened for MR-1, SLC2A1, and CLCN1 genes, which are the causative genes of paroxysmal nonkinesigenic dyskinesia (PNKD), paroxysmal exertion-induced dyskinesia, and myotonia congenita (MC), respectively. In addition, 200 genetically matched healthy individuals were recruited as controls., Results: A total of 16 genetic variants including 4 in MR-1 gene, 8 in SLC2A1 gene, and 4 in CLCN1 gene were detected. Among them, SLC2A1 c.363G>A mutation was detected in one case, and CLCN1 c.1205C>T mutation was detected in other two cases. Neither of them was found in 200 controls as well as 1000 Genomes database and ExAC database. Both mutations were predicted to be pathogenic by SIFT and PolyPhen2. The SLC2A1 c.363G>A mutation was novel., Conclusions: The phenotypic overlap may lead to the difficulty in distinguishing PKD from PNKD and MC. For those PRRT2- negative PKD cases, screening of SLC2A1 and CLCN1 genes are useful in confirming the diagnosis.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.