1
|
Silveira KC, Ambrose A, Athey T, Taylor S, Mercimek-Andrews S, Kannu P. Dissecting CASK: Novel splice site variant associated with male MICPCH phenotype. Clin Genet 2024; 106:764-768. [PMID: 39212003 DOI: 10.1111/cge.14610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
CASK (MIM#300172), encoding a calcium/calmodulin-dependent serine protein kinase, is crucial for synaptic transmission and gene regulation during neural development. Pathogenic variants of CASK are known to cause several neurodevelopmental disorders, including X-linked intellectual disability and microcephaly with pontine and cerebellar hypoplasia (MICPCH). This study introduces a novel, de novo synonymous CASK variant (NM_001367721.1: c.1737G>A, p.(Glu579=)), discovered in a male patient diagnosed with MICPCH, characterized by microcephaly, developmental delay, visual impairment, and myoclonic seizures. The variant disrupts a donor splice-site at the end of exon 18. Transcriptomic analysis of blood identified 12 different CASK transcripts secondary to the synonymous variant. Nearly one third of these transcripts were predicted to result in nonsense mediated decay or protein degradation. Protein modeling revealed structural alterations in the PDZ functional domain of CASK, due to exon 18 deletion. Our findings highlight the utility of transcriptomic analysis in demonstrating the underlying disease mechanism in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Karina C Silveira
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anastasia Ambrose
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Taryn Athey
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Alberta Health Services, Edmonton Zone, Alberta, Canada
| | - Sherryl Taylor
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Saadet Mercimek-Andrews
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Alberta Health Services, Edmonton Zone, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Peter Kannu
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Alberta Health Services, Edmonton Zone, Alberta, Canada
| |
Collapse
|
2
|
Yahia A, Li D, Lejerkrans S, Rajagopalan S, Kalnak N, Tammimies K. Whole exome sequencing and polygenic assessment of a Swedish cohort with severe developmental language disorder. Hum Genet 2024; 143:169-183. [PMID: 38300321 PMCID: PMC10881898 DOI: 10.1007/s00439-023-02636-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/25/2023] [Indexed: 02/02/2024]
Abstract
Developmental language disorder (DLD) overlaps clinically, genetically, and pathologically with other neurodevelopmental disorders (NDD), corroborating the concept of the NDD continuum. There is a lack of studies to understand the whole genetic spectrum in individuals with DLD. Previously, we recruited 61 probands with severe DLD from 59 families and examined 59 of them and their families using microarray genotyping with a 6.8% diagnostic yield. Herein, we investigated 53 of those probands using whole exome sequencing (WES). Additionally, we used polygenic risk scores (PRS) to understand the within family enrichment of neurodevelopmental difficulties and examine the associations between the results of language-related tests in the probands and language-related PRS. We identified clinically significant variants in four probands, resulting in a 7.5% (4/53) molecular diagnostic yield. Those variants were in PAK2, MED13, PLCB4, and TNRC6B. We also prioritized additional variants for future studies for their role in DLD, including high-impact variants in PARD3 and DIP2C. PRS did not explain the aggregation of neurodevelopmental difficulties in these families. We did not detect significant associations between the language-related tests and language-related PRS. Our results support using WES as the first-tier genetic test for DLD as it can identify monogenic DLD forms. Large-scale sequencing studies for DLD are needed to identify new genes and investigate the polygenic contribution to the condition.
Collapse
Affiliation(s)
- Ashraf Yahia
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Danyang Li
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | - Sanna Lejerkrans
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Shyam Rajagopalan
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru, India
| | - Nelli Kalnak
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden
- Department of Speech-Language Pathology, Helsingborg Hospital, Helsingborg, Sweden
| | - Kristiina Tammimies
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Region Stockholm, Stockholm, Sweden.
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden.
| |
Collapse
|
3
|
Weeratunga S, Gormal RS, Liu M, Eldershaw D, Livingstone EK, Malapaka A, Wallis TP, Bademosi AT, Jiang A, Healy MD, Meunier FA, Collins BM. Interrogation and validation of the interactome of neuronal Munc18-interacting Mint proteins with AlphaFold2. J Biol Chem 2024; 300:105541. [PMID: 38072052 PMCID: PMC10820826 DOI: 10.1016/j.jbc.2023.105541] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024] Open
Abstract
Munc18-interacting proteins (Mints) are multidomain adaptors that regulate neuronal membrane trafficking, signaling, and neurotransmission. Mint1 and Mint2 are highly expressed in the brain with overlapping roles in the regulation of synaptic vesicle fusion required for neurotransmitter release by interacting with the essential synaptic protein Munc18-1. Here, we have used AlphaFold2 to identify and then validate the mechanisms that underpin both the specific interactions of neuronal Mint proteins with Munc18-1 as well as their wider interactome. We found that a short acidic α-helical motif within Mint1 and Mint2 is necessary and sufficient for specific binding to Munc18-1 and binds a conserved surface on Munc18-1 domain3b. In Munc18-1/2 double knockout neurosecretory cells, mutation of the Mint-binding site reduces the ability of Munc18-1 to rescue exocytosis, and although Munc18-1 can interact with Mint and Sx1a (Syntaxin1a) proteins simultaneously in vitro, we find that they have mutually reduced affinities, suggesting an allosteric coupling between the proteins. Using AlphaFold2 to then examine the entire cellular network of putative Mint interactors provides a structural model for their assembly with a variety of known and novel regulatory and cargo proteins including ADP-ribosylation factor (ARF3/ARF4) small GTPases and the AP3 clathrin adaptor complex. Validation of Mint1 interaction with a new predicted binder TJAP1 (tight junction-associated protein 1) provides experimental support that AlphaFold2 can correctly predict interactions across such large-scale datasets. Overall, our data provide insights into the diversity of interactions mediated by the Mint family and show that Mints may help facilitate a key trigger point in SNARE (soluble N-ethylmaleimide-sensitive factor attachment receptor) complex assembly and vesicle fusion.
Collapse
Affiliation(s)
- Saroja Weeratunga
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Meihan Liu
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Denaye Eldershaw
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Emma K Livingstone
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Anusha Malapaka
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Adekunle T Bademosi
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Anmin Jiang
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Michael D Healy
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Frederic A Meunier
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia.
| |
Collapse
|
4
|
Tello JA, Jiang L, Zohar Y, Restifo LL. Drosophila CASK regulates brain size and neuronal morphogenesis, providing a genetic model of postnatal microcephaly suitable for drug discovery. Neural Dev 2023; 18:6. [PMID: 37805506 PMCID: PMC10559581 DOI: 10.1186/s13064-023-00174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/08/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND CASK-related neurodevelopmental disorders are untreatable. Affected children show variable severity, with microcephaly, intellectual disability (ID), and short stature as common features. X-linked human CASK shows dosage sensitivity with haploinsufficiency in females. CASK protein has multiple domains, binding partners, and proposed functions at synapses and in the nucleus. Human and Drosophila CASK show high amino-acid-sequence similarity in all functional domains. Flies homozygous for a hypomorphic CASK mutation (∆18) have motor and cognitive deficits. A Drosophila genetic model of CASK-related disorders could have great scientific and translational value. METHODS We assessed the effects of CASK loss of function on morphological phenotypes in Drosophila using established genetic, histological, and primary neuronal culture approaches. NeuronMetrics software was used to quantify neurite-arbor morphology. Standard nonparametric statistics methods were supplemented by linear mixed effects modeling in some cases. Microfluidic devices of varied dimensions were fabricated and numerous fluid-flow parameters were used to induce oscillatory stress fields on CNS tissue. Dissociation into viable neurons and neurite outgrowth in vitro were assessed. RESULTS We demonstrated that ∆18 homozygous flies have small brains, small heads, and short bodies. When neurons from developing CASK-mutant CNS were cultured in vitro, they grew small neurite arbors with a distinctive, quantifiable "bushy" morphology that was significantly rescued by transgenic CASK+. As in humans, the bushy phenotype showed dosage-sensitive severity. To overcome the limitations of manual tissue trituration for neuronal culture, we optimized the design and operation of a microfluidic system for standardized, automated dissociation of CNS tissue into individual viable neurons. Neurons from CASK-mutant CNS dissociated in the microfluidic system recapitulate the bushy morphology. Moreover, for any given genotype, device-dissociated neurons grew larger arbors than did manually dissociated neurons. This automated dissociation method is also effective for rodent CNS. CONCLUSIONS These biological and engineering advances set the stage for drug discovery using the Drosophila model of CASK-related disorders. The bushy phenotype provides a cell-based assay for compound screening. Nearly a dozen genes encoding CASK-binding proteins or transcriptional targets also have brain-development mutant phenotypes, including ID. Hence, drugs that improve CASK phenotypes might also benefit children with disorders due to mutant CASK partners.
Collapse
Affiliation(s)
- Judith A Tello
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
- Department of Neurology, University of Arizona Health Sciences, 1501 N. Campbell Ave, Tucson, AZ, 85724-5023, USA
- Present address: Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, 10010, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85721, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85721, USA
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, 85721, USA
- BIO5 Interdisciplinary Research Institute, University of Arizona, Tucson, AZ, 85721, USA
| | - Linda L Restifo
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Neurology, University of Arizona Health Sciences, 1501 N. Campbell Ave, Tucson, AZ, 85724-5023, USA.
- BIO5 Interdisciplinary Research Institute, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Cellular & Molecular Medicine, University of Arizona Health Sciences, Tucson, AZ, 85724, USA.
| |
Collapse
|
5
|
Schiano C, Luongo L, Maione S, Napoli C. Mediator complex in neurological disease. Life Sci 2023; 329:121986. [PMID: 37516429 DOI: 10.1016/j.lfs.2023.121986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Neurological diseases, including traumatic brain injuries, stroke (haemorrhagic and ischemic), and inherent neurodegenerative diseases cause acquired disability in humans, representing a leading cause of death worldwide. The Mediator complex (MED) is a large, evolutionarily conserved multiprotein that facilities the interaction between transcription factors and RNA Polymerase II in eukaryotes. Some MED subunits have been found altered in the brain, although their specific functions in neurodegenerative diseases are not fully understood. Mutations in MED subunits were associated with a wide range of genetic diseases for MED12, MED13, MED13L, MED20, MED23, MED25, and CDK8 genes. In addition, MED12 and MED23 were deregulated in the Alzheimer's Disease. Interestingly, most of the genomic mutations have been found in the subunits of the kinase module. To date, there is only one evidence on MED1 involvement in post-stroke cognitive deficits. Although the underlying neurodegenerative disorders may be different, we are confident that the signal cascades of the biological-cognitive mechanisms of brain adaptation, which begin after brain deterioration, may also differ. Here, we analysed relevant studies in English published up to June 2023. They were identified through a search of electronic databases including PubMed, Medline, EMBASE and Scopus, including search terms such as "Mediator complex", "neurological disease", "brains". Thematic content analysis was conducted to collect and summarize all studies demonstrating MED alteration to understand the role of this central transcriptional regulatory complex in the brain. Improved and deeper knowledge of the regulatory mechanisms in neurological diseases can increase the ability of physicians to predict onset and progression, thereby improving diagnostic care and providing appropriate treatment decisions.
Collapse
Affiliation(s)
- Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Italy.
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Italy; IRCSS, Neuromed, Pozzilli, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Italy; IRCSS, Neuromed, Pozzilli, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Italy; Clinical Department of Internal Medicine and Specialistic Units, Division of Clinical Immunology and Immunohematology, Transfusion Medicine, and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Universitaria Policlinico (AOU), Italy
| |
Collapse
|
6
|
Mori T, Zhou M, Tabuchi K. Diverse Clinical Phenotypes of CASK-Related Disorders and Multiple Functional Domains of CASK Protein. Genes (Basel) 2023; 14:1656. [PMID: 37628707 PMCID: PMC10454856 DOI: 10.3390/genes14081656] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
CASK-related disorders are a form of rare X-linked neurological diseases and most of the patients are females. They are characterized by several symptoms, including microcephaly with pontine and cerebellar hypoplasia (MICPCH), epilepsy, congenital nystagmus, and neurodevelopmental disorders. Whole-genome sequencing has identified various mutations, including nonsense and missense mutations, from patients with CASK-related disorders, revealing correlations between specific mutations and clinical phenotypes. Notably, missense mutations associated with epilepsy and intellectual disability were found throughout the whole region of the CASK protein, while missense mutations related to microcephaly and MICPCH were restricted in certain domains. To investigate the pathophysiology of CASK-related disorders, research groups have employed diverse methods, including the generation of CASK knockout mice and the supplementation of CASK to rescue the phenotypes. These approaches have yielded valuable insights into the identification of functional domains of the CASK protein associated with a specific phenotype. Additionally, recent advancements in the AI-based prediction of protein structure, such as AlphaFold2, and the application of genome-editing techniques to generate CASK mutant mice carrying missense mutations from patients with CASK-related disorders, allow us to understand the pathophysiology of CASK-related disorders in more depth and to develop novel therapeutic methods for the fundamental treatment of CASK-related disorders.
Collapse
Affiliation(s)
- Takuma Mori
- Department of Neuroinnovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan;
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Mengyun Zhou
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Katsuhiko Tabuchi
- Department of Neuroinnovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan;
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| |
Collapse
|
7
|
Guo Q, Kouyama-Suzuki E, Shirai Y, Cao X, Yanagawa T, Mori T, Tabuchi K. Structural Analysis Implicates CASK-Liprin-α2 Interaction in Cerebellar Granular Cell Death in MICPCH Syndrome. Cells 2023; 12:cells12081177. [PMID: 37190086 DOI: 10.3390/cells12081177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Microcephaly with pontine and cerebellar hypoplasia (MICPCH) syndrome is a neurodevelopmental disorder caused by the deficiency of the X-chromosomal gene CASK. However, the molecular mechanisms by which CASK deficiency causes cerebellar hypoplasia in this syndrome remain elusive. In this study, we used CASK knockout (KO) mice as models for MICPCH syndrome and investigated the effect of CASK mutants. Female CASK heterozygote KO mice replicate the progressive cerebellar hypoplasia observed in MICPCH syndrome. CASK KO cultured cerebellar granule (CG) cells show progressive cell death that can be rescued by co-infection with lentivirus expressing wild-type CASK. Rescue experiments with CASK deletion mutants identify that the CaMK, PDZ, and SH3, but not L27 and guanylate kinase domains of CASK are required for the survival of CG cells. We identify missense mutations in the CaMK domain of CASK derived from human patients that fail to rescue the cell death of cultured CASK KO CG cells. Machine learning-based structural analysis using AlphaFold 2.2 predicts that these mutations disrupt the structure of the binding interface with Liprin-α2. These results suggest that the interaction with Liprin-α2 via the CaMK domain of CASK may be involved in the pathophysiology of cerebellar hypoplasia in MICPCH syndrome.
Collapse
Affiliation(s)
- Qi Guo
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Emi Kouyama-Suzuki
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Yoshinori Shirai
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, China
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
- Department of NeuroHealth Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
- Department of NeuroHealth Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan
| |
Collapse
|
8
|
Rodríguez-García ME, Cotrina-Vinagre FJ, Sánchez-Calvin MT, de Aragón AM, de Las Heras RS, Dinman JD, de Vries BBA, Nabais Sá MJ, Quijada-Fraile P, Martínez-Azorín F. A novel de novo variant in CASK causes a severe neurodevelopmental disorder that masks the phenotype of a novel de novo variant in EEF2. J Hum Genet 2023:10.1038/s10038-023-01150-4. [PMID: 37072624 DOI: 10.1038/s10038-023-01150-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/10/2023] [Accepted: 04/01/2023] [Indexed: 04/20/2023]
Abstract
We report a 9-year-old Spanish boy with severe psychomotor developmental delay, short stature, microcephaly and abnormalities of the brain morphology, including cerebellar atrophy. Whole-exome sequencing (WES) uncovered two novel de novo variants, a hemizygous variant in CASK (Calcium/Calmodulin Dependent Serine Protein Kinase) and a heterozygous variant in EEF2 (Eukaryotic Translation Elongation Factor 2). CASK gene encodes the peripheral plasma membrane protein CASK that is a scaffold protein located at the synapses in the brain. The c.2506-6 A > G CASK variant induced two alternative splicing events that account for the 80% of the total transcripts, which are likely to be degraded by NMD. Pathogenic variants in CASK have been associated with severe neurological disorders such as mental retardation with or without nystagmus also called FG syndrome 4 (FGS4), and intellectual developmental disorder with microcephaly and pontine and cerebellar hypoplasia (MICPCH). Heterozygous variants in EEF2, which encodes the elongation factor 2 (eEF2), have been associated to Spinocerebellar ataxia 26 (SCA26) and more recently to a childhood-onset neurodevelopmental disorder with benign external hydrocephalus. The yeast model system used to investigate the functional consequences of the c.34 A > G EEF2 variant supported its pathogenicity by demonstrating it affects translational fidelity. In conclusion, the phenotype associated with the CASK variant is more severe and masks the milder phenotype of EEF2 variant.
Collapse
Affiliation(s)
- María Elena Rodríguez-García
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN). Instituto de Investigación Hospital 12 de Octubre (i + 12), E-28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, E-28041, Madrid, Spain
| | - Francisco Javier Cotrina-Vinagre
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN). Instituto de Investigación Hospital 12 de Octubre (i + 12), E-28041, Madrid, Spain
| | | | | | | | - Jonathan D Dinman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA
| | - Bert B A de Vries
- Department of Human Genetics, Radboud University Medical Center and Donders Institute for Brain, Cognition and Behavior, 6525 GA, Nijmegen, The Netherlands
| | - Maria João Nabais Sá
- Centre for Predictive and Preventive Genetics (CGPP) and UnIGENe, Institute for Molecular and Cell Biology (IBMC), i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Pilar Quijada-Fraile
- Unidad Pediátrica de Enfermedades Raras, Enfermedades Mitocondriales y Metabólicas Hereditarias, Hospital 12 de Octubre, E-28041, Madrid, Spain
| | - Francisco Martínez-Azorín
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN). Instituto de Investigación Hospital 12 de Octubre (i + 12), E-28041, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, E-28041, Madrid, Spain.
| |
Collapse
|
9
|
Yeo XY, Lim YT, Chae WR, Park C, Park H, Jung S. Alterations of presynaptic proteins in autism spectrum disorder. Front Mol Neurosci 2022; 15:1062878. [PMID: 36466804 PMCID: PMC9715400 DOI: 10.3389/fnmol.2022.1062878] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/31/2022] [Indexed: 01/05/2025] Open
Abstract
The expanded use of hypothesis-free gene analysis methods in autism research has significantly increased the number of genetic risk factors associated with the pathogenesis of autism. A further examination of the implicated genes directly revealed the involvement in processes pertinent to neuronal differentiation, development, and function, with a predominant contribution from the regulators of synaptic function. Despite the importance of presynaptic function in synaptic transmission, the regulation of neuronal network activity, and the final behavioral output, there is a relative lack of understanding of the presynaptic contribution to the pathology of autism. Here, we will review the close association among autism-related mutations, autism spectrum disorders (ASD) phenotypes, and the altered presynaptic protein functions through a systematic examination of the presynaptic risk genes relating to the critical stages of synaptogenesis and neurotransmission.
Collapse
Affiliation(s)
- Xin Yi Yeo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi Tang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Woo Ri Chae
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of BioNano Technology, Gachon University, Seongnam, South Korea
| | - Chungwon Park
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Hyokeun Park
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Sangyong Jung
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Xie G, Zhang Y, Yang W, Yang L, Wang R, Xu M, Sun L, Zhang B, Cui X. Case report: A novel CASK mutation in a Chinese female child with microcephaly with pontine and cerebellar hypoplasia. Front Genet 2022; 13:856636. [PMID: 36159992 PMCID: PMC9490368 DOI: 10.3389/fgene.2022.856636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Microcephaly with pontine and cerebellar hypoplasia (MICPCH) is a rare X-linked dominant genetic disease, and most MICPCHs are ascribed to CASK mutations, while few are revealed in Chinese patients. This study aims to identify the pathogenic mutation in a Chinese proband with MICPCH. Methods: A 3-year-old female Chinese proband with MICPCH and her parents were included. Clinical data were collected from the medical records and recalled by the proband's mother. Whole genome sequencing and Sanger sequencing were used to find the pathogenic mutation of MICPCH. Results: The proband presented with postnatal progressive microcephaly, cerebellar hypoplasia, intellectual disability, motor and language development retardation and limb hypertonia. Genetic analysis indicated that there was a novel compound heterozygote nonsynonymous mutation, c.755T>C(p.Leu252Pro) in exon8 of CASK gene in the proband, but not in her parents. This CASK mutation has not been reported in other databases. Conclusion: This study broadens the mutation spectrum of the CASK gene and is of great value for precise prenatal diagnosis and genetic counseling.
Collapse
Affiliation(s)
- Guilan Xie
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenfang Yang
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Liren Yang
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Ruiqi Wang
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Mengmeng Xu
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Landi Sun
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Boxing Zhang
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaoyi Cui
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Peking University Health Science Center, Beijing, China
| |
Collapse
|
11
|
Dubbs H, Ortiz-Gonzalez X, Marsh ED. Pathogenic variants in CASK: Expanding the genotype-phenotype correlations. Am J Med Genet A 2022; 188:2617-2626. [PMID: 35670295 DOI: 10.1002/ajmg.a.62863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/04/2022] [Accepted: 05/16/2022] [Indexed: 01/24/2023]
Abstract
Pathogenic variants in CASK, an X-linked gene that plays a role in brain development and synaptic function, are the cause of both microcephaly with pontine and cerebellar hypoplasia (MICPCH), and X-linked intellectual disability (XLID) with or without nystagmus. MICPCH is caused by loss of function variants in CASK, typically affects females, and is associated with moderate-to-severe intellectual disability (ID). Additional findings, present in about one-third of individuals, include feeding difficulties, ophthalmologic issues, hypertonicity, epilepsy, and sensorineural hearing loss. Only a few affected males with MICPCH phenotype have been reported and most have had profound developmental disability and intractable epilepsy. The XLID phenotype is typically caused by missense variants and most often manifests in males; carrier females are mildly affected or unaffected. Nystagmus is often present. In total, over 175 patients have been reported in the literature. We now report an additional 11 patients with pathogenic variants in CASK that expand these phenotypes and reported genotype-phenotype correlations.
Collapse
Affiliation(s)
- Holly Dubbs
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xilma Ortiz-Gonzalez
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric D Marsh
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Lai D, Gade M, Yang E, Koh HY, Lu J, Walley NM, Buckley AF, Sands TT, Akman CI, Mikati MA, McKhann GM, Goldman JE, Canoll P, Alexander AL, Park KL, Von Allmen GK, Rodziyevska O, Bhattacharjee MB, Lidov HGW, Vogel H, Grant GA, Porter BE, Poduri AH, Crino PB, Heinzen EL. Somatic variants in diverse genes leads to a spectrum of focal cortical malformations. Brain 2022; 145:2704-2720. [PMID: 35441233 PMCID: PMC9612793 DOI: 10.1093/brain/awac117] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/19/2022] [Accepted: 03/13/2022] [Indexed: 11/14/2022] Open
Abstract
Post-zygotically acquired genetic variants, or somatic variants, that arise during cortical development have emerged as important causes of focal epilepsies, particularly those due to malformations of cortical development. Pathogenic somatic variants have been identified in many genes within the PI3K-AKT-mTOR-signalling pathway in individuals with hemimegalencephaly and focal cortical dysplasia (type II), and more recently in SLC35A2 in individuals with focal cortical dysplasia (type I) or non-dysplastic epileptic cortex. Given the expanding role of somatic variants across different brain malformations, we sought to delineate the landscape of somatic variants in a large cohort of patients who underwent epilepsy surgery with hemimegalencephaly or focal cortical dysplasia. We evaluated samples from 123 children with hemimegalencephaly (n = 16), focal cortical dysplasia type I and related phenotypes (n = 48), focal cortical dysplasia type II (n = 44), or focal cortical dysplasia type III (n = 15). We performed high-depth exome sequencing in brain tissue-derived DNA from each case and identified somatic single nucleotide, indel and large copy number variants. In 75% of individuals with hemimegalencephaly and 29% with focal cortical dysplasia type II, we identified pathogenic variants in PI3K-AKT-mTOR pathway genes. Four of 48 cases with focal cortical dysplasia type I (8%) had a likely pathogenic variant in SLC35A2. While no other gene had multiple disease-causing somatic variants across the focal cortical dysplasia type I cohort, four individuals in this group had a single pathogenic or likely pathogenic somatic variant in CASK, KRAS, NF1 and NIPBL, genes previously associated with neurodevelopmental disorders. No rare pathogenic or likely pathogenic somatic variants in any neurological disease genes like those identified in the focal cortical dysplasia type I cohort were found in 63 neurologically normal controls (P = 0.017), suggesting a role for these novel variants. We also identified a somatic loss-of-function variant in the known epilepsy gene, PCDH19, present in a small number of alleles in the dysplastic tissue from a female patient with focal cortical dysplasia IIIa with hippocampal sclerosis. In contrast to focal cortical dysplasia type II, neither focal cortical dysplasia type I nor III had somatic variants in genes that converge on a unifying biological pathway, suggesting greater genetic heterogeneity compared to type II. Importantly, we demonstrate that focal cortical dysplasia types I, II and III are associated with somatic gene variants across a broad range of genes, many associated with epilepsy in clinical syndromes caused by germline variants, as well as including some not previously associated with radiographically evident cortical brain malformations.
Collapse
Affiliation(s)
- Dulcie Lai
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Meethila Gade
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hyun Yong Koh
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.,Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jinfeng Lu
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nicole M Walley
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anne F Buckley
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Tristan T Sands
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Cigdem I Akman
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Mohamad A Mikati
- Department of Neurobiology, Duke University, Durham, NC 27708, USA.,Division of Pediatric Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guy M McKhann
- Department of Neurosurgery, Columbia University, New York Presbyterian Hospital, New York, NY 10032, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Allyson L Alexander
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kristen L Park
- Department of Pediatrics and Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Gretchen K Von Allmen
- Department of Neurology, McGovern Medical School, Houston, TX 77030, USA.,Division of Child Neurology, Department of Pediatrics, McGovern Medical School, Houston, TX 77030, USA
| | - Olga Rodziyevska
- Division of Child Neurology, Department of Pediatrics, McGovern Medical School, Houston, TX 77030, USA
| | | | - Hart G W Lidov
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Gerald A Grant
- Department of Neurosurgery, Lucile Packard Children's Hospital at Stanford, School of Medicine, Stanford, CA 94305, USA
| | - Brenda E Porter
- Department of Neurology and Neurological Sciences, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Annapurna H Poduri
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.,Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erin L Heinzen
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
13
|
Zhang R, Jia P, Yao Y, Zhu F. Case Report: Identification of a novel CASK missense variant in a Chinese family with MICPCH. Front Genet 2022; 13:933785. [PMID: 36092876 PMCID: PMC9452731 DOI: 10.3389/fgene.2022.933785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Mental retardation and microcephaly with pontine and cerebellar hypoplasia (MICPCH) is a rare genetic disorder that results in varying levels of pontocerebellar hypoplasia, microcephaly, and severe intellectual disabilities. Prior genetic analyses have identified the CASK gene as a driver of MICPCH. Herein, we analyzed a Chinese family with MICPCH. The index patient was an 8-year-old male. He and his 3-year-old brother suffered from microcephaly, pontocerebellar hypoplasia, serious mental retardation, ataxia, gait disorder, and inability to speak. Through a combination of whole-exome sequencing and subsequent Sanger sequencing, a novel X-linked missense mutation, c.1882G>C (p.D628H) in the CASK gene, was identified in two siblings, as well as their mother and grandmother, who exhibited mild mental retardation. Other family members with negative genetic testing were normal. In silico analyses indicated that this missense mutation was predicted to reduce CASK protein stability, disrupt the SRC homology 3 (SH3) domain, and abolish its function. In summary, we identified a novel missense variate in CASK associated with MICPCH. Our work facilitates the diagnosis of the disease in this family and broadens the gene variant spectrum of the CASK in MICPCH patients.
Collapse
Affiliation(s)
- Runfeng Zhang
- College of Life Sciences, Hubei Normal University, Huangshi, China
| | - Peng Jia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanyi Yao
- Medical Genetic Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
- *Correspondence: Feng Zhu, ; Yanyi Yao,
| | - Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Feng Zhu, ; Yanyi Yao,
| |
Collapse
|
14
|
Zhang Y, Nie Y, Mu Y, Zheng J, Xu X, Zhang F, Shu J, Liu Y. A de novo variant in CASK gene causing intellectual disability and brain hypoplasia: a case report and literature review. Ital J Pediatr 2022; 48:73. [PMID: 35550617 PMCID: PMC9097383 DOI: 10.1186/s13052-022-01248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Background The pathogenic variation of CASK gene can cause CASK related mental disorders. The main clinical manifestations are microcephaly with pontine and cerebellar hypoplasia, X-linked mental disorders with or without nystagmus and FG syndrome. The main pathogenic mechanism is the loss of function of related protein caused by variant. We reported a Chinese male newborn with a de novo variant in CASK gene. Case presentation We present an 18-day-old baby with growth retardation and brain hypoplasia. Whole-exome sequencing was performed, which detected a hemizygous missense variant c.764G > A of CASK gene. The variant changed the 255th amino acid from Arg to His. Software based bioinformatics analyses were conducted to infer its functional effect. Conclusions In this paper, a de novo variant of CASK gene was reported. Moreover, a detailed description of all the cases described in the literature is reported. CASK variants cause a variety of clinical phenotypes. Its diagnosis is difficult due to the lack of typical clinical symptoms. Genetic testing should be performed as early as possible if this disease is suspected. This case provides an important reference for the diagnosis and treatment of future cases.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Neonatology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.,Graduate College of Tianjin Medical University, Tianjin, China
| | - Yanyan Nie
- Department of Neonatology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Yu Mu
- Department of Neonatology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Jie Zheng
- Graduate College of Tianjin Medical University, Tianjin, China
| | - Xiaowei Xu
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China.,Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Fang Zhang
- Department of Neonatology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Jianbo Shu
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China. .,Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| | - Yang Liu
- Department of Neonatology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| |
Collapse
|
15
|
Mukherjee K, LaConte LEW, Srivastava S. The Non-Linear Path from Gene Dysfunction to Genetic Disease: Lessons from the MICPCH Mouse Model. Cells 2022; 11:1131. [PMID: 35406695 PMCID: PMC8997851 DOI: 10.3390/cells11071131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/09/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Most human disease manifests as a result of tissue pathology, due to an underlying disease process (pathogenesis), rather than the acute loss of specific molecular function(s). Successful therapeutic strategies thus may either target the correction of a specific molecular function or halt the disease process. For the vast majority of brain diseases, clear etiologic and pathogenic mechanisms are still elusive, impeding the discovery or design of effective disease-modifying drugs. The development of valid animal models and their proper characterization is thus critical for uncovering the molecular basis of the underlying pathobiological processes of brain disorders. MICPCH (microcephaly and pontocerebellar hypoplasia) is a monogenic condition that results from variants of an X-linked gene, CASK (calcium/calmodulin-dependent serine protein kinase). CASK variants are associated with a wide range of clinical presentations, from lethality and epileptic encephalopathies to intellectual disabilities, microcephaly, and autistic traits. We have examined CASK loss-of-function mutations in model organisms to simultaneously understand the pathogenesis of MICPCH and the molecular function/s of CASK. Our studies point to a highly complex relationship between the potential molecular function/s of CASK and the phenotypes observed in model organisms and humans. Here we discuss the implications of our observations from the pathogenesis of MICPCH as a cautionary narrative against oversimplifying molecular interpretations of data obtained from genetically modified animal models of human diseases.
Collapse
Affiliation(s)
- Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA; (L.E.W.L.); (S.S.)
- Department of Psychiatry, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Leslie E. W. LaConte
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA; (L.E.W.L.); (S.S.)
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Sarika Srivastava
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA; (L.E.W.L.); (S.S.)
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
16
|
Cuttler K, Hassan M, Carr J, Cloete R, Bardien S. Emerging evidence implicating a role for neurexins in neurodegenerative and neuropsychiatric disorders. Open Biol 2021; 11:210091. [PMID: 34610269 PMCID: PMC8492176 DOI: 10.1098/rsob.210091] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Synaptopathies are brain disorders characterized by dysfunctional synapses, which are specialized junctions between neurons that are essential for the transmission of information. Synaptic dysfunction can occur due to mutations that alter the structure and function of synaptic components or abnormal expression levels of a synaptic protein. One class of synaptic proteins that are essential to their biology are cell adhesion proteins that connect the pre- and post-synaptic compartments. Neurexins are one type of synaptic cell adhesion molecule that have, recently, gained more pathological interest. Variants in both neurexins and their common binding partners, neuroligins, have been associated with several neuropsychiatric disorders. In this review, we summarize some of the key physiological functions of the neurexin protein family and the protein networks they are involved in. Furthermore, examination of published literature has implicated neurexins in both neuropsychiatric and neurodegenerative disorders. There is a clear link between neurexins and neuropsychiatric disorders, such as autism spectrum disorder and schizophrenia. However, multiple expression studies have also shown changes in neurexin expression in several neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Therefore, this review highlights the potential importance of neurexins in brain disorders and the importance of doing more targeted studies on these genes and proteins.
Collapse
Affiliation(s)
- Katelyn Cuttler
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Maryam Hassan
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Cape Town, South Africa
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| | - Ruben Cloete
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Cape Town, South Africa
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| |
Collapse
|
17
|
Mondal A, Mukherjee S, Dar W, Singh S, Pati S. Role of glucose 6-phosphate dehydrogenase (G6PD) deficiency and its association to Autism Spectrum Disorders. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166185. [PMID: 34087423 DOI: 10.1016/j.bbadis.2021.166185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/27/2022]
Abstract
Autism Spectrum Disorder (ASD) is a common group of neurodevelopmental disorders which causes significant alterations in social and communication skills along with repetitive behavior and limited interests. The physiological understanding of ASD is ambiguous. Several reports suggested that environmental, genetic and epigenetic changes, neuroinflammation, mitochondrial dysfunction and metabolic alterations orchestrate the pathological outcomes of ASD. A recent report from Saudi Arabia found a mutation in X-chromosomal housekeeping glucose 6-phosphate dehydrogenase (G6PD) gene in two male ASD patients. Although, the involvement of G6PD-deficiency in the pathogenesis of ASD is poorly understood. Several reports suggested that G6PD deficiency impedes cellular detoxification of reactive oxygen species (ROS), which may result in neuronal damage and neuroinflammation. A deficiency of G6PD in newborn children may play a fundamental role in the pathogenesis of ASD. In this review, we will discuss the implications of G6PD deficiency in pathogenesis, male biasness and theranostics in ASD patients.
Collapse
Affiliation(s)
- Abir Mondal
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Soumyadeep Mukherjee
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Waseem Dar
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| | - Soumya Pati
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India.
| |
Collapse
|
18
|
Dutta RK, Larsson M, Arnesen T, Heie A, Walz M, Alesina P, Gimm O, Söderkvist P. X-chromosome variants are associated with aldosterone producing adenomas. Sci Rep 2021; 11:10562. [PMID: 34006971 PMCID: PMC8131628 DOI: 10.1038/s41598-021-89986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/04/2021] [Indexed: 11/09/2022] Open
Abstract
Aldosterone-producing adenomas (APAs) are a major cause of primary aldosteronism (PA) and are characterized by constitutively producing aldosterone, which leads to hypertension. Several mutations have been identified in ion channels or ion channel-associated genes that result in APAs. To date, no studies have used a genome-wide association study (GWAS) approach to search for predisposing loci for APAs. Thus, we investigated Scandinavian APA cases (n = 35) and Swedish controls (n = 60) in a GWAS and discovered a susceptibility locus on chromosome Xq13.3 (rs2224095, OR = 7.9, 95% CI = 2.8-22.4, P = 1 × 10-7) in a 4-Mb region that was significantly associated with APA. Direct genotyping of sentinel SNP rs2224095 in a replication cohort of APAs (n = 83) and a control group (n = 740) revealed persistently strong significance (OR = 6.1, 95% CI = 3.5-10.6, p < 0.0005). We sequenced an adjacent gene, MAGEE1, of the sentinel SNP and identified a rare variant in one APA, p.Gly327Glu, which is complementary to other mutations in our primary cohort. Expression quantitative trait loci (eQTL) were investigated on the X-chromosome, and 24 trans-eQTL were identified. Some of the genes identified by trans-eQTL point towards a novel mechanistic explanation for the association of the SNPs with APAs. In conclusion, our study provides further insights into the genetic basis of APAs.
Collapse
Affiliation(s)
- Ravi Kumar Dutta
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden. .,Department of Clinical and Experimental Medicine, Linköping University, 58183, Linköping, Sweden.
| | - Malin Larsson
- Science for Life Laboratory, Department of Physics, Chemistry and Biology, Linköping University, 58183, Linköping, Sweden
| | - Thomas Arnesen
- Department of Surgery, Haukeland University Hospital, Bergen, Norway.,Department of Biological Sciences, University of Bergen, Bergen, Norway.,Department of Biosciences, University of Bergen, Bergen, Norway
| | - Anette Heie
- Department of Surgery, Haukeland University Hospital, Bergen, Norway.,Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Martin Walz
- Klinik Für Chirurgie and Zentrum Für Minimal Invasive Chirurgie, Klinikum Essen-Mitte, Essen, Germany
| | - Piero Alesina
- Klinik Für Chirurgie and Zentrum Für Minimal Invasive Chirurgie, Klinikum Essen-Mitte, Essen, Germany
| | - Oliver Gimm
- Department of Surgery and Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| | - Peter Söderkvist
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| |
Collapse
|
19
|
CASK related disorder: Epilepsy and developmental outcome. Eur J Paediatr Neurol 2021; 31:61-69. [PMID: 33640666 DOI: 10.1016/j.ejpn.2021.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE CASK pathogenic variants are associated with variable features, as intellectual disability, optic atrophy, brainstem/cerebellar hypoplasia, and epileptic encephalopathy. Few studies describe the electroclinical features of epilepsy in patients with CASK pathogenic variants and their relationship with developmental delay. METHODS this national multicentre cohort included genetically confirmed patients with different CASK pathogenic variants. Our findings were compared with cohorts reported in the literature. RESULTS we collected 34 patients (29 females) showing from moderate (4 patients) to severe (22) and profound (8) developmental delay; all showed pontine and cerebellar hypoplasia, all except three with microcephaly. Seventeen out of 34 patients (50%) suffered from epileptic seizures, including spasms (11 patients, 32.3%), generalized (5) or focal seizures (1). In 8/17 individuals (47.1%), epilepsy started at or beyond the age of 24 months. Seven (3 males) out of the 11 children with spasms showed EEG features and a course supporting the diagnosis of a developmental and epileptic encephalopathy (DEE). Drug resistance was frequent in our cohort (52.9% of patients with epilepsy). EEG abnormalities included poorly organized background activity with diffuse or multifocal epileptiform abnormalities and sleep-activation, with possible appearance over the follow-up period. Developmental delay degree was not statistically different among patients with or without seizures but feeding difficulties were more frequent in patients with epilepsy. CONCLUSIONS epilepsy is a frequent comorbidity with a high incidence of spasms and drug resistance. Overall developmental disability does not seem to be more severe in the group of patients with epilepsy nor to be linked to specific epilepsy/EEG characteristics. A childhood onset of epilepsy is frequent, with possible worsening over time, so that serial and systematic monitoring is mandatory.
Collapse
|
20
|
Nishio Y, Kidokoro H, Takeo T, Narita H, Sawamura F, Narita K, Kawano Y, Nakata T, Muramatsu H, Hara S, Kaname T, Natsume J. The eldest case of MICPCH with CASK mutation exhibiting gross motor regression. Brain Dev 2021; 43:459-463. [PMID: 33272775 DOI: 10.1016/j.braindev.2020.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/02/2020] [Accepted: 11/15/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND MICPCH is manifested as microcephaly associated with pontocerebellar hypoplasia and global developmental delay but developmental regression has never been reported. We describe the detailed clinical history of a woman with intellectual disability and microcephaly with pontine and cerebellar hypoplasia (MICPCH) with a CASK mutation who exhibited gross motor regression after adolescence. CASE The patient experienced severe motor and intellectual developmental delay with microcephaly from infancy. The initial diagnosis was Rett syndrome based on her clinical features, including hand stereotypes and the absence of structural abnormality on magnetic resonance imaging (MRI) performed at the age of 5 years. Although gross motor abilities developed slowly and she could walk independently, she never acquired speech or understanding of languages. After adolescence, her motor ability gradually regressed so that she was unable to stand without support and moved with a wheelchair. At the age of 31 years, because of her atypical clinical course for Rett syndrome, whole exome sequencing was performed, which revealed a de novo heterozygous c.2068 + 1G > A mutation in the CASK gene (NM_001126055). Brain MRI revealed mild pontocerebellar hypoplasia compatible with the clinical phenotype of MICPCH. DISCUSSION This case suggests that MICPCH with a CASK mutation might cause developmental regression after adolescence and might be regarded as a neurodegenerative disorder.
Collapse
Affiliation(s)
- Yosuke Nishio
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Pediatrics, TOYOTA Memorial Hospital, Toyota, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Toshiki Takeo
- Department of Pediatrics, TOYOTA Memorial Hospital, Toyota, Japan
| | - Hajime Narita
- Department of Pediatrics, TOYOTA Memorial Hospital, Toyota, Japan
| | - Fumi Sawamura
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kotaro Narita
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiko Kawano
- Department of Pediatrics, TOYOTA Memorial Hospital, Toyota, Japan
| | - Tomohiko Nakata
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Hara
- Department of Pediatrics, TOYOTA Memorial Hospital, Toyota, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Jun Natsume
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Disability Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
21
|
Zhao J, Hou M, Wang H, Liu Q, Sun D, Wei W. Microcephaly, disproportionate pontine, and cerebellar hypoplasia syndrome: Two novel mutations in the CASK gene were discovered in Chinese females. Int J Dev Neurosci 2021; 81:277-284. [PMID: 33629417 DOI: 10.1002/jdn.10100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/08/2021] [Accepted: 02/21/2021] [Indexed: 11/10/2022] Open
Abstract
Microcephaly, disproportionate pontine and cerebellar hypoplasia (MICPCH) syndrome is a rare and genetic disorder, which is mainly caused by mutations in the CASK gene. We described four variations in the CASK gene in Chinese female patients with MICPCH, who presented with microcephaly, developmental delay, and motor disorder. The CASK mutations were identified using NGS (the next-generation sequencing), copy number variation sequencing. Two novel variations in the CASK gene were revealed including a frameshift mutation c.1000_1001insG (p.Asp334GlyfsTer32) and a nonsense mutation c.2110A > T (p.Lys704Ter). Two other aberrations were c.316C > T (p.Arg106Ter) and Xp11.4-p11.3 (41,700,001-44,660,000) × 1 loss. We provided clinical manifestations and neuroimaging findings of the four patients. The genetic variation spectrum of MICPCH caused by CASK was updated. Furthermore, we expounded on the molecular mechanism of the disease and noticed that it was not possible to relate the magnitude of the genetic alteration to a particular phenotype.
Collapse
Affiliation(s)
- Jianhui Zhao
- Department of Neurology & Rehabilitation, Qingdao Women and Children's Hospital, Qingdao, P.R. China
| | - Mei Hou
- Department of Neurology & Rehabilitation, Qingdao Women and Children's Hospital, Qingdao, P.R. China
| | - Haiqiao Wang
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Qiuyan Liu
- Department of Neurology & Rehabilitation, Qingdao Women and Children's Hospital, Qingdao, P.R. China
| | - Dianrong Sun
- Department of Neurology & Rehabilitation, Qingdao Women and Children's Hospital, Qingdao, P.R. China
| | - Wei Wei
- Kangso Medical Inspection Co, Ltd., Beijing, P.R. China
| |
Collapse
|
22
|
Pan YE, Tibbe D, Harms FL, Reißner C, Becker K, Dingmann B, Mirzaa G, Kattentidt-Mouravieva AA, Shoukier M, Aggarwal S, Missler M, Kutsche K, Kreienkamp HJ. Missense mutations in CASK, coding for the calcium-/calmodulin-dependent serine protein kinase, interfere with neurexin binding and neurexin-induced oligomerization. J Neurochem 2020; 157:1331-1350. [PMID: 33090494 DOI: 10.1111/jnc.15215] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
Mutations in the X-linked gene coding for the calcium-/calmodulin-dependent serine protein kinase (CASK) are associated with severe neurological disorders ranging from intellectual disability (in males) to mental retardation and microcephaly with pontine and cerebellar hypoplasia. CASK is involved in transcription control, in the regulation of trafficking of the post-synaptic NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, and acts as a presynaptic scaffolding protein. For CASK missense mutations, it is mostly unclear which of CASK's molecular interactions and cellular functions are altered and contribute to patient phenotypes. We identified five CASK missense mutations in male patients affected by neurodevelopmental disorders. These and five previously reported mutations were systematically analysed with respect to interaction with CASK interaction partners by co-expression and co-immunoprecipitation. We show that one mutation in the L27 domain interferes with binding to synapse-associated protein of 97 kDa. Two mutations in the guanylate kinase (GK) domain affect binding of CASK to the nuclear factors CASK-interacting nucleosome assembly protein (CINAP) and T-box, brain, 1 (Tbr1). A total of five mutations in GK as well as PSD-95/discs large/ZO-1 (PDZ) domains affect binding of CASK to the pre-synaptic cell adhesion molecule Neurexin. Upon expression in neurons, we observe that binding to Neurexin is not required for pre-synaptic localization of CASK. We show by bimolecular fluorescence complementation assay that Neurexin induces oligomerization of CASK, and that mutations in GK and PDZ domains interfere with the Neurexin-induced oligomerization of CASK. Our data are supported by molecular modelling, where we observe that the cooperative activity of PDZ, SH3 and GK domains is required for Neurexin binding and oligomerization of CASK.
Collapse
Affiliation(s)
- Yingzhou Edward Pan
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Debora Tibbe
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Leonie Harms
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Reißner
- Institut für Anatomie und Molekulare Neurobiologie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Bri Dingmann
- Medical Genetics Department, Seattle Children's Hospital, Seattle, Washington, DC, USA
| | - Ghayda Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, DC, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, DC, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | | | - Moneef Shoukier
- Pränatal-Medizin München, Frauenärzte und Humangenetiker MVZ, München, Germany
| | - Shagun Aggarwal
- Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
| | - Markus Missler
- Institut für Anatomie und Molekulare Neurobiologie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Kerstin Kutsche
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Mukherjee K, Patel PA, Rajan DS, LaConte LEW, Srivastava S. Survival of a male patient harboring CASK Arg27Ter mutation to adolescence. Mol Genet Genomic Med 2020; 8:e1426. [PMID: 32696595 PMCID: PMC7549553 DOI: 10.1002/mgg3.1426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/01/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background CASK is an X‐linked gene in mammals and its deletion in males is incompatible with life. CASK heterozygous mutations in female patients associate with intellectual disability, microcephaly, pontocerebellar hypoplasia, and optic nerve hypoplasia, whereas CASK hemizygous mutations in males manifest as early infantile epileptic encephalopathy with a grim prognosis. Here, we report a rare case of survival of a male patient harboring a CASK null mutation to adolescent age. Methods Trio whole exome sequencing analysis was performed from blood genomic DNA. Magnetic resonance imaging (MRI), magnetic resonance spectroscopy (MRS), and electroencephalogram (EEG) analyses were performed to determine anomalies in brain development, metabolite concentrations, and electrical activity, respectively. Results Trio‐WES analysis identified a de novo c.79C>T (p.Arginine27Ter) mutation in CASK causing a premature translation termination at the very N‐terminus of the protein. The 17‐years, and 11‐month‐old male patient displayed profound intellectual disability, microcephaly, dysmorphism, ponto‐cerebellar hypoplasia, and intractable epilepsy. His systemic symptoms included overall reduced somatic growth, dysautonomia, ventilator and G tube dependence, and severe osteopenia. Brain MRI revealed a severe cerebellar and brain stem hypoplasia with progressive cerebral atrophy. EEG spectral analysis revealed a global functional defect with generalized background slowing and delta waves dominating even in the awake state. Conclusion This case study is the first to report survival of a male patient carrying a CASK loss‐of‐function mutation to adolescence and highlights that improved palliative care could extend survival. Moreover, the genomic position encoding Arg27 in CASK may possess an increased susceptibility to mutations.
Collapse
Affiliation(s)
- Konark Mukherjee
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Paras A Patel
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Deepa S Rajan
- Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leslie E W LaConte
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Sarika Srivastava
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.,Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
24
|
Chanchani SR, Xie H, Sekhon G, Melikishvili AM, Moyer Harasink S, Pall H, Giampietro PF. A male infant with Xq22.2q22.3 duplication containing PLP1 and MID2. Mol Genet Genomic Med 2020; 8:e1078. [PMID: 31951325 PMCID: PMC7057127 DOI: 10.1002/mgg3.1078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/26/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022] Open
Abstract
Background The Xq22.2 q23 is a complex genomic region which includes the genes MID2 and PLP1 associated with FG syndrome 5 and Pelizaeus–Merzbacher disease, respectively. There is limited information regarding the clinical outcomes observed in patients with deletions within this region. Methods We report on a male infant with intrauterine growth retardation (IUGR) who developed head titubation and spasticity during his postnatal hospital course. Results Chromosome microarray revealed a 6.7 Mb interstitial duplication of Xq22.2q22.3. Fluorescence in situ hybridization showed that the patient's mother also possessed the identical duplication in the Xq22.3q22.3 region. Among the 34 OMIM genes in this interval, the duplication of the PLP1 (OMIM# 300401) and MID2 (OMIM# 300204) appears to be the most significant contributors to the patient's clinical features. Mutations and duplications of PLP1 are associated with X‐linked recessive Pelizaeus–Merzbacher disease (PMD). A single case of a Xq22.3 duplication including the MID2 has been reported in boy with features of FG syndrome. However, our patient's clinical features are not consistent with the FG syndrome phenotype. Conclusion Our patient's clinical features appear to be influenced by the PLP1 duplication but the clinical effect of other dosage sensitive genes influencing brain development cannot be ruled out.
Collapse
Affiliation(s)
- Swati R Chanchani
- Department of Pediatrics St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Hongyan Xie
- Tricore Reference Laboratory, Albuquerque, NM, USA
| | | | | | - Sue Moyer Harasink
- Department of Pediatrics St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Harpreet Pall
- Department of Pediatrics St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
25
|
Fabbretti E. P2X3 receptors are transducers of sensory signals. Brain Res Bull 2019; 151:119-124. [PMID: 30660716 DOI: 10.1016/j.brainresbull.2018.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/27/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022]
Abstract
Peripheral stimuli are transduced by specific receptors expressed by sensory neurons and are further processed in the dorsal horn of spinal cord before to be transmitted to the brain. While relative few receptor subtypes mediate the initial depolarisation of sensory neurons, an impressive number of molecules and ion channels integrate these inputs into coded signals. Soluble mediators and ambient conditions further shape these processes, potentially triggering peripheral and central sensitisation, or sensory downregulation. Extracellular ATP is a major signaling molecule that acts via purinergic receptors and is a powerful modulator of cell communication as well as a neurotransmitter at peripheral/central synapses. In particular, ATP-mediated signals are transduced by P2X3 receptors expressed mainly by peripheral sensory neurons. Recent evidence suggests that P2X3 receptor function not only induces neuron depolarisation and firing with consequent neurotransmitter release, but it also triggers intracellular molecular changes that amplify purinergic signaling with important consequences.
Collapse
Affiliation(s)
- Elsa Fabbretti
- Department of Life Science, University of Trieste, via Giorgieri 5, 34127, Trieste, Italy.
| |
Collapse
|
26
|
Mori T, Kasem EA, Suzuki-Kouyama E, Cao X, Li X, Kurihara T, Uemura T, Yanagawa T, Tabuchi K. Deficiency of calcium/calmodulin-dependent serine protein kinase disrupts the excitatory-inhibitory balance of synapses by down-regulating GluN2B. Mol Psychiatry 2019; 24:1079-1092. [PMID: 30610199 PMCID: PMC6756202 DOI: 10.1038/s41380-018-0338-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 11/24/2022]
Abstract
Calcium/calmodulin-dependent serine protein kinase (CASK) is a membrane-associated guanylate kinase (MAGUK) protein that is associated with neurodevelopmental disorders. CASK is thought to have both pre- and postsynaptic functions, but the mechanism and consequences of its functions in the brain have yet to be elucidated, because homozygous CASK-knockout (CASK-KO) mice die before brain maturation. Taking advantage of the X-chromosome inactivation (XCI) mechanism, here we examined the synaptic functions of CASK-KO neurons in acute brain slices of heterozygous CASK-KO female mice. We also analyzed CASK-knockdown (KD) neurons in acute brain slices generated by in utero electroporation. Both CASK-KO and CASK-KD neurons showed a disruption of the excitatory and inhibitory (E/I) balance. We further found that the expression level of the N-methyl-D-aspartate receptor subunit GluN2B was decreased in CASK-KD neurons and that overexpressing GluN2B rescued the disrupted E/I balance in CASK-KD neurons. These results suggest that the down-regulation of GluN2B may be involved in the mechanism of the disruption of synaptic E/I balance in CASK-deficient neurons.
Collapse
Affiliation(s)
- Takuma Mori
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Enas A. Kasem
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan ,0000 0004 0578 3577grid.411978.2Department of Zoology, Faculty of Science, Kafr Elsheikh University, Kafr Elsheihk, 33511 Egypt
| | - Emi Suzuki-Kouyama
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Xueshan Cao
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Xue Li
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Taiga Kurihara
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Takeshi Uemura
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan ,0000 0001 1507 4692grid.263518.bInstitute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621 Japan ,0000 0004 1754 9200grid.419082.6CREST, JST, Saitama, 332-0012 Japan
| | - Toru Yanagawa
- 0000 0001 2369 4728grid.20515.33Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575 Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan. .,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan. .,PRESTO, JST, Saitama, 332-0012, Japan.
| |
Collapse
|
27
|
Bozarth X, Foss K, Mefford HC. A de novo in-frame deletion of CASK
gene causes early onset infantile spasms and supratentorial cerebral malformation in a female patient. Am J Med Genet A 2018; 176:2425-2429. [DOI: 10.1002/ajmg.a.40429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/06/2018] [Accepted: 06/10/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Xiuhua Bozarth
- Department of Neurology; Seattle Children's Hospital, University of Washington; Seattle Washington
| | - Kimberly Foss
- Department of Genetics; Seattle Children's Hospital; Seattle Washington
| | - Heather C. Mefford
- Division of Genetic Medicine, Department of Pediatrics; University of Washington; Seattle Washington
| |
Collapse
|
28
|
Cristofoli F, Devriendt K, Davis EE, Van Esch H, Vermeesch JR. Novel CASK mutations in cases with syndromic microcephaly. Hum Mutat 2018; 39:993-1001. [PMID: 29691940 PMCID: PMC5995665 DOI: 10.1002/humu.23536] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 11/05/2022]
Abstract
Mutations in CASK cause a wide spectrum of phenotypes in humans ranging from mild X-linked intellectual disability to a severe microcephaly (MC) and pontocerebellar hypoplasia syndrome. Nevertheless, predicting pathogenicity and phenotypic consequences of novel CASK mutations through the exclusive consideration of genetic information and population-based data remains a challenge. Using whole exome sequencing, we identified four novel CASK mutations in individuals with syndromic MC. To understand the functional consequences of the different point mutations on the development of MC and cerebellar defects, we established a transient loss-of-function zebrafish model, and demonstrate recapitulation of relevant neuroanatomical phenotypes. Furthermore, we utilized in vivo complementation studies to demonstrate that the three point mutations confer a loss-of-function effect. This work endorses zebrafish as a tractable model to rapidly assess the effect of novel CASK variants on brain development.
Collapse
Affiliation(s)
- Francesca Cristofoli
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Koen Devriendt
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for the Genetics of Cognition, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Joris R Vermeesch
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
29
|
LaConte LEW, Srivastava S, Mukherjee K. Probing Protein Kinase-ATP Interactions Using a Fluorescent ATP Analog. Methods Mol Biol 2018; 1647:171-183. [PMID: 28809002 DOI: 10.1007/978-1-4939-7201-2_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Eukaryotic protein kinases are an intensely investigated class of enzymes which have garnered attention due to their usefulness as drug targets. Determining the regulation of ATP binding to a protein kinase is not only critical for understanding function in a cellular context but also for designing kinase-specific molecular inhibitors. Here, we provide a general procedure for characterizing ATP binding to eukaryotic protein kinases. The protocol can be adapted to identify the conditions under which a particular kinase is activated. The approach is simple, requiring only a fluorescent ATP analog such as TNP-ATP or MANT-ATP and an instrument to monitor changes in fluorescence. Although the interaction kinetics between a kinase and a given ATP analog may differ from that of native ATP, this disadvantage is offset by the ease of performing and interpreting this assay. Importantly, it can be optimized to probe a large variety of conditions under which the kinase-nucleotide binding might be affected.
Collapse
Affiliation(s)
- Leslie E W LaConte
- Virginia Tech Carilion Research Institute, Roanoke, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Sarika Srivastava
- Virginia Tech Carilion Research Institute, Roanoke, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Konark Mukherjee
- Virginia Tech Carilion Research Institute, Roanoke, 2 Riverside Circle, Roanoke, VA, 24016, USA. .,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24060, USA.
| |
Collapse
|
30
|
Hayashi S, Uehara DT, Tanimoto K, Mizuno S, Chinen Y, Fukumura S, Takanashi JI, Osaka H, Okamoto N, Inazawa J. Comprehensive investigation of CASK mutations and other genetic etiologies in 41 patients with intellectual disability and microcephaly with pontine and cerebellar hypoplasia (MICPCH). PLoS One 2017; 12:e0181791. [PMID: 28783747 PMCID: PMC5546575 DOI: 10.1371/journal.pone.0181791] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/09/2017] [Indexed: 01/10/2023] Open
Abstract
The CASK gene (Xp11.4) is highly expressed in the mammalian nervous system and plays several roles in neural development and synaptic function. Loss-of-function mutations of CASK are associated with intellectual disability and microcephaly with pontine and cerebellar hypoplasia (MICPCH), especially in females. Here, we present a comprehensive investigation of 41 MICPCH patients, analyzed by mutational search of CASK and screening of candidate genes using an SNP array, targeted resequencing and whole-exome sequencing (WES). In total, we identified causative or candidate genomic aberrations in 37 of the 41 cases (90.2%). CASK aberrations including a rare mosaic mutation in a male patient, were found in 32 cases, and a mutation in ITPR1, another known gene in which mutations are causative for MICPCH, was found in one case. We also found aberrations involving genes other than CASK, such as HDAC2, MARCKS, and possibly HS3ST5, which may be associated with MICPCH. Moreover, the targeted resequencing screening detected heterozygous variants in RELN in two cases, of uncertain pathogenicity, and WES analysis suggested that concurrent mutations of both DYNC1H1 and DCTN1 in one case could lead to MICPCH. Our results not only identified the etiology of MICPCH in nearly all the investigated patients but also suggest that MICPCH is a genetically heterogeneous condition, in which CASK inactivating mutations appear to account for the majority of cases.
Collapse
Affiliation(s)
- Shin Hayashi
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Hard Tissue Genome Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, United States of America
- * E-mail: (SH); (JI)
| | - Daniela Tiaki Uehara
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kousuke Tanimoto
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Genome Laboratory, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seiji Mizuno
- Department of Pediatrics, Central Hospital, Aichi Human Service Center, Kasugai, Japan
| | - Yasutsugu Chinen
- Department of Pediatrics, University of the Ryukyus School of Medicine, Nishihara, Japan
| | - Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jun-ichi Takanashi
- Department of Pediatrics, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical School, Tochigi, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Hard Tissue Genome Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail: (SH); (JI)
| |
Collapse
|
31
|
Seto T, Hamazaki T, Nishigaki S, Kudo S, Shintaku H, Ondo Y, Shimojima K, Yamamoto T. A novel CASK mutation identified in siblings exhibiting developmental disorders with/without microcephaly. Intractable Rare Dis Res 2017; 6:177-182. [PMID: 28944139 PMCID: PMC5608927 DOI: 10.5582/irdr.2017.01031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The calcium/calmodulin-dependent serine protein kinase gene (CASK) mutations are associated with various neurological disorders; a syndrome of intellectual disability (ID) and microcephaly with pontine and cerebellar hypoplasia (MICPCH), FG syndrome, X-linked ID with/without nystagmus, epileptic encephalopathy, and autistic spectrum disorder (ASD). Next generation sequencing was performed to elucidate genetic causes in siblings exhibiting developmental disorders, and a novel CASK mutation, c.1424G>T (p.Ser475Ile), was detected in a male patient with ID, ASD, and microcephaly. Radiological examination of his brain showed no structural abnormality. The identified mutation was shared with the healthy mother and a younger sister exhibiting ASD. Although the mother showed a skewed X-chromosome inactivation (XCI) pattern, the sister showed a paradoxical XCI pattern. This would explain why this sister possessed a normal intellectual level, but showed the same ASD symptoms as the affected brother. A novel CASK mutation was identified in two siblings with ID and/or ASD, suggesting a relationship between the CASK mutation and ASD. Recently performed large molecular cohorts for patients with developmental disorders suggest that CASK is one of the genes related to developmental disorders. For better understanding of genotype-phenotype correlation in ASD cases with CASK mutations, more information should be accumulated.
Collapse
Affiliation(s)
- Toshiyuki Seto
- Department of Pediatrics, Osaka City University, Osaka, Japan
| | | | | | - Satoshi Kudo
- Department of Pediatrics, Osaka City University, Osaka, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University, Osaka, Japan
| | - Yumiko Ondo
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Keiko Shimojima
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiyuki Yamamoto
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
- Address correspondence to: Dr. Toshiyuki Yamamoto, Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ward, Tokyo 162-8666, Japan. E-mail:
| |
Collapse
|
32
|
Yang F, Sun S, Tan G, Costanzo M, Hill DE, Vidal M, Andrews BJ, Boone C, Roth FP. Identifying pathogenicity of human variants via paralog-based yeast complementation. PLoS Genet 2017; 13:e1006779. [PMID: 28542158 PMCID: PMC5466341 DOI: 10.1371/journal.pgen.1006779] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 06/09/2017] [Accepted: 04/25/2017] [Indexed: 11/21/2022] Open
Abstract
To better understand the health implications of personal genomes, we now face a largely unmet challenge to identify functional variants within disease-associated genes. Functional variants can be identified by trans-species complementation, e.g., by failure to rescue a yeast strain bearing a mutation in an orthologous human gene. Although orthologous complementation assays are powerful predictors of pathogenic variation, they are available for only a few percent of human disease genes. Here we systematically examine the question of whether complementation assays based on paralogy relationships can expand the number of human disease genes with functional variant detection assays. We tested over 1,000 paralogous human-yeast gene pairs for complementation, yielding 34 complementation relationships, of which 33 (97%) were novel. We found that paralog-based assays identified disease variants with success on par with that of orthology-based assays. Combining all homology-based assay results, we found that complementation can often identify pathogenic variants outside the homologous sequence region, presumably because of global effects on protein folding or stability. Within our search space, paralogy-based complementation more than doubled the number of human disease genes with a yeast-based complementation assay for disease variation. Functional complementation assays of human disease-associated gene variants can reveal many more human disease variants at high confidence than current computational approaches, even using highly-diverged model organisms. However, this has generally only been possible for a minority of human disease genes for which orthologous complementation is known in the relevant model organism, so that alternative assays are urgently needed. Here we show that complementation relationships can be found for many additional human disease genes by exploiting paralogous human-yeast gene relationships, and that disease variant identification using paralogy-based assays performs on par with orthology-based assays.
Collapse
Affiliation(s)
- Fan Yang
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Song Sun
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Guihong Tan
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Costanzo
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - David E. Hill
- Center for Cancer Systems Biology (CCSB), Dana- Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana- Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brenda J. Andrews
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Charles Boone
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Frederick P. Roth
- Donnelly Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Center for Cancer Systems Biology (CCSB), Dana- Farber Cancer Institute, Boston, Massachusetts, United States of America
- Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
33
|
Grau C, Starkovich M, Azamian MS, Xia F, Cheung SW, Evans P, Henderson A, Lalani SR, Scott DA. Xp11.22 deletions encompassing CENPVL1, CENPVL2, MAGED1 and GSPT2 as a cause of syndromic X-linked intellectual disability. PLoS One 2017; 12:e0175962. [PMID: 28414775 PMCID: PMC5393878 DOI: 10.1371/journal.pone.0175962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/03/2017] [Indexed: 12/27/2022] Open
Abstract
By searching a clinical database of over 60,000 individuals referred for array-based CNV analyses and online resources, we identified four males from three families with intellectual disability, developmental delay, hypotonia, joint hypermobility and relative macrocephaly who carried small, overlapping deletions of Xp11.22. The maximum region of overlap between their deletions spanned ~430 kb and included two pseudogenes, CENPVL1 and CENPVL2, whose functions are not known, and two protein coding genes-the G1 to S phase transition 2 gene (GSPT2) and the MAGE family member D1 gene (MAGED1). Deletions of this ~430 kb region have not been previously implicated in human disease. Duplications of GSPT2 have been documented in individuals with intellectual disability, but the phenotypic consequences of a loss of GSPT2 function have not been elucidated in humans or mouse models. Changes in MAGED1 have not been associated with intellectual disability in humans, but loss of MAGED1 function is associated with neurocognitive and neurobehavioral phenotypes in mice. In all cases, the Xp11.22 deletion was inherited from an unaffected mother. Studies performed on DNA from one of these mothers did not show evidence of skewed X-inactivation. These results suggest that deletions of an ~430 kb region on chromosome Xp11.22 that encompass CENPVL1, CENPVL2, GSPT2 and MAGED1 cause a distinct X-linked syndrome characterized by intellectual disability, developmental delay, hypotonia, joint hypermobility and relative macrocephaly. Loss of GSPT2 and/or MAGED1 function may contribute to the intellectual disability and developmental delay seen in males with these deletions.
Collapse
Affiliation(s)
- Christina Grau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Molly Starkovich
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mahshid S. Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fan Xia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics, Houston, Texas, Unite States of America
| | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics, Houston, Texas, Unite States of America
| | - Patricia Evans
- Departments of Pediatrics and Neurology, University of Texas Southwestern Medical School, Dallas, Texas, United States of America
| | - Alex Henderson
- The Newcastle upon Tyne Hospitals, Newcastle upon Tyne, England
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
34
|
Dunn P, Prigatano GP, Szelinger S, Roth J, Siniard AL, Claasen AM, Richholt RF, De Both M, Corneveaux JJ, Moskowitz AM, Balak C, Piras IS, Russell M, Courtright AL, Belnap N, Rangasamy S, Ramsey K, Opitz JM, Craig DW, Narayanan V, Huentelman MJ, Schrauwen I. A de novo splice site mutation inCASKcauses FG syndrome-4 and congenital nystagmus. Am J Med Genet A 2017; 173:611-617. [DOI: 10.1002/ajmg.a.38069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/14/2016] [Indexed: 11/07/2022]
|
35
|
Huang TN, Hsueh YP. Calcium/calmodulin-dependent serine protein kinase (CASK), a protein implicated in mental retardation and autism-spectrum disorders, interacts with T-Brain-1 (TBR1) to control extinction of associative memory in male mice. J Psychiatry Neurosci 2017; 42:37-47. [PMID: 28234597 PMCID: PMC5373711 DOI: 10.1503/jpn.150359] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Human genetic studies have indicated that mutations in calcium/calmodulin-dependent serine protein kinase (CASK) result in X-linked mental retardation and autism-spectrum disorders. We aimed to establish a mouse model to study how Cask regulates mental ability. METHODS Because Cask encodes a multidomain scaffold protein, a possible strategy to dissect how CASK regulates mental ability and cognition is to disrupt specific protein-protein interactions of CASK in vivo and then investigate the impact of individual specific protein interactions. Previous in vitro analyses indicated that a rat CASK T724A mutation reduces the interaction between CASK and T-brain-1 (TBR1) in transfected COS cells. Because TBR1 is critical for glutamate receptor, ionotropic, N-methyl-D-aspartate receptor subunit 2B (Grin2b) expression and is a causative gene for autism and intellectual disability, we then generated CASK T740A (corresponding to rat CASK T724A) mutant mice using a gene-targeting approach. Immunoblotting, coimmunoprecipitation, histological methods and behavioural assays (including home cage, open field, auditory and contextual fear conditioning and conditioned taste aversion) were applied to investigate expression of CASK and its related proteins, the protein-protein interactions of CASK, and anatomic and behavioural features of CASK T740A mice. RESULTS The CASK T740A mutation attenuated the interaction between CASK and TBR1 in the brain. However, CASK T740A mice were generally healthy, without obvious defects in brain morphology. The most dramatic defect among the mutant mice was in extinction of associative memory, though acquisition was normal. LIMITATIONS The functions of other CASK protein interactions cannot be addressed using CASK T740A mice. CONCLUSION Disruption of the CASK and TBR1 interaction impairs extinction, suggesting the involvement of CASK in cognitive flexibility.
Collapse
Affiliation(s)
| | - Yi-Ping Hsueh
- Correspondence to: Y.P. Hsueh, Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529, Taiwan;
| |
Collapse
|
36
|
Giovani ÉM, Marinho KCT, Andia-Merlin R. Dental treatment of a patient with Opitz G/BBB syndrome. SPECIAL CARE IN DENTISTRY 2016; 37:102-106. [PMID: 27642052 DOI: 10.1111/scd.12200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Opitz G/BBB syndrome is a genetic condition characterized by several abnormalities along the midline of the body, such as hypertelorism, craniofacial deformities, and dysphagia. This study reports the clinical features of Optiz syndrome and its importance in the knowledge of patients who are developmentally challenged as a whole, in order to establish adequate dental treatment for a certain clinical case. A 19-year-old patient visited the Paulista University for a dental treatment. The extraoral examination revealed ocular hypertelorism (wide-spaced eyes), oblique eyelids, epicanthus, low-set cart, and intellectual disability. During the intraoral examination, large caries lesions were observed surrounding the braces of the fixed orthodontic appliance and poor oral hygiene. Preventive and restorative treatments were carried out. It was concluded that the knowledge of patients with special needs as a whole is mandatory for an adequate dental treatment. This is a case report that highlights the importance of dentist and interdisciplinary care attendance for all patient systems, the examination and analyses should not be restricted to the oral cavity.
Collapse
Affiliation(s)
- Élcio Magdalena Giovani
- Chairman, Professor, Integrated Clinic Discipline, Coordinator of Center for Studies and Special Service for Patients, Professor, Postgraduate Dentistry Courses, UNIP, São Paulo, SP, Brazil
| | | | - Ruth Andia-Merlin
- Associate Professor, Integrated Clinic and Center for Studies and Special Service for Patients, UNIP, São Paulo, SP, Brazil
| |
Collapse
|
37
|
X-linked intellectual disability gene CASK regulates postnatal brain growth in a non-cell autonomous manner. Acta Neuropathol Commun 2016; 4:30. [PMID: 27036546 PMCID: PMC4818453 DOI: 10.1186/s40478-016-0295-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 01/07/2023] Open
Abstract
The phenotypic spectrum among girls with heterozygous mutations in the X-linked intellectual disability (XLID) gene CASK (calcium/calmodulin-dependent serine protein kinase) includes postnatal microcephaly, ponto-cerebellar hypoplasia, seizures, optic nerve hypoplasia, growth retardation and hypotonia. Although CASK knockout mice were previously reported to exhibit perinatal lethality and a 3-fold increased apoptotic rate in the brain, CASK deletion was not found to affect neuronal physiology and their electrical properties. The pathogenesis of CASK associated disorders and the potential function of CASK therefore remains unknown. Here, using Cre-LoxP mediated gene excision experiments; we demonstrate that deleting CASK specifically from mouse cerebellar neurons does not alter the cerebellar architecture or function. We demonstrate that the neuron-specific deletion of CASK in mice does not cause perinatal lethality but induces severe recurrent epileptic seizures and growth retardation before the onset of adulthood. Furthermore, we demonstrate that although neuron-specific haploinsufficiency of CASK is inconsequential, the CASK mutation associated human phenotypes are replicated with high fidelity in CASK heterozygous knockout female mice (CASK(+/-)). These data suggest that CASK-related phenotypes are not purely neuronal in origin. Surprisingly, the observed microcephaly in CASK(+/-) animals is not associated with a specific loss of CASK null brain cells indicating that CASK regulates postnatal brain growth in a non-cell autonomous manner. Using biochemical assay, we also demonstrate that CASK can interact with metabolic proteins. CASK knockdown in human cell lines cause reduced cellular respiration and CASK(+/-) mice display abnormalities in muscle and brain oxidative metabolism, suggesting a novel function of CASK in metabolism. Our data implies that some phenotypic components of CASK heterozygous deletion mutation associated disorders represent systemic manifestation of metabolic stress and therefore amenable to therapeutic intervention.
Collapse
|
38
|
Zhu J, Shang Y, Zhang M. Mechanistic basis of MAGUK-organized complexes in synaptic development and signalling. Nat Rev Neurosci 2016; 17:209-23. [DOI: 10.1038/nrn.2016.18] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Autism and Intellectual Disability-Associated KIRREL3 Interacts with Neuronal Proteins MAP1B and MYO16 with Potential Roles in Neurodevelopment. PLoS One 2015; 10:e0123106. [PMID: 25902260 PMCID: PMC4406691 DOI: 10.1371/journal.pone.0123106] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/27/2015] [Indexed: 11/19/2022] Open
Abstract
Cell-adhesion molecules of the immunoglobulin superfamily play critical roles in brain development, as well as in maintaining synaptic plasticity, the dysfunction of which is known to cause cognitive impairment. Recently dysfunction of KIRREL3, a synaptic molecule of the immunoglobulin superfamily, has been implicated in several neurodevelopmental conditions including intellectual disability, autism spectrum disorder, and in the neurocognitive delay associated with Jacobsen syndrome. However, the molecular mechanisms of its physiological actions remain largely unknown. Using a yeast two-hybrid screen, we found that the KIRREL3 extracellular domain interacts with brain expressed proteins MAP1B and MYO16 and its intracellular domain can potentially interact with ATP1B1, UFC1, and SHMT2. The interactions were confirmed by co-immunoprecipitation and colocalization analyses of proteins expressed in human embryonic kidney cells, mouse neuronal cells, and rat primary neuronal cells. Furthermore, we show KIRREL3 colocalization with the marker for the Golgi apparatus and synaptic vesicles. Previously, we have shown that KIRREL3 interacts with the X-linked intellectual disability associated synaptic scaffolding protein CASK through its cytoplasmic domain. In addition, we found a genomic deletion encompassing MAP1B in one patient with intellectual disability, microcephaly and seizures and deletions encompassing MYO16 in two unrelated patients with intellectual disability, autism and microcephaly. MAP1B has been previously implicated in synaptogenesis and is involved in the development of the actin-based membrane skeleton. MYO16 is expressed in hippocampal neurons and also indirectly affects actin cytoskeleton through its interaction with WAVE1 complex. We speculate KIRREL3 interacting proteins are potential candidates for intellectual disability and autism spectrum disorder. Moreover, our findings provide further insight into understanding the molecular mechanisms underlying the physiological action of KIRREL3 and its role in neurodevelopment.
Collapse
|
40
|
Moog U, Bierhals T, Brand K, Bautsch J, Biskup S, Brune T, Denecke J, de Die-Smulders CE, Evers C, Hempel M, Henneke M, Yntema H, Menten B, Pietz J, Pfundt R, Schmidtke J, Steinemann D, Stumpel CT, Van Maldergem L, Kutsche K. Phenotypic and molecular insights into CASK-related disorders in males. Orphanet J Rare Dis 2015; 10:44. [PMID: 25886057 PMCID: PMC4449965 DOI: 10.1186/s13023-015-0256-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/20/2015] [Indexed: 12/05/2022] Open
Abstract
Background Heterozygous loss-of-function mutations in the X-linked CASK gene cause progressive microcephaly with pontine and cerebellar hypoplasia (MICPCH) and severe intellectual disability (ID) in females. Different CASK mutations have also been reported in males. The associated phenotypes range from nonsyndromic ID to Ohtahara syndrome with cerebellar hypoplasia. However, the phenotypic spectrum in males has not been systematically evaluated to date. Methods We identified a CASK alteration in 8 novel unrelated male patients by targeted Sanger sequencing, copy number analysis (MLPA and/or FISH) and array CGH. CASK transcripts were investigated by RT-PCR followed by sequencing. Immunoblotting was used to detect CASK protein in patient-derived cells. The clinical phenotype and natural history of the 8 patients and 28 CASK-mutation positive males reported previously were reviewed and correlated with available molecular data. Results CASK alterations include one nonsense mutation, one 5-bp deletion, one mutation of the start codon, and five partial gene deletions and duplications; seven were de novo, including three somatic mosaicisms, and one was familial. In three subjects, specific mRNA junction fragments indicated in tandem duplication of CASK exons disrupting the integrity of the gene. The 5-bp deletion resulted in multiple aberrant CASK mRNAs. In fibroblasts from patients with a CASK loss-of-function mutation, no CASK protein could be detected. Individuals who are mosaic for a severe CASK mutation or carry a hypomorphic mutation still showed detectable amount of protein. Conclusions Based on eight novel patients and all CASK-mutation positive males reported previously three phenotypic groups can be distinguished that represent a clinical continuum: (i) MICPCH with severe epileptic encephalopathy caused by hemizygous loss-of-function mutations, (ii) MICPCH associated with inactivating alterations in the mosaic state or a partly penetrant mutation, and (iii) syndromic/nonsyndromic mild to severe ID with or without nystagmus caused by CASK missense and splice mutations that leave the CASK protein intact but likely alter its function or reduce the amount of normal protein. Our findings facilitate focused testing of the CASK gene and interpreting sequence variants identified by next-generation sequencing in cases with a phenotype resembling either of the three groups. Electronic supplementary material The online version of this article (doi:10.1186/s13023-015-0256-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ute Moog
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Kristina Brand
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Jan Bautsch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | | | - Thomas Brune
- Universitätskinderklinik, Universitätsklinikum Magdeburg, Magdeburg, Germany.
| | - Jonas Denecke
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Neuropädiatrie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| | - Christine E de Die-Smulders
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht UMC+, Maastricht, The Netherlands.
| | - Christina Evers
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Marco Henneke
- Klinik für Kinder- und Jugendmedizin, Universitätsmedizin Göttingen, Göttingen, Germany.
| | - Helger Yntema
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Björn Menten
- Center for Medical Genetics, Ghent University, Ghent, Belgium.
| | - Joachim Pietz
- Section of Neuropediatrics, Center for Child and Adolescent Medicine, Heidelberg, Germany.
| | - Rolph Pfundt
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Jörg Schmidtke
- Institut für Humangenetik, Medizinische Hochschule Hannover, Hannover, Germany.
| | - Doris Steinemann
- Institut für Zell- und Molekularpathologie, Medizinische Hochschule Hannover, Hannover, Germany.
| | - Constance T Stumpel
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht UMC+, Maastricht, The Netherlands.
| | | | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
41
|
Hunter JM, Kiefer J, Balak CD, Jooma S, Ahearn ME, Hall JG, Baumbach-Reardon L. Review of X-linked syndromes with arthrogryposis or early contractures-aid to diagnosis and pathway identification. Am J Med Genet A 2015; 167A:931-73. [DOI: 10.1002/ajmg.a.36934] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/05/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Jesse M. Hunter
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Jeff Kiefer
- Knowledge Mining; Translational Genomics Research Institute; Phoenix Arizona
| | - Christopher D. Balak
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Sonya Jooma
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Mary Ellen Ahearn
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Judith G. Hall
- Departments of Medical Genetics and Pediatrics; University of British Columbia and BC Children's Hospital Vancouver; British Columbia Canada
| | - Lisa Baumbach-Reardon
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| |
Collapse
|
42
|
Rakshambikai R, Manoharan M, Gnanavel M, Srinivasan N. Typical and atypical domain combinations in human protein kinases: functions, disease causing mutations and conservation in other primates. RSC Adv 2015. [DOI: 10.1039/c4ra11685b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A twist in the evolution of human kinases resulting in kinases with hybrid and rogue properties.
Collapse
Affiliation(s)
| | - Malini Manoharan
- Molecular Biophysics Unit
- Indian Institute of Science
- Bangalore 560012
- India
| | - Mutharasu Gnanavel
- Molecular Biophysics Unit
- Indian Institute of Science
- Bangalore 560012
- India
| | | |
Collapse
|
43
|
Novel approaches for targeting kinases: allosteric inhibition, allosteric activation and pseudokinases. Future Med Chem 2014; 6:541-61. [PMID: 24649957 DOI: 10.4155/fmc.13.216] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Protein kinases are involved in many essential cellular processes and their deregulation can lead to a variety of diseases, including cancer. The pharmaceutical industry has invested heavily in the identification of kinase inhibitors to modulate these disease-promoting pathways, resulting in several successful drugs. However, the field is challenging as it is difficult to identify novel selective inhibitors with good pharmacokinetic/pharmacodynamic properties. In addition, resistance to kinase inhibitor treatment frequently arises. The identification of non-ATP site targeting ('allosteric') inhibitors, the identification of kinase activators and the expansion of kinase target space to include the less studied members of the family, including atypical- and pseudo-kinases, are potential avenues to overcome these challenges. In this perspective, the opportunities and challenges of following these approaches and others will be discussed.
Collapse
|
44
|
Malik BR, Hodge JJL. CASK and CaMKII function in Drosophila memory. Front Neurosci 2014; 8:178. [PMID: 25009461 PMCID: PMC4070058 DOI: 10.3389/fnins.2014.00178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/06/2014] [Indexed: 12/04/2022] Open
Abstract
Calcium (Ca2+) and Calmodulin (CaM)-dependent serine/threonine kinase II (CaMKII) plays a central role in synaptic plasticity and memory due to its ability to phosphorylate itself and regulate its own kinase activity. Autophosphorylation at threonine 287 (T287) switches CaMKII to a Ca2+ independent and constitutively active state replicated by overexpression of a phosphomimetic CaMKII-T287D transgene or blocked by expression of a T287A transgene. A second pair of sites, T306 T307 in the CaM binding region once autophosphorylated, prevents CaM binding and inactivates the kinase during synaptic plasticity and memory, and can be blocked by a TT306/7AA transgene. Recently the synaptic scaffolding molecule called CASK (Ca2+/CaM-associated serine kinase) has been shown to control both sets of CaMKII autophosphorylation events during neuronal growth, Ca2+ signaling and memory in Drosophila. Deletion of either full length CASK or just its CaMK-like and L27 domains removed middle-term memory (MTM) and long-term memory (LTM), with CASK function in the α′/ß′ mushroom body neurons being required for memory. In a similar manner directly changing the levels of CaMKII autophosphorylation (T287D, T287A, or TT306/7AA) in the α′/ß′ neurons also removed MTM and LTM. In the CASK null mutant expression of either the Drosophila or human CASK transgene in the α′/ß′ neurons was found to completely rescue memory, confirming that CASK signaling in α′/β′ neurons is necessary and sufficient for Drosophila memory formation and that the neuronal function of CASK is conserved between Drosophila and human. Expression of human CASK in Drosophila also rescued the effect of CASK deletion on the activity state of CaMKII, suggesting that human CASK may also regulate CaMKII autophosphorylation. Mutations in human CASK have recently been shown to result in intellectual disability and neurological defects suggesting a role in plasticity and learning possibly via regulation of CaMKII autophosphorylation.
Collapse
Affiliation(s)
- Bilal R Malik
- School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - James J L Hodge
- School of Physiology and Pharmacology, University of Bristol Bristol, UK
| |
Collapse
|
45
|
LaConte LEW, Chavan V, Mukherjee K. Identification and glycerol-induced correction of misfolding mutations in the X-linked mental retardation gene CASK. PLoS One 2014; 9:e88276. [PMID: 24505460 PMCID: PMC3914952 DOI: 10.1371/journal.pone.0088276] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/09/2014] [Indexed: 11/21/2022] Open
Abstract
The overwhelming amount of available genomic sequence variation information demands a streamlined approach to examine known pathogenic mutations of any given protein. Here we seek to outline a strategy to easily classify pathogenic missense mutations that cause protein misfolding and are thus good candidates for chaperone-based therapeutic strategies, using previously identified mutations in the gene CASK. We applied a battery of bioinformatics algorithms designed to predict potential impact on protein structure to five pathogenic missense mutations in the protein CASK that have been shown to underlie pathologies ranging from X-linked mental retardation to autism spectrum disorder. A successful classification of the mutations as damaging was not consistently achieved despite the known pathogenicity. In addition to the bioinformatics analyses, we performed molecular modeling and phylogenetic comparisons. Finally, we developed a simple high-throughput imaging assay to measure the misfolding propensity of the CASK mutants in situ. Our data suggests that a phylogenetic analysis may be a robust method for predicting structurally damaging mutations in CASK. Mutations in two evolutionarily invariant residues (Y728C and W919R) exhibited a strong propensity to misfold and form visible aggregates in the cytosolic milieu. The remaining mutations (R28L, Y268H, and P396S) showed no evidence of aggregation and maintained their interactions with known CASK binding partners liprin-α3 Mint-1, and Veli, indicating an intact structure. Intriguingly, the protein aggregation caused by the Y728C and W919R mutations was reversed by treating the cells with a chemical chaperone (glycerol), providing a possible therapeutic strategy for treating structural mutations in CASK in the future.
Collapse
Affiliation(s)
- Leslie E. W. LaConte
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, United States of America
| | - Vrushali Chavan
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, United States of America
| | - Konark Mukherjee
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, United States of America
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
46
|
Gillespie JM, Hodge JJL. CASK regulates CaMKII autophosphorylation in neuronal growth, calcium signaling, and learning. Front Mol Neurosci 2013; 6:27. [PMID: 24062638 PMCID: PMC3769642 DOI: 10.3389/fnmol.2013.00027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/24/2013] [Indexed: 11/27/2022] Open
Abstract
Calcium (Ca2+)/calmodulin (CaM)-dependent kinase II (CaMKII) activity plays a fundamental role in learning and memory. A key feature of CaMKII in memory formation is its ability to be regulated by autophosphorylation, which switches its activity on and off during synaptic plasticity. The synaptic scaffolding protein CASK (calcium (Ca2+)/calmodulin (CaM) associated serine kinase) is also important for learning and memory, as mutations in CASK result in intellectual disability and neurological defects in humans. We show that in Drosophila larvae, CASK interacts with CaMKII to control neuronal growth and calcium signaling. Furthermore, deletion of the CaMK-like and L27 domains of CASK (CASK β null) or expression of overactive CaMKII (T287D) produced similar effects on synaptic growth and Ca2+ signaling. CASK overexpression rescues the effects of CaMKII overactivity, consistent with the notion that CASK and CaMKII act in a common pathway that controls these neuronal processes. The reduction in Ca2+ signaling observed in the CASK β null mutant caused a decrease in vesicle trafficking at synapses. In addition, the decrease in Ca2+ signaling in CASK mutants was associated with an increase in Ether-à-go-go (EAG) potassium (K+) channel localization to synapses. Reducing EAG restored the decrease in Ca2+ signaling observed in CASK mutants to the level of wildtype, suggesting that CASK regulates Ca2+ signaling via EAG. CASK knockdown reduced both appetitive associative learning and odor evoked Ca2+ responses in Drosophila mushroom bodies, which are the learning centers of Drosophila. Expression of human CASK in Drosophila rescued the effect of CASK deletion on the activity state of CaMKII, suggesting that human CASK may also regulate CaMKII autophosphorylation.
Collapse
Affiliation(s)
- John M Gillespie
- School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | | |
Collapse
|
47
|
Abstract
CASK (Ca2+/calmodulin-activated serine kinase) is a synaptic protein that interacts with the cytosolic tail of adhesion molecules such as neurexins, syncam and syndecans. It belongs to the MAGUK (membrane-associated guanylate kinase) family of scaffolding proteins which are known to decorate cell–cell junctions. CASK is an essential gene in mammals, critical for neurodevelopment. Mutations in the CASK gene in humans result in phenotypes that range from intellectual disability to lethality. Despite its importance, CASK has a single genetic isoform located in the short arm of the X chromosome near an evolutionary breakpoint. Surprisingly, CASK is a non-essential gene in invertebrates and displays functional divergence. In the present article, we describe the phylogenetic differences in existing CASK orthologues. The CASK gene has undergone a huge expansion in size (~55-fold). Almost all of this expansion is a direct result of an increase in the size of the introns. The coding region of CASK orthologues, and hence the protein, exhibit a high degree of evolutionary conservation. Within the protein, domain arrangement is completely conserved and substitution rates are higher in the connecting loop regions [L27 (Lin2, Lin7)] than within the domain. Our analyses of single residue substitutions and genotype–phenotype relationships suggest that, other than intronic expansion, the dramatic functional changes of CASK are driven by subtle (non-radical) primary structure changes within the CASK protein and concomitant changes in its protein interactors.
Collapse
|
48
|
Malik BR, Gillespie JM, Hodge JJL. CASK and CaMKII function in the mushroom body α'/β' neurons during Drosophila memory formation. Front Neural Circuits 2013; 7:52. [PMID: 23543616 PMCID: PMC3608901 DOI: 10.3389/fncir.2013.00052] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/09/2013] [Indexed: 11/13/2022] Open
Abstract
Ca(2+)/CaM serine/threonine kinase II (CaMKII) is a central molecule in mechanisms of synaptic plasticity and memory. A vital feature of CaMKII in plasticity is its ability to switch to a calcium (Ca(2+)) independent constitutively active state after autophosphorylation at threonine 287 (T287). A second pair of sites, T306 T307 in the calmodulin (CaM) binding region once autophosphorylated, prevent subsequent CaM binding and inactivates the kinase during synaptic plasticity and memory. Recently a synaptic molecule called Ca(2+)/CaM-dependent serine protein kinase (CASK) has been shown to control both sets of CaMKII autophosphorylation events and hence is well poised to be a key regulator of memory. We show deletion of full length CASK or just its CaMK-like and L27 domains disrupts middle-term memory (MTM) and long-term memory (LTM), with CASK function in the α'/β' subset of mushroom body neurons being required for memory. Likewise directly changing the levels of CaMKII autophosphorylation in these neurons removed MTM and LTM. The requirement of CASK and CaMKII autophosphorylation was not developmental as their manipulation just in the adult α'/β' neurons was sufficient to remove memory. Overexpression of CASK or CaMKII in the α'/β' neurons also occluded MTM and LTM. Overexpression of either Drosophila or human CASK in the α'/β' neurons of the CASK mutant completely rescued memory, confirming that CASK signaling in α'/β' neurons is necessary and sufficient for Drosophila memory formation and that the neuronal function of CASK is conserved between Drosophila and human. At the cellular level CaMKII overexpression in the α'/β' neurons increased activity dependent Ca(2+) responses while reduction of CaMKII decreased it. Likewise reducing CASK or directly expressing a phosphomimetic CaMKII T287D transgene in the α'/β' similarly decreased Ca(2+) signaling. Our results are consistent with CASK regulating CaMKII autophosphorylation in a pathway required for memory formation that involves activity dependent changes in Ca(2+) signaling in the α'/β' neurons.
Collapse
Affiliation(s)
- Bilal R Malik
- School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | | | | |
Collapse
|
49
|
Takanashi JI, Okamoto N, Yamamoto Y, Hayashi S, Arai H, Takahashi Y, Maruyama K, Mizuno S, Shimakawa S, Ono H, Oyanagi R, Kubo S, Barkovich AJ, Inazawa J. Clinical and radiological features of Japanese patients with a severe phenotype due to CASK mutations. Am J Med Genet A 2012; 158A:3112-8. [PMID: 23165780 DOI: 10.1002/ajmg.a.35640] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 08/05/2012] [Indexed: 11/11/2022]
Abstract
Heterozygous loss of function mutations of CASK at Xp11.4 in females cause severe intellectual disability (ID) and microcephaly with pontine and cerebellar hypoplasia (MICPCH). However, the longitudinal clinical and radiological course of affected patients, including patterns of postnatal growth, has not been described. Neurodevelopmental and imaging information was retrospectively accrued for 16 Japanese (15 female and 1 male) patients with ID and MICPCH associated with CASK mutations. All records were analyzed; patient age ranged from 2 to 16 years at the time of the most recent examinations. The growth pattern, neurological development, neurological signs/symptoms, and facial features were similar in the 15 female patients. Their head circumference at birth was within the normal range in about half, and their height and weight were frequently normal. This was followed by early development of severe microcephaly and postnatal growth retardation. The patients acquired head control almost normally between 3 and 6 months, followed by motor delay. More than half of the female patients had epilepsy. Their MRIs showed microcephaly, brainstem, and cerebellar hypoplasia in early infancy, and a normal or large appearing corpus callosum. The male patient showed a more severe clinical phenotype. These uniform clinical and radiological features should facilitate an early diagnosis and be useful for medical care of females with ID and MICPCH associated with CASK mutations.
Collapse
|
50
|
Zhu J, Shang Y, Chen J, Zhang M. Structure and function of the guanylate kinase-like domain of the MAGUK family scaffold proteins. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s11515-012-1244-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|