1
|
Qi H, Pan D, Zhang Y, Zhu Y, Zhang X, Fu T. NEXMIF Combined with KIDINS220 Gene Mutation Caused Neurodevelopmental Disorder and Epilepsy: One Case Report. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:588-594. [PMID: 39129698 PMCID: PMC11319755 DOI: 10.62641/aep.v52i4.1625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
of Medical History: A male infant, 8 months old, was admitted to hospital with cough and fever. The clinical symptoms were found to be mental retardation, obesity, dystonia, movement limitation, and visual retardation. Early development was normal, but after 6 months, the child developed upright head instability, difficulty grasping, and seizures. Symptoms and Signs: The child presents with mental retardation, obesity, increased muscle tone, motor dysfunction, visual impairment, and seizures. DIAGNOSIS A whole exon test was performed to detect a neurite extension and migration factor (NEXMIF) gene mutation (NM_001008537.2: c.1042C > T (p. Arg348*)), which is known to be associated with intellectual disability and neurological symptoms. In addition, the test revealed a mutation in the Kinase D interacting substrate of 220 kDa (KIDINS220) gene (NM_020738.2: c.3242_3243insC (p. Leu1082AIafs*5)) with a heterozygous mutation in the father and wild type in the mother. TREATMENT The patient was treated with anti-infection, aerosol inhalation, calcium supplement, and anti-epileptic drugs (levetiracetam), and the disease was controlled. Home and hospital rehabilitation is also underway. CLINICAL OUTCOME The condition of the child improved after treatment and no seizures occurred again. The patient needs continuous rehabilitation treatment and follow-up observation. CONCLUSION For male children with unexplained neurodevelopmental disorders and comorbidities such as obesity, dystonia, and seizures, mutations in related genes such as NEXMIF should be considered. Clinical practice should improve genetic testing as early as possible to provide a basis for genetic counseling.
Collapse
Affiliation(s)
- Hongli Qi
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| | - Dongju Pan
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| | - Ying Zhang
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| | - Yunhui Zhu
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| | - Xie Zhang
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| | - Tingting Fu
- Department of Pediatrics, Pu'er People's Hospital, 665000 Pu'er, Yunnan, China
| |
Collapse
|
2
|
Ray NR, Kunkle BW, Hamilton‐Nelson K, Kurup JT, Rajabli F, Qiao M, Vardarajan BN, Cosacak MI, Kizil C, Jean‐Francois M, Cuccaro M, Reyes‐Dumeyer D, Cantwell L, Kuzma A, Vance JM, Gao S, Hendrie HC, Baiyewu O, Ogunniyi A, Akinyemi RO, Lee W, Martin ER, Wang L, Beecham GW, Bush WS, Xu W, Jin F, Wang L, Farrer LA, Haines JL, Byrd GS, Schellenberg GD, Mayeux R, Pericak‐Vance MA, Reitz C. Extended genome-wide association study employing the African genome resources panel identifies novel susceptibility loci for Alzheimer's disease in individuals of African ancestry. Alzheimers Dement 2024; 20:5247-5261. [PMID: 38958117 PMCID: PMC11350055 DOI: 10.1002/alz.13880] [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: 09/20/2023] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Despite a two-fold risk, individuals of African ancestry have been underrepresented in Alzheimer's disease (AD) genomics efforts. METHODS Genome-wide association studies (GWAS) of 2,903 AD cases and 6,265 controls of African ancestry. Within-dataset results were meta-analyzed, followed by functional genomics analyses. RESULTS A novel AD-risk locus was identified in MPDZ on chromosome (chr) 9p23 (rs141610415, MAF = 0.002, p = 3.68×10-9). Two additional novel common and nine rare loci were identified with suggestive associations (P < 9×10-7). Comparison of association and linkage disequilibrium (LD) patterns between datasets with higher and lower degrees of African ancestry showed differential association patterns at chr12q23.2 (ASCL1), suggesting that this association is modulated by regional origin of local African ancestry. DISCUSSION These analyses identified novel AD-associated loci in individuals of African ancestry and suggest that degree of African ancestry modulates some associations. Increased sample sets covering as much African genetic diversity as possible will be critical to identify additional loci and deconvolute local genetic ancestry effects. HIGHLIGHTS Genetic ancestry significantly impacts risk of Alzheimer's Disease (AD). Although individuals of African ancestry are twice as likely to develop AD, they are vastly underrepresented in AD genomics studies. The Alzheimer's Disease Genetics Consortium has previously identified 16 common and rare genetic loci associated with AD in African American individuals. The current analyses significantly expand this effort by increasing the sample size and extending ancestral diversity by including populations from continental Africa. Single variant meta-analysis identified a novel genome-wide significant AD-risk locus in individuals of African ancestry at the MPDZ gene, and 11 additional novel loci with suggestive genome-wide significance at p < 9×10-7. Comparison of African American datasets with samples of higher degree of African ancestry demonstrated differing patterns of association and linkage disequilibrium at one of these loci, suggesting that degree and/or geographic origin of African ancestry modulates the effect at this locus. These findings illustrate the importance of increasing number and ancestral diversity of African ancestry samples in AD genomics studies to fully disentangle the genetic architecture underlying AD, and yield more effective ancestry-informed genetic screening tools and therapeutic interventions.
Collapse
Grants
- P30 AG013854 NIA NIH HHS
- International Parkinson Fonds
- P50 MH060451 NIMH NIH HHS
- P30 AG066444 NIA NIH HHS
- R01 AG28786-01A1 North Carolina A&T University
- U01AG46161 NIA NIH HHS
- AG05128 Duke University
- Medical Research Council
- U01AG057659 NIH HHS
- R01 AG022374 NIA NIH HHS
- U19 AG074865 NIA NIH HHS
- P50 AG023501 NIA NIH HHS
- U01 AG046152 NIA NIH HHS
- P30 AG010124 NIA NIH HHS
- U01 HG006375 NHGRI NIH HHS
- Biogen
- U01 AG058654 NIA NIH HHS
- NIMH MH60451 NINDS NIH HHS
- U54 AG052427 NIA NIH HHS
- P30 AG066518 NIA NIH HHS
- UO1 HG004610 Group Health Research Institute
- RC2 AG036528 NIA NIH HHS
- P30 AG028377 NIA NIH HHS
- R01AG048927 NIH HHS
- UO1 HG006375 Group Health Research Institute
- R01 AG22018 Rush University
- U01AG46152 NIA NIH HHS
- P50 AG008671 NIA NIH HHS
- P30 AG10133 Indiana University
- P50 AG005142 NIA NIH HHS
- U01 AG10483 Boston University
- Higher Education Funding Council for England
- R01 AG035137 NIA NIH HHS
- R01 AG009029 NIA NIH HHS
- P50 AG005131 NIA NIH HHS
- P50 AG005128 NIA NIH HHS
- P30 AG010133 NIA NIH HHS
- U24 AG021886 NIA NIH HHS
- R01 AG031581 NIA NIH HHS
- 5R01AG012101 New York University
- R01 AG009956 NIA NIH HHS
- P50 AG016574 NIA NIH HHS
- P50 AG005146 NIA NIH HHS
- U01AG058654 NIH HHS
- AG025688 Emory University
- P30AG10161 NIA NIH HHS
- Alzheimer's Drug Discovery Foundation
- U01 AG061356 NIA NIH HHS
- RC2 AG036650 NIA NIH HHS
- Servier
- Janssen Alzheimer Immunotherapy Research & Development, LLC.
- U01 AG032984 NIA NIH HHS
- U01 HG008657 NHGRI NIH HHS
- Brain Net Europe
- R01 AG019085 NIA NIH HHS
- Lumosity
- R01 AG013616 NIA NIH HHS
- U01 AG024904 NIA NIH HHS
- Translational Genomics Research Institute
- P50 AG008702 NIA NIH HHS
- Bristol-Myers Squibb Company
- R01 AG030146 NIA NIH HHS
- R01AG041797 NIA FBS (Columbia University)
- U01 AG072579 NIA NIH HHS
- Piramal Imaging
- DeNDRoN
- UL1 RR029893 NCRR NIH HHS
- Takeda Pharmaceutical Company
- 1R01AG035137 New York University
- R01 AG15819 Rush University
- R01AG30146 NIA NIH HHS
- R01AG15819 NIA NIH HHS
- P50 NS039764 NINDS NIH HHS
- P01 AG003991 NIA NIH HHS
- Office of Research and Development
- Genentech, Inc.
- U01 AG016976 NIA NIH HHS
- US Department of Veterans Affairs Administration
- P30 AG008051 NIA NIH HHS
- P50 AG005681 NIA NIH HHS
- P30 AG013846 NIA NIH HHS
- U24 AG056270 NIA NIH HHS
- RC2 AG036502 NIA NIH HHS
- P01 AG026276 NIA NIH HHS
- R01 AG017917 NIA NIH HHS
- Araclon Biotech
- U01 AG057659 NIA NIH HHS
- R01 MH080295 NIMH NIH HHS
- Hersenstichting Nederland Breinbrekend Werk
- R01 AG026390 NIA NIH HHS
- R01 AG028786 NIA NIH HHS
- KL2 RR024151 NCRR NIH HHS
- Internationale Stiching Alzheimer Onderzoek
- P30AG066462 NIH HHS
- U24 AG026390 NIA FBS (Columbia University)
- Novartis Pharmaceuticals Corporation
- P50 AG005136 NIA NIH HHS
- Meso Scale Diagnostics, LLC.
- CereSpir, Inc.
- P30 AG012300 NIA NIH HHS
- P01 AG03991 University of Washington
- RF1AG059018 NIH HHS
- Canadian Institute of Health Research
- RF1 AG059018 NIA NIH HHS
- BioClinica, Inc.
- U01 AG062943 NIA NIH HHS
- R01 AG012101 NIA NIH HHS
- GE Healthcare
- P50 AG016573 NIA NIH HHS
- U24 AG21886 National Cell Repository for Alzheimer's Disease (NCRAD)
- P50 AG016570 NIA NIH HHS
- P50 AG005134 NIA NIH HHS
- P30 AG066462 NIA NIH HHS
- Stichting MS Research
- P30 AG008017 NIA NIH HHS
- R01AG33193 Boston University
- Howard Hughes Medical Institute
- R01 AG042437 NIA NIH HHS
- U24 AG041689 NIA NIH HHS
- P01 AG019724 NIA NIH HHS
- R01AG36042 NIA NIH HHS
- RC2AG036547 NIA NIH HHS
- R01 AG036042 NIA NIH HHS
- P30 AG010161 NIA NIH HHS
- AG019757 University of Miami
- Kronos Science
- P30 AG08051 New York University
- IIRG-05-14147 Alzheimer's Association
- AG010491 University of Miami
- R01 AG033193 NIA NIH HHS
- P50 AG025688 NIA NIH HHS
- IIRG-08-89720 Alzheimer's Association
- AbbVie
- R37 AG015473 NIA NIH HHS
- U24 AG026395 NIA NIH HHS
- R01 AG032990 NIA NIH HHS
- North Bristol NHS Trust Research and Innovation Department
- AG021547 University of Miami
- R01 AG01101 Rush University
- Transition Therapeutics
- R01 AG072547 NIA NIH HHS
- AG027944 University of Miami
- AG041232 NIA NIH HHS
- A2111048 BrightFocus Foundation
- U01 AG052410 NIA NIH HHS
- Johnson & Johnson Pharmaceutical Research & Development LLC.
- R01 CA129769 NCI NIH HHS
- P50 AG005133 NIA NIH HHS
- U01 AG010483 NIA NIH HHS
- UO1 AG006781 Group Health Research Institute
- Merck & Co., Inc.
- U01AG32984 NIA NIH HHS
- U01 AG024904 NIH HHS
- RC2 AG036547 NIA NIH HHS
- P01 AG002219 NIA NIH HHS
- R01 AG17917 Rush University
- U01 AG006781 NIA NIH HHS
- R01 AG041797 NIA NIH HHS
- NIBIB NIH HHS
- P01 AG010491 NIA NIH HHS
- P50 AG005144 NIA NIH HHS
- U01AG062943 NIH HHS
- R01 AG064614 NIA NIH HHS
- Glaxo Smith Kline
- U01AG072579 NIH HHS
- Biomedical Laboratory Research Program
- U19AG074865 NIH HHS
- R01 AG048927 NIA NIH HHS
- RF1 AG057473 NIA NIH HHS
- R01 AG037212 NIA NIH HHS
- R01 AG022018 NIA NIH HHS
- U24AG056270 NIH HHS
- R01 AG021547 NIA NIH HHS
- R01 AG041232 NIA NIH HHS
- P50 AG005138 NIA NIH HHS
- RF1AG57473 NIA NIH HHS
- R01 AG019757 NIA NIH HHS
- R01 AG020688 NIA NIH HHS
- AG07562 University of Pittsburgh
- R01AG072547 NIH HHS
- Alzheimer's Research Trust
- Pfizer Inc.
- Illinois Department of Public Health
- Elan Pharmaceuticals, Inc.
- NHS trusts
- R01 AG030653 NIA NIH HHS
- AG052410 NIA NIH HHS
- P20 MD000546 NIMHD NIH HHS
- R01 AG027944 NIA NIH HHS
- Eli Lilly and Company
- R01 AG017173 NIA NIH HHS
- R01 AG025259 NIA NIH HHS
- U01 HG004610 NHGRI NIH HHS
- U24-AG041689 University of Pennsylvania
- P30 AG010129 NIA NIH HHS
- U01 AG046161 NIA NIH HHS
- Wellcome Trust
- P30 AG019610 NIA NIH HHS
- IXICO Ltd.
- P50 AG016582 NIA NIH HHS
- R01 AG048015 NIA NIH HHS
- NeuroRx Research
- R01AG17917 NIA NIH HHS
- U01AG61356 NIA NIH HHS
- R01AG36836 NIA NIH HHS
- 5R01AG022374 New York University
- EuroImmun; F. Hoffmann-La Roche Ltd
- R01 AG041718 NIA NIH HHS
- 1RC2AG036502 New York University
- Newcastle University
- AG041718 University of Pittsburgh
- P30 AG028383 NIA NIH HHS
- AG05144 University of Kentucky
- AG030653 University of Pittsburgh
- R01AG48015 NIA NIH HHS
- R01 AG026916 NIA NIH HHS
- P50 AG033514 NIA NIH HHS
- R01 NS059873 NINDS NIH HHS
- # NS39764 NINDS NIH HHS
- ADGC National Institutes of Health, National Institute on Aging (NIH-NIA)
- Neurotrack Technologies
- Fujirebio
- Lundbeck
- MP-V BrightFocus Foundation
- BRACE
- R01 AG015819 NIA NIH HHS
- R01 AG036836 NIA NIH HHS
- Eisai Inc.
- 5R01AG013616 New York University
- W81XWH-12-2-0012 Department of Defense
- R01AG064614 NIH HHS
- AG02365 University of Pittsburgh
- NIH
- University of Pennsylvania
- NACC
- Boston University
- Columbia University
- Duke University
- Emory University
- Indiana University
- Johns Hopkins University
- Massachusetts General Hospital
- Mayo Clinic
- New York University
- Northwestern University
- Oregon Health & Science University
- Rush University
- NIA
- University of Alabama at Birmingham
- University of Arizona
- University of California, Davis
- University of California, Irvine
- University of California, Los Angeles
- University of California, San Diego
- University of California, San Francisco
- University of Kentucky
- University of Michigan
- University of Pittsburgh
- University of Southern California
- University of Miami
- University of Washington
- Vanderbilt University
- NINDS
- Alzheimer's Association
- Office of Research and Development
- BrightFocus Foundation
- Wellcome Trust
- Howard Hughes Medical Institute
- Medical Research Council
- Newcastle University
- Higher Education Funding Council for England
- Alzheimer's Research Trust
- BRACE
- Stichting MS Research
- Department of Defense
- National Institute of Biomedical Imaging and Bioengineering
- AbbVie
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Biogen
- Bristol‐Myers Squibb Company
- Eli Lilly and Company
- Genentech, Inc.
- Fujirebio
- GE Healthcare
- Lundbeck
- Merck & Co., Inc.
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Servier
- Takeda Pharmaceutical Company
- Illinois Department of Public Health
- Translational Genomics Research Institute
Collapse
|
3
|
Yao D, Binan L, Bezney J, Simonton B, Freedman J, Frangieh CJ, Dey K, Geiger-Schuller K, Eraslan B, Gusev A, Regev A, Cleary B. Scalable genetic screening for regulatory circuits using compressed Perturb-seq. Nat Biotechnol 2024; 42:1282-1295. [PMID: 37872410 PMCID: PMC11035494 DOI: 10.1038/s41587-023-01964-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/22/2023] [Indexed: 10/25/2023]
Abstract
Pooled CRISPR screens with single-cell RNA sequencing readout (Perturb-seq) have emerged as a key technique in functional genomics, but they are limited in scale by cost and combinatorial complexity. In this study, we modified the design of Perturb-seq by incorporating algorithms applied to random, low-dimensional observations. Compressed Perturb-seq measures multiple random perturbations per cell or multiple cells per droplet and computationally decompresses these measurements by leveraging the sparse structure of regulatory circuits. Applied to 598 genes in the immune response to bacterial lipopolysaccharide, compressed Perturb-seq achieves the same accuracy as conventional Perturb-seq with an order of magnitude cost reduction and greater power to learn genetic interactions. We identified known and novel regulators of immune responses and uncovered evolutionarily constrained genes with downstream targets enriched for immune disease heritability, including many missed by existing genome-wide association studies. Our framework enables new scales of interrogation for a foundational method in functional genomics.
Collapse
Affiliation(s)
- Douglas Yao
- Program in Systems, Synthetic, and Quantitative Biology, Harvard University, Cambridge, MA, USA
| | - Loic Binan
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jon Bezney
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke Simonton
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jahanara Freedman
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Chris J Frangieh
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kushal Dey
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Alexander Gusev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Genentech, South San Francisco, CA, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA, USA.
- Department of Biology, Boston University, Boston, MA, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Program in Bioinformatics, Boston University, Boston, MA, USA.
- Biological Design Center, Boston University, Boston, MA, USA.
| |
Collapse
|
4
|
Alstrup M, Cesca F, Krawczun-Rygmaczewska A, López-Menéndez C, Pose-Utrilla J, Castberg FC, Bjerager MO, Finnila C, Kruer MC, Bakhtiari S, Padilla-Lopez S, Manwaring L, Keren B, Afenjar A, Galatolo D, Scalise R, Santorelli FM, Shillington A, Vezain M, Martinovic J, Stevens C, Gowda VK, Srinivasan VM, Thiffault I, Pastinen T, Baranano K, Lee A, Granadillo J, Glassford MR, Keegan CE, Matthews N, Saugier-Veber P, Iglesias T, Østergaard E. Refining the phenotype of SINO syndrome: A comprehensive cohort report of 14 novel cases. Genet Med 2024; 26:101219. [PMID: 39033379 DOI: 10.1016/j.gim.2024.101219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
PURPOSE Spastic paraplegia, intellectual disability, nystagmus, and obesity syndrome (SINO) is a rare autosomal dominant condition caused by heterozygous variants in KIDINS220. A total of 12 individuals are reported, comprising 8 with SINO and 4 with an autosomal recessive condition attributed to biallelic KIDINS220 variants. METHODS In our international cohort, we have included 14 individuals, carrying 13 novel pathogenic KIDINS220 variants in heterozygous form. We assessed the clinical and molecular data of our cohort and previously reported individuals and, based on functional experiments, reached a better understanding of the pathogenesis behind the KIDINS220-related disease. RESULTS Using fetal tissue and in vitro assays, we demonstrate that the variants generate KIDINS220 truncated forms that mislocalize in punctate intracellular structures, with decreased levels of the full-length protein, suggesting a trans-dominant negative effect. A total of 92% had their diagnosis within 3 years, with symptoms of developmental delay, spasticity, hypotonia, lack of eye contact, and nystagmus. We identified a KIDINS220 variant associated with fetal hydrocephalus and show that 58% of examined individuals present brain ventricular dilatation. We extend the phenotypic spectrum of SINO syndrome to behavioral manifestations not previously highlighted. CONCLUSION Our study provides further insights into the clinical spectrum, etiology, and predicted functional impact of KIDINS220 variants.
Collapse
Affiliation(s)
- Morten Alstrup
- Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Fabrizia Cesca
- Department of Life Sciences, University of Trieste, Trieste, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy.
| | - Alicja Krawczun-Rygmaczewska
- Department of Life Sciences, University of Trieste, Trieste, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Celia López-Menéndez
- Instituto de Investigaciones Biomédicas Sols-Morreale. Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III. Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas Sols-Morreale. Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III. Madrid, Spain
| | - Filip Christian Castberg
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics, North Zealand Hospital, Hilleroed, Denmark
| | | | | | - Michael C Kruer
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ; Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | - Somayeh Bakhtiari
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ; Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | - Sergio Padilla-Lopez
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ; Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | - Linda Manwaring
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Boris Keren
- Département de génétique, AP-HP.Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alexandra Afenjar
- APHP. Sorbonne Université, Centre de Référence Malformations et maladies congénitales du cervelet et déficiences intellectuelles de causes rares, UF de génétique clinique, Hôpital Trousseau, Paris, France
| | - Daniele Galatolo
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Roberta Scalise
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Fillippo M Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Amelle Shillington
- Department of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| | - Myriam Vezain
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, France
| | - Jelena Martinovic
- Department of Fetal Pathology, AP-HP Antoine Beclere Hospital, University Paris Saclay, Clamart, France
| | - Cathy Stevens
- Department of Pediatrics, University of Tennessee College of Medicine, Chattanooga, TN
| | - Vykuntaraju K Gowda
- Department of Pediatric Neurology, Indira Gandhi institute of child health, Bangalore, India
| | | | - Isabelle Thiffault
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO; University of Missouri Kansas City School of Medicine, Kansas City, MO; Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO
| | - Tomi Pastinen
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO; University of Missouri Kansas City School of Medicine, Kansas City, MO
| | - Kristin Baranano
- Johns Hopkins University, The Johns Hopkins Hospital, Baltimore, MD
| | - Angela Lee
- Department of Pediatrics, Division of Genetics and Genomics, Washington University, Saint Louis, MO
| | - Jorge Granadillo
- Department of Pediatrics, Division of Genetics and Genomics, Washington University, Saint Louis, MO
| | - Megan R Glassford
- Department of Pediatrics, Division of Genetics, Metabolism, and Genomic Medicine, University of Michigan, Ann Arbor, MI; Department of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Catherine E Keegan
- Department of Pediatrics, Division of Genetics, Metabolism, and Genomic Medicine, University of Michigan, Ann Arbor, MI
| | - Nicole Matthews
- WVU Medicine Children's Hospital, Division of Genetics, Morgantown, WV
| | - Pascale Saugier-Veber
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, France; CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Rouen, France
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas Sols-Morreale. Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III. Madrid, Spain.
| | - Elsebet Østergaard
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Herr LA, Fiala GJ, Sagar, Schaffer AM, Hummel JF, Zintchenko M, Raute K, Velasco Cárdenas RMH, Heizmann B, Ebert K, Fehrenbach K, Janowska I, Chan S, Tanriver Y, Minguet S, Schamel WW. Kidins220 and Aiolos promote thymic iNKT cell development by reducing TCR signals. SCIENCE ADVANCES 2024; 10:eadj2802. [PMID: 38489359 PMCID: PMC10942104 DOI: 10.1126/sciadv.adj2802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
Development of T cells is controlled by the signal strength of the TCR. The scaffold protein kinase D-interacting substrate of 220 kilodalton (Kidins220) binds to the TCR; however, its role in T cell development was unknown. Here, we show that T cell-specific Kidins220 knockout (T-KO) mice have strongly reduced invariant natural killer T (iNKT) cell numbers and modest decreases in conventional T cells. Enhanced apoptosis due to increased TCR signaling in T-KO iNKT thymocytes of developmental stages 2 and 3 shows that Kidins220 down-regulates TCR signaling at these stages. scRNA-seq indicated that the transcription factor Aiolos is down-regulated in Kidins220-deficient iNKT cells. Analysis of an Aiolos KO demonstrated that Aiolos is a downstream effector of Kidins220 during iNKT cell development. In the periphery, T-KO iNKT cells show reduced TCR signaling upon stimulation with α-galactosylceramide, suggesting that Kidins220 promotes TCR signaling in peripheral iNKT cells. Thus, Kidins220 reduces or promotes signaling dependent on the iNKT cell developmental stage.
Collapse
Affiliation(s)
- Laurenz A. Herr
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Gina J. Fiala
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Maria Schaffer
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Jonas F. Hummel
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Marina Zintchenko
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Raute
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Rubí M.-H. Velasco Cárdenas
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Beate Heizmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Karolina Ebert
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Kerstin Fehrenbach
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Iga Janowska
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Yakup Tanriver
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
- Department of Medicine IV: Nephrology and Primary Care, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Wolfgang W. Schamel
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Zhang J, Zhang Y, Shang Q, Cheng Y, Su Y, Zhang J, Wang T, Ding J, Li Y, Xie Y, Xing Q. Gain-of-Function KIDINS220 Variants Disrupt Neuronal Development and Cause Cerebral Palsy. Mov Disord 2024; 39:498-509. [PMID: 38148610 DOI: 10.1002/mds.29694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Kinase D-interacting substrate of 220 kDa (KIDINS220) is a multifunctional scaffolding protein essential for neuronal development. It has been implicated in neurological diseases with either autosomal dominant (AD) or autosomal recessive (AR) inheritance patterns. The molecular mechanisms underlying the AR/AD dual nature of KIDINS220 remain elusive, posing challenges to genetic interpretation and clinical interventions. Moreover, increased KIDINS220 exhibited neurotoxicity, but its role in neurodevelopment remains unclear. OBJECTIVE The aim was to investigate the genotype-phenotype correlations of KIDINS220 and elucidate its pathophysiological role in neuronal development. METHODS Whole-exome sequencing was performed in a four-generation family with cerebral palsy. CRISPR/Cas9 was used to generate KIDINS220 mutant cell lines. In utero electroporation was employed to investigate the effect of KIDINS220 variants on neurogenesis in vivo. RESULTS We identified in KIDINS220 a pathogenic nonsense variant (c.4177C > T, p.Q1393*) that associated with AD cerebral palsy. We demonstrated that the nonsense variants located in the terminal exon of KIDINS220 are gain-of-function (GoF) variants, which enable the mRNA to escape nonsense-mediated decay and produce a truncated yet functional KIDINS220 protein. The truncated protein exhibited significant resistance to calpain and consequently accumulated within cells, resulting in the hyperactivation of Rac1 and defects in neuronal development. CONCLUSIONS Our findings demonstrate that the location of variants within KIDINS220 plays a crucial role in determining inheritance patterns and corresponding clinical outcomes. The proposed interaction between Rac1 and KIDINS220 provides new insights into the pathogenesis of cerebral palsy, implying potential therapeutic perspectives. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jin Zhang
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Yandong Zhang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qing Shang
- Department of Pediatric Rehabilitation Medicine, Children's Hospital of Zhengzhou University and Henan Children's Hospital, Zhengzhou, China
| | - Ye Cheng
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Yu Su
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Junjie Zhang
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Ting Wang
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Jian Ding
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Yunqian Li
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| | - Yunli Xie
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qinghe Xing
- Children's Hospital of Fudan University and Institutes of Biomedical Sciences of Fudan University, Shanghai, China
| |
Collapse
|
7
|
Mušálková D, Přistoupilová A, Jedličková I, Hartmannová H, Trešlová H, Nosková L, Hodaňová K, Bittmanová P, Stránecký V, Jiřička V, Langmajerová M, Woodbury‐Smith M, Zarrei M, Trost B, Scherer SW, Bleyer AJ, Vevera J, Kmoch S. Increased burden of rare protein-truncating variants in constrained, brain-specific and synaptic genes in extremely impulsively violent males with antisocial personality disorder. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12882. [PMID: 38359179 PMCID: PMC10869132 DOI: 10.1111/gbb.12882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 02/17/2024]
Abstract
The genetic correlates of extreme impulsive violence are poorly understood, and there have been few studies that have characterized a large group of affected individuals both clinically and genetically. We performed whole exome sequencing (WES) in 290 males with the life-course-persistent, extremely impulsively violent form of antisocial personality disorder (APD) and analyzed the spectrum of rare protein-truncating variants (rPTVs). Comparisons were made with 314 male controls and publicly available genotype data. Functional annotation tools were used for biological interpretation. Participants were significantly more likely to harbor rPTVs in genes that are intolerant to loss-of-function variants (odds ratio [OR] 2.06; p < 0.001), specifically expressed in brain (OR 2.80; p = 0.036) and enriched for those involved in neurotransmitter transport and synaptic processes. In 60 individuals (20%), we identified rPTVs that we classified as clinically relevant based on their clinical associations, biological function and gene expression patterns. Of these, 37 individuals harbored rPTVs in 23 genes that are associated with a monogenic neurological disorder, and 23 individuals harbored rPTVs in 20 genes reportedly intolerant to loss-of-function variants. The analysis presents evidence in support of a model where presence of either one or several private, functionally relevant mutations contribute significantly to individual risk of life-course-persistent APD and reveals multiple individuals who could be affected by clinically unrecognized neuropsychiatric Mendelian disease. Thus, Mendelian diseases and increased rPTV burden may represent important factors for the development of extremely impulsive violent life-course-persistent forms of APD irrespective of their clinical presentation.
Collapse
Affiliation(s)
- Dita Mušálková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Anna Přistoupilová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Ivana Jedličková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Hana Hartmannová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Helena Trešlová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Lenka Nosková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Kateřina Hodaňová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Petra Bittmanová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Václav Jiřička
- Department of PsychologyPrison Service of the Czech RepublicPragueCzech Republic
- Department of Psychiatry, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Michaela Langmajerová
- Department of Psychiatry, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Marc Woodbury‐Smith
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick ChildrenTorontoOntarioCanada
- Faculty of Medical Sciences, Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Mehdi Zarrei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Brett Trost
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Molecular Genetics and McLaughlin CentreUniversity of TorontoTorontoOntarioCanada
| | - Anthony J. Bleyer
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
- Section on Nephrology, Wake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jan Vevera
- Department of Psychiatry, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
- Department of PsychiatryUniversity Hospital PilsenPilsenCzech Republic
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
- Department of Psychiatry, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| |
Collapse
|
8
|
Zhang F, Chen J, Li Y, Ye J, Wang C. Neuronal Scaffold Protein ARMS Interacts with Synaptotagmin-4 C2AB through the Ankyrin Repeat Domain with an Unexpected Mode. Int J Mol Sci 2023; 24:16993. [PMID: 38069318 PMCID: PMC10707181 DOI: 10.3390/ijms242316993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
The ankyrin repeat-rich membrane spanning (ARMS), a transmembrane neuronal scaffold protein, plays a fundamental role in neuronal physiology, including neuronal development, polarity, differentiation, survival and angiogenesis, through interactions with diverse partners. Previous studies have shown that the ARMS negatively regulates brain-derived neurotrophic factor (BDNF) secretion by interacting with Synaptotagmin-4 (Syt4), thereby affecting neurogenesis and the development and function of the nervous system. However, the molecular mechanisms of the ARMS/Syt4 complex assembly remain unclear. Here, we confirmed that the ARMS directly interacts with Syt4 through its N-terminal ankyrin repeats 1-8. Unexpectedly, both the C2A and C2B domains of Syt4 are necessary for binding with the ARMS. We then combined the predicted complex structural models from AlphaFold2 with systematic biochemical analyses using point mutagenesis to underline the molecular basis of ARMS/Syt4 complex formation and to identify two conserved residues, E15 and W72, of the ARMS, as essential residues mediating the assembly of the complex. Furthermore, we showed that ARMS proteins are unable to interact with Syt1 or Syt3, indicating that the interaction between ARMS and Syt4 is specific. Taken together, the findings from this study provide biochemical details on the interaction between the ARMS and Syt4, thereby offering a biochemical basis for the further understanding of the potential mechanisms and functional implications of the ARMS/Syt4 complex formation, especially with regard to the modulation of BDNF secretion and associated neuropathies.
Collapse
Affiliation(s)
- Fa Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiasheng Chen
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yahong Li
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jin Ye
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Chao Wang
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
9
|
Ray NR, Kunkle BW, Hamilton-Nelson K, Kurup JT, Rajabli F, Cosacak MI, Kizil C, Jean-Francois M, Cuccaro M, Reyes-Dumeyer D, Cantwell L, Kuzma A, Vance JM, Gao S, Hendrie HC, Baiyewu O, Ogunniyi A, Akinyemi RO, Lee WP, Martin ER, Wang LS, Beecham GW, Bush WS, Farrer LA, Haines JL, Byrd GS, Schellenberg GD, Mayeux R, Pericak-Vance MA, Reitz C. Extended genome-wide association study employing the African Genome Resources Panel identifies novel susceptibility loci for Alzheimer's Disease in individuals of African ancestry. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.29.23294774. [PMID: 37693582 PMCID: PMC10491365 DOI: 10.1101/2023.08.29.23294774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Despite a two-fold increased risk, individuals of African ancestry have been significantly underrepresented in Alzheimer's Disease (AD) genomics efforts. METHODS GWAS of 2,903 AD cases and 6,265 cognitive controls of African ancestry. Within-dataset results were meta-analyzed, followed by gene-based and pathway analyses, and analysis of RNAseq and whole-genome sequencing data. RESULTS A novel AD risk locus was identified in MPDZ on chromosome 9p23 (rs141610415, MAF=.002, P =3.68×10 -9 ). Two additional novel common and nine novel rare loci approached genome-wide significance at P <9×10 -7 . Comparison of association and LD patterns between datasets with higher and lower degrees of African ancestry showed differential association patterns at chr12q23.2 ( ASCL1 ), suggesting that the association is modulated by regional origin of local African ancestry. DISCUSSION Increased sample sizes and sample sets from Africa covering as much African genetic diversity as possible will be critical to identify additional disease-associated loci and improve deconvolution of local genetic ancestry effects.
Collapse
|
10
|
Del Puerto A, Lopez-Fonseca C, Simón-García A, Martí-Prado B, Barrios-Muñoz AL, Pose-Utrilla J, López-Menéndez C, Alcover-Sanchez B, Cesca F, Schiavo G, Campanero MR, Fariñas I, Iglesias T, Porlan E. Kidins220 sets the threshold for survival of neural stem cells and progenitors to sustain adult neurogenesis. Cell Death Dis 2023; 14:500. [PMID: 37542079 PMCID: PMC10403621 DOI: 10.1038/s41419-023-05995-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 06/22/2023] [Accepted: 07/13/2023] [Indexed: 08/06/2023]
Abstract
In the adult mammalian brain, neural stem cells (NSCs) located in highly restricted niches sustain the generation of new neurons that integrate into existing circuits. A reduction in adult neurogenesis is linked to ageing and neurodegeneration, whereas dysregulation of proliferation and survival of NSCs have been hypothesized to be at the origin of glioma. Thus, unravelling the molecular underpinnings of the regulated activation that NSCs must undergo to proliferate and generate new progeny is of considerable relevance. Current research has identified cues promoting or restraining NSCs activation. Yet, whether NSCs depend on external signals to survive or if intrinsic factors establish a threshold for sustaining their viability remains elusive, even if this knowledge could involve potential for devising novel therapeutic strategies. Kidins220 (Kinase D-interacting substrate of 220 kDa) is an essential effector of crucial pathways for neuronal survival and differentiation. It is dramatically altered in cancer and in neurological and neurodegenerative disorders, emerging as a regulatory molecule with important functions in human disease. Herein, we discover severe neurogenic deficits and hippocampal-based spatial memory defects accompanied by increased neuroblast death and high loss of newly formed neurons in Kidins220 deficient mice. Mechanistically, we demonstrate that Kidins220-dependent activation of AKT in response to EGF restraints GSK3 activity preventing NSCs apoptosis. We also show that NSCs with Kidins220 can survive with lower concentrations of EGF than the ones lacking this molecule. Hence, Kidins220 levels set a molecular threshold for survival in response to mitogens, allowing adult NSCs growth and expansion. Our study identifies Kidins220 as a key player for sensing the availability of growth factors to sustain adult neurogenesis, uncovering a molecular link that may help paving the way towards neurorepair.
Collapse
Affiliation(s)
- Ana Del Puerto
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Autovía A6, Km 7,5, 28040, Madrid, Spain
| | - Coral Lopez-Fonseca
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Ana Simón-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Beatriz Martí-Prado
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departmento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, C/ Dr. Moliner, 50, 46100, Burjassot, Spain
| | - Ana L Barrios-Muñoz
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Celia López-Menéndez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Fabrizia Cesca
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 5, 34127, Trieste, Italy
| | - Giampietro Schiavo
- Department of Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Miguel R Campanero
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departmento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, C/ Dr. Moliner, 50, 46100, Burjassot, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
| | - Eva Porlan
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain.
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
| |
Collapse
|
11
|
Mucke HA. Patent highlights October-November 2022. Pharm Pat Anal 2023; 12:95-102. [PMID: 37477491 DOI: 10.4155/ppa-2023-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
A snapshot of recent noteworthy developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
12
|
Morin post-treatment surpassed calpeptin in ameliorating 3-NP-induced cortical neurotoxicity via modulation of glutamate/calpain axis, Kidins220, and BDNF/TrkB/AKT/CREB trajectory. Int Immunopharmacol 2023; 116:109771. [PMID: 36736222 DOI: 10.1016/j.intimp.2023.109771] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023]
Abstract
The neuroprotective capacity of morin hydrate (MH), a potent antioxidant flavonoid, and calpeptin (CP), a calpain inhibitor, was documented against different insults but not Huntington's disease (HD). Accordingly, we aim to assess the neuroprotective potential of MH and/or CP in a 3-nitropropionic acid (3-NP)-induced HD model. The 3-NP-treated rats were post-treated with saline, MH, CP, or MH + CP for a week. Post-treatment with MH and/or CP amended motor function (beam walking test) and short-/ long-term spatial memory (novel object recognition test) and improved cortical microscopic architecture. On the molecular level, MH, and to a lesser extent CP, inhibited the cortical content/expression of glutamate, calpain, and Kidins220 and abated the inflammatory molecules, nuclear factor (NF)-κB, tumor necrosis factor-α, and interleukin-1β, as well as lipid peroxidation. However, MH, but barely CP, activated the molecules of the neuroprotective trajectory; viz., brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase receptor B (TrkB), protein kinase B (AKT), and cAMP response element-binding protein (CREB). Compared to the single treatments, the combination regimen mediated further reductions in the cortical contents of glutamate, calpain, and Kidins220, effects that extended to entail the anti-inflammatory/anti-oxidant potentials of MH and to a greater extent CP. However, the combination of MH strengthened the fair effect of CP on the survival signaling pathway BDNF/TrkB/AKT/CREB. In conclusion, MH, CP, and especially their combination, afforded neuroprotection against HD through curbing the glutamate/calpain axis, Kidins220, as well as NF-κB-mediated neuroinflammation/oxidative stress, besides activating the BDNF/TrkB/AKT/CREB hub that was partly dependent on calpain inhibition.
Collapse
|
13
|
Yao D, Binan L, Bezney J, Simonton B, Freedman J, Frangieh CJ, Dey K, Geiger-Schuller K, Eraslan B, Gusev A, Regev A, Cleary B. Compressed Perturb-seq: highly efficient screens for regulatory circuits using random composite perturbations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525200. [PMID: 36747806 PMCID: PMC9900787 DOI: 10.1101/2023.01.23.525200] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pooled CRISPR screens with single-cell RNA-seq readout (Perturb-seq) have emerged as a key technique in functional genomics, but are limited in scale by cost and combinatorial complexity. Here, we reimagine Perturb-seq's design through the lens of algorithms applied to random, low-dimensional observations. We present compressed Perturb-seq, which measures multiple random perturbations per cell or multiple cells per droplet and computationally decompresses these measurements by leveraging the sparse structure of regulatory circuits. Applied to 598 genes in the immune response to bacterial lipopolysaccharide, compressed Perturb-seq achieves the same accuracy as conventional Perturb-seq at 4 to 20-fold reduced cost, with greater power to learn genetic interactions. We identify known and novel regulators of immune responses and uncover evolutionarily constrained genes with downstream targets enriched for immune disease heritability, including many missed by existing GWAS or trans-eQTL studies. Our framework enables new scales of interrogation for a foundational method in functional genomics.
Collapse
Affiliation(s)
- Douglas Yao
- Program in Systems, Synthetic, and Quantitative Biology, Harvard University, Cambridge, MA
| | - Loic Binan
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Jon Bezney
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
- Current address: Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Brooke Simonton
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Jahanara Freedman
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Chris J Frangieh
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Kushal Dey
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | | | - Alexander Gusev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA
- These authors jointly supervised this work
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA
- Current address: Genentech, South San Francisco, CA
- These authors jointly supervised this work
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA
- Department of Biology, Boston University, Boston, MA
- Department of Biomedical Engineering, Boston University, Boston, MA
- Program in Bioinformatics, Boston University, Boston, MA
- Biological Design Center, Boston University, Boston, MA
- These authors jointly supervised this work
| |
Collapse
|
14
|
Transcriptomics of angiotensin II-induced long noncoding and coding RNAs in endothelial cells. J Hypertens 2022; 40:1303-1313. [PMID: 35762471 DOI: 10.1097/hjh.0000000000003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Angiotensin II (Ang II)-induced endothelial dysfunction plays an important role in the pathogenesis of cardiovascular diseases such as systemic hypertension, cardiac hypertrophy and atherosclerosis. Recently, long noncoding RNAs (lncRNAs) have been shown to play an essential role in the pathobiology of cardiovascular diseases; however, the effect of Ang II on lncRNAs and coding RNAs expression in endothelial cells has not been evaluated. Accordingly, we sought to evaluate the expression profiles of lncRNAs and coding RNAs in endothelial cells following treatment with Ang II. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and treated with Ang II (10-6 mol/l) for 24 h. The cells were then profiled for the expression of lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS In HUVECs following Ang II treatment, from a total of 30 584 lncRNA targets screened, 25 targets were significantly upregulated, while 69 were downregulated. In the same HUVECs samples, from 26 106 mRNA targets screened, 28 targets were significantly upregulated and 67 were downregulated. Of the differentially expressed lncRNAs, RP11-354P11.2 and RP11-360F5.1 were the most upregulated (11-fold) and downregulated (three-fold) lncRNAs, respectively. Assigning the differentially regulated genes into functional groups using bioinformatics reveals numerous genes involved in the nucleotide excision repair and ECM-receptor interaction. CONCLUSION This is the first study to profile the Ang II-induced differentially expressed lncRNAs and mRNAs in human endothelial cells. Our results reveal novel targets and substantially extend the list of potential candidate genes involved in Ang II-induced endothelial dysfunction and cardiovascular diseases.
Collapse
|
15
|
Almacellas-Barbanoj A, Albini M, Satapathy A, Jaudon F, Michetti C, Krawczun-Rygmaczewska A, Huang H, Manago F, Papaleo F, Benfenati F, Cesca F. Kidins220/ARMS modulates brain morphology and anxiety-like traits in adult mice. Cell Death Dis 2022; 8:58. [PMID: 35140204 PMCID: PMC8828717 DOI: 10.1038/s41420-022-00854-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/25/2022] [Indexed: 11/17/2022]
Abstract
Kinase D interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), is a transmembrane scaffold protein that participates in fundamental aspects of neuronal physiology including cell survival, differentiation, and synaptic plasticity. The Kidins220 constitutive knockout line displays developmental defects in the nervous and cardiovascular systems that lead to embryonic lethality, which has so far precluded the study of this protein in the adult. Moreover, Kidins220 mRNA is tightly regulated by alternative splicing, whose impact on nervous system physiology has not yet been addressed in vivo. Here, we have asked to what extent the absence of Kidins220 splicing and the selective knockout of Kidins220 impact on adult brain homeostasis. To answer this question, we used a floxed line that expresses only the full-length, non-spliced Kidins220 mRNA, and a forebrain-specific, CaMKII-Cre driven Kidins220 conditional knockout (cKO) line. Kidins220 cKO brains are characterized by enlarged ventricles in the absence of cell death, and by deficient dendritic arborization in several cortical regions. The deletion of Kidins220 leads to behavioral changes, such as reduced anxiety-like traits linked to alterations in TrkB-BDNF signaling and sex-dependent alterations of hippocampal-dependent spatial memory. Kidins220 floxed mice present similarly enlarged brain ventricles and increased associative memory. Thus, both the absolute levels of Kidins220 expression and its splicing pattern are required for the correct brain development and related expression of behavioral phenotypes. These findings are relevant in light of the increasing evidence linking mutations in the human KIDINS220 gene to the onset of severe neurodevelopmental disorders.
Collapse
Affiliation(s)
- Amanda Almacellas-Barbanoj
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Albini
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Annyesha Satapathy
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Huiping Huang
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Francesca Manago
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132, Genova, Italy. .,Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.
| |
Collapse
|
16
|
Jaudon F, Albini M, Ferroni S, Benfenati F, Cesca F. A developmental stage- and Kidins220-dependent switch in astrocyte responsiveness to brain-derived neurotrophic factor. J Cell Sci 2021; 134:jcs258419. [PMID: 34279618 DOI: 10.1242/jcs.258419] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022] Open
Abstract
Astroglial cells are key to maintain nervous system homeostasis. Neurotrophins are known for their pleiotropic effects on neuronal physiology but also exert complex functions to glial cells. Here, we investigated (i) the signaling competence of mouse embryonic and postnatal primary cortical astrocytes exposed to brain-derived neurotrophic factor (BDNF) and, (ii) the role of kinase D-interacting substrate of 220 kDa (Kidins220), a transmembrane scaffold protein that mediates neurotrophin signaling in neurons. We found a shift from a kinase-based response in embryonic cells to a response predominantly relying on intracellular Ca2+ transients [Ca2+]i within postnatal cultures, associated with a decrease in the synthesis of full-length BDNF receptor TrkB, with Kidins220 contributing to the BDNF-activated kinase and [Ca2+]i pathways. Finally, Kidins220 participates in the homeostatic function of astrocytes by controlling the expression of the ATP-sensitive inward rectifier potassium channel 10 (Kir4.1) and the metabolic balance of embryonic astrocytes. Overall, our data contribute to the understanding of the complex role played by astrocytes within the central nervous system, and identify Kidins220 as a novel actor in the increasing number of pathologies characterized by astrocytic dysfunctions. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Martina Albini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
17
|
Gungor S, Oktay Y, Hiz S, Aranguren-Ibáñez Á, Kalafatcilar I, Yaramis A, Karaca E, Yis U, Sonmezler E, Ekinci B, Aslan M, Yilmaz E, Özgör B, Balaraju S, Szabo N, Laurie S, Beltran S, MacArthur DG, Hathazi D, Töpf A, Roos A, Lochmuller H, Vernos I, Horvath R. Autosomal recessive variants in TUBGCP2 alter the γ-tubulin ring complex leading to neurodevelopmental disease. iScience 2021; 24:101948. [PMID: 33458610 PMCID: PMC7797523 DOI: 10.1016/j.isci.2020.101948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/20/2020] [Accepted: 12/11/2020] [Indexed: 12/23/2022] Open
Abstract
Microtubules help building the cytoskeleton of neurons and other cells. Several components of the gamma-tubulin (γ-tubulin) complex have been previously reported in human neurodevelopmental diseases. We describe two siblings from a consanguineous Turkish family with dysmorphic features, developmental delay, brain malformation, and epilepsy carrying a homozygous mutation (p.Glu311Lys) in TUBGCP2 encoding the γ-tubulin complex 2 (GCP2) protein. This variant is predicted to disrupt the electrostatic interaction of GCP2 with GCP3. In primary fibroblasts carrying the variant, we observed a faint delocalization of γ-tubulin during the cell cycle but normal GCP2 protein levels. Through mass spectrometry, we observed dysregulation of multiple proteins involved in the assembly and organization of the cytoskeleton and the extracellular matrix, controlling cellular adhesion and of proteins crucial for neuronal homeostasis including axon guidance. In summary, our functional and proteomic studies link TUBGCP2 and the γ-tubulin complex to the development of the central nervous system in humans.
Collapse
Affiliation(s)
- Serdal Gungor
- Inonu University, Faculty of Medicine, Turgut Ozal Research Center, Department of Paediatric Neurology, Malatya, Turkey
| | - Yavuz Oktay
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University and Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Semra Hiz
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Dokuz Eylul University, Faculty of Medicine, Department of Pediatric Neurology Izmir, Turkey
| | - Álvaro Aranguren-Ibáñez
- Centre for Genomic Regulation (CRG), the Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Ipek Kalafatcilar
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Dokuz Eylul University, Faculty of Medicine, Department of Pediatric Neurology Izmir, Turkey
| | - Ahmet Yaramis
- Pediatric Neurology Clinic, Private Office, Diyarbakir, Turkey
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University and Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Uluc Yis
- Dokuz Eylul University, Faculty of Medicine, Department of Pediatric Neurology Izmir, Turkey
| | - Ece Sonmezler
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Burcu Ekinci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Mahmut Aslan
- Dokuz Eylul University, Faculty of Medicine, Department of Pediatric Neurology Izmir, Turkey
| | - Elmasnur Yilmaz
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Bilge Özgör
- Inonu University, Faculty of Medicine, Turgut Ozal Research Center, Department of Paediatric Neurology, Malatya, Turkey
| | - Sunitha Balaraju
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurosciences, John Van Geest Cambridge Centre for Brain Repair, University of Cambridge School of Clinical Medicine, Robinson Way, Cambridge CB2 0PY, UK
| | - Nora Szabo
- Department of Clinical Neurosciences, John Van Geest Cambridge Centre for Brain Repair, University of Cambridge School of Clinical Medicine, Robinson Way, Cambridge CB2 0PY, UK
- Budai Children Hospital, Észak-Közép-budai Centrum, Új Szent János Kórház és Szakrendelő, Budapest, Hungary
| | - Steven Laurie
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sergi Beltran
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Daniel G. MacArthur
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Denisa Hathazi
- Department of Clinical Neurosciences, John Van Geest Cambridge Centre for Brain Repair, University of Cambridge School of Clinical Medicine, Robinson Way, Cambridge CB2 0PY, UK
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| | - Andreas Roos
- Leibniz Institut für Analytische Wissenschaften, ISAS, Dortmund, Germany & Pediatric Neurology, University Hospital, University of Duisburg-Essen, Faculty of Medicine, Essen, Germany
| | - Hanns Lochmuller
- Children's Hospital of Eastern Ontario Research Institute; Division of Neurology, Department of Medicine, the Ottawa Hospital; and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Isabelle Vernos
- Centre for Genomic Regulation (CRG), the Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Spain
| | - Rita Horvath
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
Kidins220 deficiency causes ventriculomegaly via SNX27-retromer-dependent AQP4 degradation. Mol Psychiatry 2021; 26:6411-6426. [PMID: 34002021 PMCID: PMC8760065 DOI: 10.1038/s41380-021-01127-9] [Citation(s) in RCA: 11] [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: 07/14/2020] [Revised: 03/29/2021] [Accepted: 04/14/2021] [Indexed: 02/04/2023]
Abstract
Several psychiatric, neurologic and neurodegenerative disorders present increased brain ventricles volume, being hydrocephalus the disease with the major manifestation of ventriculomegaly caused by the accumulation of high amounts of cerebrospinal fluid (CSF). The molecules and pathomechanisms underlying cerebral ventricular enlargement are widely unknown. Kinase D interacting substrate of 220 kDa (KIDINS220) gene has been recently associated with schizophrenia and with a novel syndrome characterized by spastic paraplegia, intellectual disability, nystagmus and obesity (SINO syndrome), diseases frequently occurring with ventriculomegaly. Here we show that Kidins220, a transmembrane protein effector of various key neuronal signalling pathways, is a critical regulator of CSF homeostasis. We observe that both KIDINS220 and the water channel aquaporin-4 (AQP4) are markedly downregulated at the ventricular ependymal lining of idiopathic normal pressure hydrocephalus (iNPH) patients. We also find that Kidins220 deficient mice develop ventriculomegaly accompanied by water dyshomeostasis and loss of AQP4 in the brain ventricular ependymal layer and astrocytes. Kidins220 is a known cargo of the SNX27-retromer, a complex that redirects endocytosed plasma membrane proteins (cargos) back to the cell surface, thus avoiding their targeting to lysosomes for degradation. Mechanistically, we show that AQP4 is a novel cargo of the SNX27-retromer and that Kidins220 deficiency promotes a striking and unexpected downregulation of the SNX27-retromer that results in AQP4 lysosomal degradation. Accordingly, SNX27 silencing decreases AQP4 levels in wild-type astrocytes whereas SNX27 overexpression restores AQP4 content in Kidins220 deficient astrocytes. Together our data suggest that the KIDINS220-SNX27-retromer-AQP4 pathway is involved in human ventriculomegaly and open novel therapeutic perspectives.
Collapse
|
19
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics identifies neuronal differentiation pathway networks activated by cathepsin inhibition treatment in neuroblastoma cells that are enhanced by concurrent 13-cis retinoic acid treatment. J Proteomics 2020; 232:104068. [PMID: 33278663 DOI: 10.1016/j.jprot.2020.104068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Neuroblastoma is the second most common pediatric cancer involving the peripheral nervous system in which stage IVS metastatic tumors regress due to spontaneous differentiation. 13-cis retinoic acid (13-cis RA) is currently used in the clinic for its differentiation effects and although it improves outcomes, relapse is seen in half of high-risk patients. Combinatorial therapies have been shown to be more effective in oncotherapy and since cathepsin inhibition reduces tumor growth, we explored the potential of coupling 13-cis RA with a cathepsin inhibitor (K777) to enhance therapeutic efficacy against neuroblastoma. Shotgun proteomics was used to identify proteins affected by K777 and dual (13-cis RA/K777) treatment in neuroblastoma SK-N-SH cells. Cathepsin inhibition was more effective in increasing proteins involved in neuronal differentiation and neurite outgrowth than 13-cis RA alone, but the combination of both treatments enhanced the neuronal differentiation effect. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of more accurate diagnostic markers and more effective treatments. In this study, we established a differentiation proteomic map of SK-N-SH cells treated with a cathepsin inhibitor (K777) and K777/13-cis RA (dual). Bioinformatic analysis revealed these treatments enhanced neuronal differentiation and axonogenesis pathways. The most affected proteins in these pathways may become valuable biomarkers of efficacy of drugs designed to enhance differentiation of neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
20
|
Li Q, Marcu DC, Palazzo O, Turner F, King D, Spires-Jones TL, Stefan MI, Busch KE. High neural activity accelerates the decline of cognitive plasticity with age in Caenorhabditis elegans. eLife 2020; 9:59711. [PMID: 33228848 PMCID: PMC7685709 DOI: 10.7554/elife.59711] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/18/2020] [Indexed: 12/11/2022] Open
Abstract
The ability to learn progressively declines with age. Neural hyperactivity has been implicated in impairing cognitive plasticity with age, but the molecular mechanisms remain elusive. Here, we show that chronic excitation of the Caenorhabditis elegans O2-sensing neurons during ageing causes a rapid decline of experience-dependent plasticity in response to environmental O2 concentration, whereas sustaining lower activity of O2-sensing neurons retains plasticity with age. We demonstrate that neural activity alters the ageing trajectory in the transcriptome of O2-sensing neurons, and our data suggest that high-activity neurons redirect resources from maintaining plasticity to sustaining continuous firing. Sustaining plasticity with age requires the K+-dependent Na+/Ca2+ (NCKX) exchanger, whereas the decline of plasticity with age in high-activity neurons acts through calmodulin and the scaffold protein Kidins220. Our findings demonstrate directly that the activity of neurons alters neuronal homeostasis to govern the age-related decline of neural plasticity and throw light on the mechanisms involved.
Collapse
Affiliation(s)
- Qiaochu Li
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel-Cosmin Marcu
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ottavia Palazzo
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Frances Turner
- Edinburgh Genomics (Genome Science), Ashworth Laboratories, The University of Edinburgh, Edinburgh, United Kingdom
| | - Declan King
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Melanie I Stefan
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,ZJU-UoE Institute, Zhejiang University, Haining, China
| | - Karl Emanuel Busch
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Jacquemin V, Antoine M, Duerinckx S, Massart A, Desir J, Perazzolo C, Cassart M, Thomas D, Segers V, Lecomte S, Abramowicz M, Pirson I. TrkA mediates effect of novel KIDINS220 mutation in human brain ventriculomegaly. Hum Mol Genet 2020; 29:3757-3764. [PMID: 33205811 DOI: 10.1093/hmg/ddaa245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/22/2022] Open
Abstract
Congenital hydrocephalus is a potentially devastating, highly heterogeneous condition whose genetic subset remains incompletely known. We here report a consanguineous family where three fetuses presented with brain ventriculomegaly and limb contractures and shared a very rare homozygous variant of KIDINS220, consisting of an in-frame deletion of three amino acids adjacent to the fourth transmembrane domain. Fetal brain imaging and autopsy showed major ventriculomegaly, reduced brain mass, and with no histomorphologic abnormalities. We demonstrate that the binding of KIDINS220 to TrkA is diminished by the deletion mutation. This family is the second that associates a KIDINS220 genetic variant with human ventriculomegaly and limb contractures, validating causality of the gene and indicating TrkA as a likely mediator of the phenotype.
Collapse
Affiliation(s)
| | - Mathieu Antoine
- IRIBHM, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Sarah Duerinckx
- IRIBHM, Université Libre de Bruxelles, 1070 Brussels, Belgium.,Neurology Department, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Annick Massart
- IRIBHM, Université Libre de Bruxelles, 1070 Brussels, Belgium.,Department of Nephrology, Hôpital Universitaire d'Anvers, 2650 Edegem, Belgium
| | - Julie Desir
- Human Genetics Center, Institute of Pathology and Genetics, 6041 Charleroi, Belgium
| | | | - Marie Cassart
- Department of Gynecology and Obstetrics, Hôpitaux Iris Sud, 1050 Brussels, Belgium
| | - Dominique Thomas
- Department of Gynecology and Obstetrics, Hôpitaux Iris Sud, 1050 Brussels, Belgium
| | - Valérie Segers
- Department of Anatomopathology, CHU Brugmann, 1020 Brussels, Belgium
| | - Sophie Lecomte
- Department of Anatomopathology, CHU Brugmann, 1020 Brussels, Belgium
| | - Marc Abramowicz
- IRIBHM, Université Libre de Bruxelles, 1070 Brussels, Belgium.,Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Isabelle Pirson
- IRIBHM, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
22
|
Jaudon F, Chiacchiaretta M, Albini M, Ferroni S, Benfenati F, Cesca F. Kidins220/ARMS controls astrocyte calcium signaling and neuron-astrocyte communication. Cell Death Differ 2020; 27:1505-1519. [PMID: 31624352 PMCID: PMC7206051 DOI: 10.1038/s41418-019-0431-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
Through their ability to modulate synaptic transmission, glial cells are key regulators of neuronal circuit formation and activity. Kidins220/ARMS (kinase-D interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning) is one of the key effectors of the neurotrophin pathways in neurons where it is required for differentiation, survival, and plasticity. However, its role in glial cells remains largely unknown. Here, we show that ablation of Kidins220 in primary cultured astrocytes induced defects in calcium (Ca2+) signaling that were linked to altered store-operated Ca2+ entry and strong overexpression of the transient receptor potential channel TRPV4. Moreover, Kidins220-/- astrocytes were more sensitive to genotoxic stress. We also show that Kidins220 expression in astrocytes is required for the establishment of proper connectivity of cocultured wild-type neurons. Altogether, our data reveal a previously unidentified role for astrocyte-expressed Kidins220 in the control of glial Ca2+ dynamics, survival/death pathways and astrocyte-neuron communication.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Martina Albini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy.
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.
| |
Collapse
|
23
|
Regulation of BDNF Release by ARMS/Kidins220 through Modulation of Synaptotagmin-IV Levels. J Neurosci 2018; 38:5415-5428. [PMID: 29769266 DOI: 10.1523/jneurosci.1653-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 04/07/2018] [Accepted: 05/03/2018] [Indexed: 11/21/2022] Open
Abstract
BDNF is a growth factor with important roles in the nervous system in both physiological and pathological conditions, but the mechanisms controlling its secretion are not completely understood. Here, we show that ARMS/Kidins220 negatively regulates BDNF secretion in neurons from the CNS and PNS. Downregulation of the ARMS/Kidins220 protein in the adult mouse brain increases regulated BDNF secretion, leading to its accumulation in the striatum. Interestingly, two mouse models of Huntington's disease (HD) showed increased levels of ARMS/Kidins220 in the hippocampus and regulated BDNF secretion deficits. Importantly, reduction of ARMS/Kidins220 in hippocampal slices from HD mice reversed the impaired regulated BDNF release. Moreover, there are increased levels of ARMS/Kidins220 in the hippocampus and PFC of patients with HD. ARMS/Kidins220 regulates Synaptotagmin-IV levels, which has been previously observed to modulate BDNF secretion. These data indicate that ARMS/Kidins220 controls the regulated secretion of BDNF and might play a crucial role in the pathogenesis of HD.SIGNIFICANCE STATEMENT BDNF is an important growth factor that plays a fundamental role in the correct functioning of the CNS. The secretion of BDNF must be properly controlled to exert its functions, but the proteins regulating its release are not completely known. Using neuronal cultures and a new conditional mouse to modulate ARMS/Kidins220 protein, we report that ARMS/Kidins220 negatively regulates BDNF secretion. Moreover, ARMS/Kidins220 is overexpressed in two mouse models of Huntington's disease (HD), causing an impaired regulation of BDNF secretion. Furthermore, ARMS/Kidins220 levels are increased in brain samples from HD patients. Future studies should address whether ARMS/Kidins220 has any function on the pathophysiology of HD.
Collapse
|
24
|
Cai S, Cai J, Jiang WG, Ye L. Kidins220 and tumour development: Insights into a complexity of cross-talk among signalling pathways (Review). Int J Mol Med 2017; 40:965-971. [PMID: 28849114 PMCID: PMC5593494 DOI: 10.3892/ijmm.2017.3093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022] Open
Abstract
The mechanistic complexes of kinase D-interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning (Kidins220/ARMS) bind and integrate a variety of cellular cues to mediate neuronal activities such as neuronal differentiation, survival, and cytoskeleton remodelling by interacting with a variety of binding partners. Accumulated evidence has also indicated its role in the regulation of vascular development. Mice with Kidins220 knockdown phenotypically present with cardiovascular abnormalities. Kidins220 also contributes to immunomodulation in combination with B cells and T cells. Moreover, emerging evidence has revealed that this protein regulates many crucial cellular processes and thus has been implicated in an increasing number of malignancies. Here, we review recent advances in our understanding of Kidins220 and its role in cancer development. Further investigation is warranted to shed light on the role played by Kidins220 in the dynamic arrangement of the cytoskeleton and epithelial–mesenchymal transition, and its implication in tumourigenesis and cancer progression.
Collapse
Affiliation(s)
- Shuo Cai
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Jun Cai
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| |
Collapse
|