1
|
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] [MESH Headings] [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
|
2
|
Rada P, Carceller-López E, Hitos AB, Gómez-Santos B, Fernández-Hernández C, Rey E, Pose-Utrilla J, García-Monzón C, González-Rodríguez Á, Sabio G, García A, Aspichueta P, Iglesias T, Valverde ÁM. Protein kinase D2 modulates hepatic insulin sensitivity in male mice. Mol Metab 2024; 90:102045. [PMID: 39401614 PMCID: PMC11535753 DOI: 10.1016/j.molmet.2024.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Protein kinase D (PKD) family is emerging as relevant regulator of metabolic homeostasis. However, the precise role of PKD2 in modulating hepatic insulin signaling has not been fully elucidated and it is the aim of this study. METHODS PKD inhibition was analyzed for insulin signaling in mouse and human hepatocytes. PKD2 was overexpressed in Huh7 hepatocytes and mouse liver, and insulin responses were evaluated. Mice with hepatocyte-specific PKD2 depletion (PKD2ΔHep) and PKD2fl/fl mice were fed a chow (CHD) or high fat diet (HFD) and glucose homeostasis and lipid metabolism were investigated. RESULTS PKD2 silencing enhanced insulin signaling in hepatocytes, an effect also found in primary hepatocytes from PKD2ΔHep mice. Conversely, a constitutively active PKD2 mutant reduced insulin-stimulated AKT phosphorylation. A more in-depth analysis revealed reduced IRS1 serine phosphorylation under basal conditions and increased IRS1 tyrosine phosphorylation in PKD2ΔHep primary hepatocytes upon insulin stimulation and, importantly PKD co-immunoprecipitates with IRS1. In vivo constitutively active PKD2 overexpression resulted in a moderate impairment of glucose homeostasis and reduced insulin signaling in the liver. On the contrary, HFD-fed PKD2ΔHep male mice displayed improved glucose and pyruvate tolerance, as well as higher peripheral insulin tolerance and enhanced hepatic insulin signaling compared to control PKD2fl/fl mice. Despite of a remodeling of hepatic lipid metabolism in HFD-fed PKD2ΔHep mice, similar steatosis grade was found in both genotypes. CONCLUSIONS Results herein have unveiled an unknown role of PKD2 in the control of insulin signaling in the liver at the level of IRS1 and point PKD2 as a therapeutic target for hepatic insulin resistance.
Collapse
Affiliation(s)
- Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Elena Carceller-López
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Ana B Hitos
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Constanza Fernández-Hernández
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Esther Rey
- Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carmelo García-Monzón
- Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Águeda González-Rodríguez
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Antonia García
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; BioBizkaia Health Research Institute, Barakaldo, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| |
Collapse
|
3
|
Yang W, Wang S, Huo X, Yang K, Guo Z, Li Y, Ji X, Hao B, Liao S. Novel autosomal recessive SINO syndrome-associated KIDINS220 variants provide insight into the genotype-phenotype correlation. Heliyon 2024; 10:e37355. [PMID: 39296002 PMCID: PMC11408833 DOI: 10.1016/j.heliyon.2024.e37355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Background KIDINS220 encodes a transmembrane scaffold protein, kinase D-interacting substrate of 220 kDa, that regulates neurotrophin signaling. Variants in KIDINS220 have been linked to spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO) syndrome or prenatal fatal cerebral ventriculomegaly and arthrogryposis (VENARG). This study aimed to investigate the genotype-phenotype correlation of pathogenic KIDINS220 variants. Methods We performed whole-exome sequencing on a patient with SINO syndrome and epilepsy. Identified pathogenic variants were confirmed using Sanger sequencing and evaluated with in silico tools. A comprehensive literature review was conducted to analyze the genetic and phenotypic data of both the newly diagnosed patient and previously reported cases with KIDINS220 variants. Results We identified novel compound heterozygous variants in KIDINS220, c.1556C > T (p.Thr519Met) and c.2374C > T (p.Arg792*), in the patient. Our analysis revealed that biallelic loss-of-function variants in KIDINS220 are associated with VENARG or autosomal recessive SINO (AR-SINO), whereas carboxy-terminal truncated variants that escape nonsense-mediated mRNA decay and lack amino acid residues 1507-1529 are linked to autosomal dominant SINO (AD-SINO). Patients with AR-SINO exhibit more severe clinical features compared to those with AD-SINO. Conclusions Our study expands the spectrum of KIDINS220 variants associated with AR-SINO and provides a valuable genotype-phenotype correlation for pathogenic KIDINS220 variants.
Collapse
Affiliation(s)
- Wenke Yang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuyue Wang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- Central Hospital of Wuhan, Wuhan, China
| | - Xiaodong Huo
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Yang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenglong Guo
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| | - Yanjun Li
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinying Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Bingtao Hao
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| | - Shixiu Liao
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| |
Collapse
|
4
|
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
|
5
|
Albini M, Almacellas-Barbanoj A, Krawczun-Rygmaczewska A, Ciano L, Benfenati F, Michetti C, Cesca F. Alterations in KIDINS220/ARMS Expression Impact Sensory Processing and Social Behavior in Adult Mice. Int J Mol Sci 2024; 25:2334. [PMID: 38397009 PMCID: PMC10889203 DOI: 10.3390/ijms25042334] [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: 01/17/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Kinase D-interacting substrate of 220 kDa (Kidins220) is a transmembrane protein that participates in neural cell survival, maturation, and plasticity. Mutations in the human KIDINS220 gene are associated with a neurodevelopmental disorder ('SINO' syndrome) characterized by spastic paraplegia, intellectual disability, and in some cases, autism spectrum disorder. To better understand the pathophysiology of KIDINS220-linked pathologies, in this study, we assessed the sensory processing and social behavior of transgenic mouse lines with reduced Kidins220 expression: the CaMKII-driven conditional knockout (cKO) line, lacking Kidins220 in adult forebrain excitatory neurons, and the Kidins220floxed line, expressing constitutively lower protein levels. We show that alterations in Kidins220 expression levels and its splicing pattern cause impaired response to both auditory and olfactory stimuli. Both transgenic lines show impaired startle response to high intensity sounds, with preserved pre-pulsed inhibition, and strongly reduced social odor recognition. In the Kidins220floxed line, olfactory alterations are associated with deficits in social memory and increased aggressive behavior. Our results broaden our knowledge of the SINO syndrome; understanding sensory information processing and its deviations under neuropathological conditions is crucial for devising future therapeutic strategies to enhance the quality of life of affected individuals.
Collapse
Affiliation(s)
- Martina Albini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Amanda Almacellas-Barbanoj
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Lorenzo Ciano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy; (M.A.); (A.A.-B.); (A.K.-R.); (L.C.); (F.B.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
6
|
Schaffer AM, Fiala GJ, Hils M, Natali E, Babrak L, Herr LA, Romero-Mulero MC, Cabezas-Wallscheid N, Rizzi M, Miho E, Schamel WWA, Minguet S. Kidins220 regulates the development of B cells bearing the λ light chain. eLife 2024; 13:e83943. [PMID: 38271217 PMCID: PMC10810608 DOI: 10.7554/elife.83943] [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: 10/12/2022] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
The ratio between κ and λ light chain (LC)-expressing B cells varies considerably between species. We recently identified Kinase D-interacting substrate of 220 kDa (Kidins220) as an interaction partner of the BCR. In vivo ablation of Kidins220 in B cells resulted in a marked reduction of λLC-expressing B cells. Kidins220 knockout B cells fail to open and recombine the genes of the Igl locus, even in genetic scenarios where the Igk genes cannot be rearranged or where the κLC confers autoreactivity. Igk gene recombination and expression in Kidins220-deficient B cells is normal. Kidins220 regulates the development of λLC B cells by enhancing the survival of developing B cells and thereby extending the time-window in which the Igl locus opens and the genes are rearranged and transcribed. Further, our data suggest that Kidins220 guarantees optimal pre-BCR and BCR signaling to induce Igl locus opening and gene recombination during B cell development and receptor editing.
Collapse
Affiliation(s)
- Anna-Maria Schaffer
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Gina Jasmin Fiala
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Miriam Hils
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of MunichMunichGermany
| | - Eriberto Natali
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Lmar Babrak
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Laurenz Alexander Herr
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Mari Carmen Romero-Mulero
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| | - Marta Rizzi
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of ViennaViennaAustria
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
- aiNET GmbHBaselSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Wolfgang WA Schamel
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Susana Minguet
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| |
Collapse
|
7
|
Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res 2023; 197:42-51. [PMID: 36780947 DOI: 10.1016/j.neures.2023.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023]
Abstract
Astrocytes are emerging in the neuroscience field as crucial modulators of brain functions, from the molecular control of synaptic plasticity to orchestrating brain-wide circuit activity for cognitive processes. The cellular pathways through which astrocytes modulate neuronal activity and plasticity are quite diverse. In this review, we focus on neurotrophic pathways, mostly those mediated by brain-derived neurotrophic factor (BDNF). Neurotrophins are a well-known family of trophic factors with pleiotropic functions in neuronal survival, maturation and activity. Within the brain, BDNF is the most abundantly expressed and most studied of all neurotrophins. While we have detailed knowledge of the effect of BDNF on neurons, much less is known about its physiology on astroglia. However, over the last years new findings emerged demonstrating that astrocytes take an active part into BDNF physiology. In this work, we discuss the state-of-the-art knowledge about astrocytes and BDNF. Indeed, astrocytes sense extracellular BDNF through its specific TrkB receptors and activate intracellular responses that greatly vary depending on the brain area, stage of development and receptors expressed. Astrocytes also uptake and recycle BDNF / proBDNF at synapses contributing to synaptic plasticity. Finally, experimental evidence is now available describing deficits in astrocytic BDNF in several neuropathologies, suggesting that astrocytic BDNF may represent a promising target for clinical translation.
Collapse
Affiliation(s)
- Martina Albini
- Department of Experimental Medicine, University of Genova, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy
| | - Fabrizia Cesca
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy.
| |
Collapse
|
8
|
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
|
9
|
Arévalo JC, Deogracias R. Mechanisms Controlling the Expression and Secretion of BDNF. Biomolecules 2023; 13:biom13050789. [PMID: 37238659 DOI: 10.3390/biom13050789] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Brain-derived nerve factor (BDNF), through TrkB receptor activation, is an important modulator for many different physiological and pathological functions in the nervous system. Among them, BDNF plays a crucial role in the development and correct maintenance of brain circuits and synaptic plasticity as well as in neurodegenerative diseases. The proper functioning of the central nervous system depends on the available BDNF concentrations, which are tightly regulated at transcriptional and translational levels but also by its regulated secretion. In this review we summarize the new advances regarding the molecular players involved in BDNF release. In addition, we will address how changes of their levels or function in these proteins have a great impact in those functions modulated by BDNF under physiological and pathological conditions.
Collapse
Affiliation(s)
- Juan Carlos Arévalo
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Rubén Deogracias
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
10
|
Sánchez-Sánchez J, Vicente-García C, Cañada-García D, Martín-Zanca D, Arévalo JC. ARMS/Kidins220 regulates nociception by controlling brain-derived neurotrophic factor secretion. Pain 2023; 164:563-576. [PMID: 35916735 DOI: 10.1097/j.pain.0000000000002741] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/15/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Pain is an alarm mechanism to prevent body damage in response to noxious stimuli. The nerve growth factor (NGF)/TrkA axis plays an essential role as pain mediator, and several clinical trials using antibodies against NGF have yielded promising results, but side effects have precluded their clinical approval. A better understanding of the mechanism of NGF/TrkA-mediated nociception is needed. Here, we find that ARMS/Kidins220, a scaffold protein for Trk receptors, is a modulator of nociception. Male mice, with ARMS/Kidins220 reduction exclusively in TrkA-expressing cells, displayed hyperalgesia to heat, inflammatory, and capsaicin stimuli, but not to cold or mechanical stimuli. Simultaneous deletion of brain-derived neurotrophic factor (BDNF) reversed the effects of ARMS/Kidins220 knock down alone. Mechanistically, ARMS/Kidins220 levels are reduced in vitro and in vivo in response to capsaicin through calpains, and this reduction leads to enhanced regulated BDNF secretion from dorsal root ganglion. Altogether, these data indicate that ARMS/Kidins220 protein levels have a role as a pain modulator in the NGF/TrkA axis regulating BDNF secretion.
Collapse
Affiliation(s)
- Julia Sánchez-Sánchez
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Cristina Vicente-García
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Daniel Cañada-García
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Dionisio Martín-Zanca
- Instituto de Biología Funcional y Genómica, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Juan C Arévalo
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
11
|
ARMS-NF-κB signaling regulates intracellular ROS to induce autophagy-associated cell death upon oxidative stress. iScience 2023; 26:106005. [PMID: 36798436 PMCID: PMC9926119 DOI: 10.1016/j.isci.2023.106005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/23/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Ankyrin repeat-rich membrane spanning (ARMS) plays roles in neural development, neuropathies, and tumor formation. Such pleiotropic function of ARMS is often attributed to diverse ARMS-interacting molecules in different cell context. However, it might be achieved by ARMS' effect on global biological mediator like reactive oxygen species (ROS). We established ARMS-knockdown in melanoma cells (siARMS) and in Drosophila eyes (GMR>dARMS RNAi ) and challenged them with H2O2. Decreased ARMS in both systems compromises nuclear translocation of NF-κB and induces ROS, which in turn augments autophagy flux and confers susceptibility to H2O2-triggered autophagic cell death. Resuming NF-κB activity or reducing ROS by antioxidants in siARMS cells and GMR>dARMS RNAi fly decreases intracellular peroxides level concurrent with reduced autophagy and attenuated cell death. Conversely, blocking NF-κB activity in wild-type flies/melanoma enhances ROS and induces autophagy with cell death. We thus uncover intracellular ROS modulated by ARMS-NFκB signaling primes autophagy for autophagic cell death upon oxidative stress.
Collapse
|
12
|
Brady LI, DeFrance B, Tarnopolsky M. Pre- and Postnatal Characterization of Autosomal Recessive KIDINS220-Associated Ventriculomegaly. Mol Syndromol 2022; 13:419-424. [PMID: 36588759 PMCID: PMC9801333 DOI: 10.1159/000522486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 01/04/2023] Open
Abstract
Introduction Heterozygous loss-of-function variants in the last 2 exons of KIDINS220 have been associated with spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO). Syndromic features of this condition include macrocephaly and dilatation of the lateral ventricles. Homozygous variants in the more proximal exons of KIDINS220 have been reported in several fetuses with a similar but more severe phenotype of limb contractures and severe ventriculomegaly identified in the second trimester of pregnancy. Case Presentation We present here a 2.5-year-old female with profound global developmental delays and spasticity who was found by fetal ultrasound in week 19 of gestation to have bilateral talipes equinovarus and severe bilateral ventriculomegaly. Postnatal genetic testing revealed biallelic variants in KIDINS220. Discussion To our knowledge, this is the first living individual reported with the autosomal recessive form of a KIDINS220-associated condition. This case provides additional information about the postnatal phenotype and a detailed history of development from prenatal ultrasonography.
Collapse
Affiliation(s)
- Lauren I. Brady
- Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Bryon DeFrance
- Department of Obstetrics and Gynecology, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Mark Tarnopolsky
- Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada,*Mark Tarnopolsky,
| |
Collapse
|
13
|
Pellegrino A, Mükusch S, Seitz V, Stein C, Herberg FW, Seitz H. Transient Receptor Potential Vanilloid 1 Signaling Is Independent on Protein Kinase A Phosphorylation of Ankyrin-Rich Membrane Spanning Protein. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2022; 10:medsci10040063. [PMID: 36412904 PMCID: PMC9680306 DOI: 10.3390/medsci10040063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
The sensory ion channel transient receptor potential vanilloid 1 (TRPV1) is mainly expressed in small to medium sized dorsal root ganglion neurons, which are involved in the transfer of acute noxious thermal and chemical stimuli. The Ankyrin-rich membrane spanning protein (ARMS) interaction with TRPV1 is modulated by protein kinase A (PKA) mediating sensitization. Here, we hypothesize that PKA phosphorylation sites of ARMS are crucial for the modulation of TRPV1 function, and that the phosphorylation of ARMS is facilitated by the A-kinase anchoring protein 79 (AKAP79). We used transfected HEK293 cells, immunoprecipitation, calcium flux, and patch clamp experiments to investigate potential PKA phosphorylation sites in ARMS and in ARMS-related peptides. Additionally, experiments were done to discriminate between PKA and protein kinase D (PKD) phosphorylation. We found different interaction ratios for TRPV1 and ARMS mutants lacking PKA phosphorylation sites. The degree of TRPV1 sensitization by ARMS mutants is independent on PKA phosphorylation. AKAP79 was also involved in the TRPV1/ARMS/PKA signaling complex. These data show that ARMS is a PKA substrate via AKAP79 in the TRPV1 signaling complex and that all four proteins interact physically, regulating TRPV1 sensitization in transfected HEK293 cells. To assess the physiological and/or therapeutic significance of these findings, similar investigations need to be performed in native neurons and/or in vivo.
Collapse
Affiliation(s)
- Antonio Pellegrino
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
| | - Sandra Mükusch
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
| | - Viola Seitz
- Institute of Experimental Anaesthesiology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
- Brandenburg Medical School Theodor Fontane, Fehrbelliner Str. 38, 16816 Neuruppin, Germany
| | - Christoph Stein
- Institute of Experimental Anaesthesiology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | | | - Harald Seitz
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
- Correspondence: ; +49-331-58187-208
| |
Collapse
|
14
|
Shen Z, Xiang M, Chen C, Ding F, Wang Y, Shang C, Xin L, Zhang Y, Cui X. Glutamate excitotoxicity: Potential therapeutic target for ischemic stroke. Biomed Pharmacother 2022; 151:113125. [PMID: 35609367 DOI: 10.1016/j.biopha.2022.113125] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/01/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate-mediated excitotoxicity is an important mechanism leading to post ischemic stroke damage. After acute stroke, the sudden reduction in cerebral blood flow is most initially followed by ion transport protein dysfunction and disruption of ion homeostasis, which in turn leads to impaired glutamate release, reuptake, and excessive N-methyl-D-aspartate receptor (NMDAR) activation, promoting neuronal death. Despite extensive evidence from preclinical studies suggesting that excessive NMDAR stimulation during ischemic stroke is a central step in post-stroke damage, NMDAR blockers have failed to translate into clinical stroke treatment. Current treatment options for stroke are very limited, and there is therefore a great need to develop new targets for neuroprotective therapeutic agents in ischemic stroke to extend the therapeutic time window. In this review, we highlight recent findings on glutamate release, reuptake mechanisms, NMDAR and its downstream cellular signaling pathways in post-ischemic stroke damage, and review the pathological changes in each link to help develop viable new therapeutic targets. We then also summarize potential neuroprotective drugs and therapeutic approaches for these new targets in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zihuan Shen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Mi Xiang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chen Chen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Fan Ding
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Yuling Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Chang Shang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Laiyun Xin
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yang Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiangning Cui
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
15
|
Bondeson DP, Paolella BR, Asfaw A, Rothberg MV, Skipper TA, Langan C, Mesa G, Gonzalez A, Surface LE, Ito K, Kazachkova M, Colgan WN, Warren A, Dempster JM, Krill-Burger JM, Ericsson M, Tang AA, Fung I, Chambers ES, Abdusamad M, Dumont N, Doench JG, Piccioni F, Root DE, Boehm J, Hahn WC, Mannstadt M, McFarland JM, Vazquez F, Golub TR. Phosphate dysregulation via the XPR1-KIDINS220 protein complex is a therapeutic vulnerability in ovarian cancer. NATURE CANCER 2022; 3:681-695. [PMID: 35437317 PMCID: PMC9246846 DOI: 10.1038/s43018-022-00360-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
Despite advances in precision medicine, the clinical prospects for patients with ovarian and uterine cancers have not substantially improved. Here, we analyzed genome-scale CRISPR/Cas9 loss-of-function screens across 851 human cancer cell lines and found that frequent overexpression of SLC34A2 – encoding a phosphate importer – is correlated to sensitivity to loss of the phosphate exporter XPR1 in vitro and in vivo. In patient-derived tumor samples, we observed frequent PAX8-dependent overexpression of SLC34A2, XPR1 copy number amplifications, and XPR1 mRNA overexpression. Mechanistically, in SLC34A2-high cancer cell lines, genetic or pharmacologic inhibition of XPR1-dependent phosphate efflux leads to the toxic accumulation of intracellular phosphate. Finally, we show that XPR1 requires the novel partner protein KIDINS220 for proper cellular localization and activity, and that disruption of this protein complex results in acidic vacuolar structures preceding cell death. These data point to the XPR1:KIDINS220 complex and phosphate dysregulation as a therapeutic vulnerability in ovarian cancer. Golub and colleagues identify the phosphate exporter XPR1 as a therapeutic vulnerability in ovarian and uterine cancers, and show that phosphate efflux inhibition reduces tumor cell viability through accumulation of intracellular phosphate.
Collapse
Affiliation(s)
| | - Brenton R Paolella
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Merck Research Laboratories, Cambridge, MA, USA
| | - Adhana Asfaw
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Carly Langan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabriel Mesa
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lauren E Surface
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Kentaro Ito
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | | | | | - Andrew A Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Iris Fung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Mai Abdusamad
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Federica Piccioni
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Merck Research Laboratories, Cambridge, MA, USA
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesse Boehm
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard Medical School, Boston, MA, USA.,Departments of Pediatric and Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | | | - Todd R Golub
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Departments of Pediatric and Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Zhang K, Sun W, Liu Y, Lv Y, Hou D, Lin Y, Xu W, Zhao J, Gai Z, Zhao S, Yuan Y. SINO Syndrome Causative KIDINS220/ARMS Gene Regulates Adipocyte Differentiation. Front Cell Dev Biol 2021; 9:619475. [PMID: 33763417 PMCID: PMC7982959 DOI: 10.3389/fcell.2021.619475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/08/2021] [Indexed: 11/24/2022] Open
Abstract
Nonsense variants in KIDINS220/ARMS were identified as the main cause of spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO) syndrome, a rare disease with birth defects in brachycephaly, neurological disorder, and obesity. The cause of neural cell dysfunction by KIDINS220/ARMS were extensively studied while the cause of obesity in SINO syndrome remains elusive. Here, we identified KIDINS220/ARMS as an adipocyte differentiation-regulating gene. A Chinese family, mother and her two sons, all showed severe symptoms of SINO syndrome. G-banding karyotyping, chromosome microarray analysis, and whole exome sequencing revealed a novel amber mutation, c.3934G>T (p. E1312X), which was close to the C-terminal region of KIDINS220/ARMS and resulted in the premature of the protein. Both the mRNA and protein levels of KIDINS220/ARMS gradually decreased during adipocyte differentiation. Knockdown of KINDINS220/ARMS could prompt adipocyte differentiation and lipid accumulation while overexpression of KIDINS220/ARMS decrease the rate of matured adipocytes. Furthermore, we demonstrated that KIDINS220/ARMS inhibits adipocyte maturation through sustained extracellular signal-regulated kinase signaling. In conclusion, this is the first report about a vertical heredity of severe dominant pathogenic mutation of KIDINS220/ARMS, suggested that KIDINS220/ARMS played a negative role in adipocyte maturation, explained the cause of obesity in SINO syndrome and could highlight the importance of adipocyte differentiation in neuron functions.
Collapse
Affiliation(s)
- Kaihui Zhang
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Wenxing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Yi Liu
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Yuqiang Lv
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Daisen Hou
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Yan Lin
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Wei Xu
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Jianyuan Zhao
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Zhongtao Gai
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Shimin Zhao
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Yiyuan Yuan
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
17
|
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
|
18
|
McMillan KJ, Banks PJ, Hellel FLN, Carmichael RE, Clairfeuille T, Evans AJ, Heesom KJ, Lewis P, Collins BM, Bashir ZI, Henley JM, Wilkinson KA, Cullen PJ. Sorting nexin-27 regulates AMPA receptor trafficking through the synaptic adhesion protein LRFN2. eLife 2021; 10:59432. [PMID: 34251337 PMCID: PMC8296521 DOI: 10.7554/elife.59432] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
The endosome-associated cargo adaptor sorting nexin-27 (SNX27) is linked to various neuropathologies through sorting of integral proteins to the synaptic surface, most notably AMPA receptors. To provide a broader view of SNX27-associated pathologies, we performed proteomics in rat primary neurons to identify SNX27-dependent cargoes, and identified proteins linked to excitotoxicity, epilepsy, intellectual disabilities, and working memory deficits. Focusing on the synaptic adhesion molecule LRFN2, we established that SNX27 binds to LRFN2 and regulates its endosomal sorting. Furthermore, LRFN2 associates with AMPA receptors and knockdown of LRFN2 results in decreased surface AMPA receptor expression, reduced synaptic activity, and attenuated hippocampal long-term potentiation. Overall, our study provides an additional mechanism by which SNX27 can control AMPA receptor-mediated synaptic transmission and plasticity indirectly through the sorting of LRFN2 and offers molecular insight into the perturbed function of SNX27 and LRFN2 in a range of neurological conditions.
Collapse
Affiliation(s)
| | - Paul J Banks
- School of Physiology, Pharmacology and Neuroscience, University of BristolBristolUnited Kingdom
| | | | | | - Thomas Clairfeuille
- Institute for Molecular Bioscience, The University of QueenslandQueenslandAustralia
| | - Ashley J Evans
- School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Kate J Heesom
- Proteomics facility, School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Philip Lewis
- Proteomics facility, School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of QueenslandQueenslandAustralia
| | - Zafar I Bashir
- School of Physiology, Pharmacology and Neuroscience, University of BristolBristolUnited Kingdom
| | - Jeremy M Henley
- School of Biochemistry, University of BristolBristolUnited Kingdom
| | | | - Peter J Cullen
- School of Biochemistry, University of BristolBristolUnited Kingdom
| |
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
|
Kanayama T, Imamura T, Mayumi A, Soma E, Sakamoto K, Hayakawa F, Tanizawa A, Kiyokawa N, Hosoi H. Functional analysis of a novel fusion protein PAX5-KIDINS220 identified in a pediatric Ph-like ALL patient. Int J Hematol 2020; 112:714-719. [PMID: 32656633 DOI: 10.1007/s12185-020-02944-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 01/10/2023]
Abstract
PAX5-KIDINS220 (PAX5-K220) is a novel chimeric fusion gene identified in a pediatric Philadelphia chromosome (Ph)-like acute lymphoblastic leukemia (ALL) patient, but the function of the encoded fusion protein has not yet been analyzed. Here, we report the functional analysis of PAX5-K220 in vitro. We successfully generated PAX5-K220 expressing cells and demonstrate that PAX5-K220 is a nuclear protein. Luciferase reporter assay reveals that PAX5-K220 inhibits wild-type PAX5 transcriptional activity in a dominant-negative fashion like other PAX5-related fusion proteins, and may contribute to lymphocyte differentiation block. However, although identified in Ph-like ALL, PAX5-K220 does not induce IL-3-independent proliferation when transduced in the IL-3-dependent Ba/F3 murine leukemia cells, but rather attenuates growth. These results reveal that PAX5-K220 certainly shares the character with other PAX5-related fusion proteins rather than other fusion proteins with tyrosine kinase activity identified in Ph-like ALL, and did not contribute to proliferation activity. Precise functional analysis of each differently partnered PAX5 fusion protein is warranted in the future for better understanding of PAX5-related translocations and their effects.
Collapse
Affiliation(s)
- Takuyo Kanayama
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Azusa Mayumi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Emi Soma
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kenichi Sakamoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.,Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Fumihiko Hayakawa
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiko Tanizawa
- Department of Human Resource Development for Cancer, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
21
|
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
|
22
|
|
23
|
Genetic and Clinical Profile of Chinese Patients with Autosomal Dominant Spastic Paraplegia. Mol Diagn Ther 2019; 23:781-789. [DOI: 10.1007/s40291-019-00426-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Sebastián-Serrano Á, Simón-García A, Belmonte-Alfaro A, Pose-Utrilla J, Santos-Galindo M, Del Puerto A, García-Guerra L, Hernández IH, Schiavo G, Campanero MR, Lucas JJ, Iglesias T. Differential regulation of Kidins220 isoforms in Huntington's disease. Brain Pathol 2019; 30:120-136. [PMID: 31264746 DOI: 10.1111/bpa.12761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disease characterized by brain atrophy particularly in the striatum that produces motor impairment, and cognitive and psychiatric disturbances. Multiple pathogenic mechanisms have been proposed including dysfunctions in neurotrophic support and calpain-overactivation, among others. Kinase D-interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), is an essential mediator of neurotrophin signaling. In adult brain, Kidins220 presents two main isoforms that differ in their carboxy-terminal length and critical protein-protein interaction domains. These variants are generated through alternative terminal exon splicing of the conventional exon 32 (Kidins220-C32) and the recently identified exon 33 (Kidins220-C33). The lack of domains encoded by exon 32 involved in key neuronal functions, including those controlling neurotrophin pathways, pointed to Kidins220-C33 as a form detrimental for neurons. However, the functional role of Kidins220-C33 in neurodegeneration or other pathologies, including HD, has not been explored. In the present work, we discover an unexpected selective downregulation of Kidins220-C33, in the striatum of HD patients, as well as in the R6/1 HD mouse model starting at early symptomatic stages. These changes are C33-specific as Kidins220-C32 variant remains unchanged. We also find the early decrease in Kidins220-C33 levels takes place in neurons, suggesting an unanticipated neuroprotective role for this isoform. Finally, using ex vivo assays and primary neurons, we demonstrate that Kidins220-C33 is downregulated by mechanisms that depend on the activation of the protease calpain. Altogether, these results strongly suggest that calpain-mediated Kidins220-C33 proteolysis modulates onset and/or progression of HD.
Collapse
Affiliation(s)
- Álvaro Sebastián-Serrano
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Simón-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alicia Belmonte-Alfaro
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 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 "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - María Santos-Galindo
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Ana Del Puerto
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucía García-Guerra
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ivó H Hernández
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain.,Facultad de Ciencias, Departamento de Biología (Unidad Docente Fisiología Animal), Universidad Autónoma de Madrid, Madrid, Spain
| | - Giampietro Schiavo
- Department of Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, UK
| | - Miguel R Campanero
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José J Lucas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
25
|
López-Menéndez C, Simón-García A, Gamir-Morralla A, Pose-Utrilla J, Luján R, Mochizuki N, Díaz-Guerra M, Iglesias T. Excitotoxic targeting of Kidins220 to the Golgi apparatus precedes calpain cleavage of Rap1-activation complexes. Cell Death Dis 2019; 10:535. [PMID: 31296845 PMCID: PMC6624258 DOI: 10.1038/s41419-019-1766-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/30/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022]
Abstract
Excitotoxic neuronal death induced by high concentrations of glutamate is a pathological event common to multiple acute or chronic neurodegenerative diseases. Excitotoxicity is mediated through overactivation of the N-Methyl-D-aspartate type of ionotropic glutamate receptors (NMDARs). Physiological stimulation of NMDARs triggers their endocytosis from the neuronal surface, inducing synaptic activity and survival. However almost nothing is known about the internalization of overactivated NMDARs and their interacting proteins, and how this endocytic process is connected with neuronal death has been poorly explored. Kinase D-interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), is a component of NMDAR complexes essential for neuronal viability by the control of ERK activation. Here we have investigated Kidins220 endocytosis induced by NMDAR overstimulation and the participation of this internalization step in the molecular mechanisms of excitotoxicity. We show that excitotoxicity induces Kidins220 and GluN1 traffic to the Golgi apparatus (GA) before Kidins220 is degraded by the protease calpain. We also find that excitotoxicity triggers an early activation of Rap1-GTPase followed by its inactivation. Kidins220 excitotoxic endocytosis and subsequent calpain-mediated downregulation governs this late inactivation of Rap1 that is associated to decreases in ERK activity preceding neuronal death. Furthermore, we identify the molecular mechanisms involved in the excitotoxic shutoff of Kidins220/Rap1/ERK prosurvival cascade that depends on calpain processing of Rap1-activation complexes. Our data fit in a model where Kidins220 targeting to the GA during early excitotoxicity would facilitate Rap1 activation and subsequent stimulation of ERK. At later times, activation of Golgi-associated calpain, would promote the degradation of GA-targeted Kidins220 and two additional components of the specific Rap1 activation complex, PDZ-GEF1, and S-SCAM. In this way, late excitotoxicity would turn off Rap1/ERK cascade and compromise neuronal survival.
Collapse
Affiliation(s)
- 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, C/ Valderrebollo, 5, 28031, 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, C/ Valderrebollo, 5, 28031, Madrid, Spain
| | - Andrea Gamir-Morralla
- 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, C/ Valderrebollo, 5, 28031, Madrid, Spain.,Institute of Physiological Chemistry, University Medical Center, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128, Mainz, Germany
| | - 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, C/ Valderrebollo, 5, 28031, Madrid, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Dept. Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/ Almansa 14, 02008, Albacete, Spain
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishiro-dai, Suita, 565-8565, Osaka, Japan
| | - Margarita Díaz-Guerra
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, 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, C/ Valderrebollo, 5, 28031, Madrid, Spain.
| |
Collapse
|
26
|
Cesca F, Schiavo G, Benfenati F. Kidins220/ARMS transgenic lines could be instrumental in the understanding of the molecular mechanisms leading to spastic paraplegia and obesity. Eur J Neurol 2018; 25:e107. [DOI: 10.1111/ene.13693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/23/2018] [Indexed: 11/30/2022]
Affiliation(s)
- F. Cesca
- Istituto Italiano di Tecnologia; Center for Synaptic Neuroscience and Technology; Genova Italy
- IRCCS Ospedale Policlinico San Martino; Genova Italy
| | | | - F. Benfenati
- Istituto Italiano di Tecnologia; Center for Synaptic Neuroscience and Technology; Genova Italy
- IRCCS Ospedale Policlinico San Martino; Genova Italy
| |
Collapse
|
27
|
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
|
28
|
Yang L, Zhang W, Peng J, Yin F. Heterozygous KIDINS220 mutation leads to spastic paraplegia and obesity in an Asian girl. Eur J Neurol 2018; 25:e53-e54. [PMID: 29667355 DOI: 10.1111/ene.13600] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 02/07/2018] [Indexed: 11/30/2022]
Affiliation(s)
- L. Yang
- Department of Pediatrics; XiangYa Hospital; Central South University; Changsha China
| | - W. Zhang
- Department of Pediatrics; XiangYa Hospital; Central South University; Changsha China
| | - J. Peng
- Department of Pediatrics; XiangYa Hospital; Central South University; Changsha China
| | - F. Yin
- Department of Pediatrics; XiangYa Hospital; Central South University; Changsha China
| |
Collapse
|
29
|
Abstract
NMDA (N-methyl-d-aspartate) receptors (NMDARs) play a central role in excitotoxic neuronal death caused by ischemic stroke, but NMDAR channel blockers have failed to be translated into clinical stroke treatments. However, recent research on NMDAR-associated signaling complexes has identified important death-signaling pathways linked to NMDARs. This led to the generation of inhibitors that inhibit these pathways downstream from the receptor without necessarily blocking NMDARs. This therapeutic approach may have fewer side effects and/or provide a wider therapeutic window for stroke as compared to the receptor antagonists. In this review, we highlight the key findings in the signaling cascades downstream of NMDARs and the novel promising therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Qiu Jing Wu
- Krembil Research Institute, University Health Network, 60 Leonard St, Toronto, ON, M5T2S8, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Michael Tymianski
- Krembil Research Institute, University Health Network, 60 Leonard St, Toronto, ON, M5T2S8, Canada. .,Department of Physiology, University of Toronto, Toronto, ON, Canada. .,Division of Neurosurgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Gamir-Morralla A, Belbin O, Fortea J, Alcolea D, Ferrer I, Lleó A, Iglesias T. Kidins220 Correlates with Tau in Alzheimer's Disease Brain and Cerebrospinal Fluid. J Alzheimers Dis 2018; 55:1327-1333. [PMID: 27858709 DOI: 10.3233/jad-160639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Identification of neurodegeneration-monitoring biomarkers would be of great clinical value for Alzheimer's disease (AD) diagnosis. Using N- or C-terminal antibodies, we studied the pro-survival synaptic effector, Kidins220, in the brain and cerebrospinal fluid (CSF) of controls and AD patients. Only the N-terminal antibody showed a positive correlation between Kidins220 and phosphorylated tau in AD brains. Using this antibody, Kidins220 was detected in CSF from AD patients where it positively correlated with CSF phosphorylated tau and tau. This study highlights the potential of Kidins220 as a CSF biomarker in AD.
Collapse
Affiliation(s)
- Andrea Gamir-Morralla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Olivia Belbin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Alcolea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Isidro Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Wang Y, Shao N, Mao X, Zhu M, Fan W, Shen Z, Xiao R, Wang C, Bao W, Xu X, Yang C, Dong J, Yu D, Wu Y, Zhu C, Wen L, Lu X, Lu YJ, Feng N. MiR-4638-5p inhibits castration resistance of prostate cancer through repressing Kidins220 expression and PI3K/AKT pathway activity. Oncotarget 2018; 7:47444-47464. [PMID: 27329728 PMCID: PMC5216953 DOI: 10.18632/oncotarget.10165] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/06/2016] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are short, conserved segments of non-coding RNA which play a significant role in prostate cancer development and progression. To identify miRNAs associated with castration resistance, we performed miRNA microarray analysis comparing castration resistant prostate cancer (CRPC) with androgen dependent prostate cancer (ADPC). We identified common underexpression of miR-4638-5p in CRPC compared to ADPC samples, which were further confirmed by quantitative PCR analysis. The role of miR-4638-5p in prostate cancer androgen-independent growth has been demonstrated both in vitro and in vivo. We also identified Kidins220 as a target gene directly regulated by miR-4638-5p and shRNA-mediated knockdown of Kidins220 phenocopied miR-4638-5p restoration. Subsequently, we revealed that Kidins220 activates PI3K/AKT pathway, which plays a key role in CRPC. Loss of miR- 4638-5p may lead to CRPC through the activity of Kidins220 and PI3K/AKT pathway. Furthermore, we found that miR-4638-5p, through regulating Kidins220 and the downstream activity of VEGF and PI3K/AKT pathway, influences prostate cancer progression via angiogenesis. The identification of miR-4638-5p down-regulation in CRPC and the understanding of the functional role of miR-4638-5p and its downstream genes/pathways have the potential to develop biomarkers for CRPC onset and to identify novel targets for novel forms of treatments of this lethal form of PCa.
Collapse
Affiliation(s)
- Yang Wang
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Ning Shao
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Xueying Mao
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Minmin Zhu
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Weifei Fan
- Jiangsu Province Geriatric Institute, Nanjing, China
| | - Zhixiang Shen
- Jiangsu Province Geriatric Institute, Nanjing, China
| | - Rong Xiao
- College of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Chuncai Wang
- College of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Wenping Bao
- College of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xinyu Xu
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Chun Yang
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Jian Dong
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Deshui Yu
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Yan Wu
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Caixia Zhu
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Liting Wen
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Xiaojie Lu
- Centre for Translational Medicine, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Yong-Jie Lu
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| |
Collapse
|
32
|
Raza MZ, Allegrini S, Dumontet C, Jordheim LP. Functions of the multi-interacting protein KIDINS220/ARMS in cancer and other pathologies. Genes Chromosomes Cancer 2017; 57:114-122. [PMID: 29181864 DOI: 10.1002/gcc.22514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/07/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022] Open
Abstract
Development of an organ and subsequently the whole system from an embryo is a highly integrated process. Although there is evidence that different systems are interconnected during developmental stages, the molecular understanding of this relationship is either not known or only to a limited extent. Nervous system development, amongst all, is maybe the most crucial and complex process. It relies on the correct distribution of specific neuronal growth factors and hormones to the specific receptors. Among the plethora of proteins that are involved in downstream signalling of neuronal growth factors, we find the kinase-D interacting substrate of 220 kDa (KIDINS220), also known as ankyrin-rich repeat membrane spanning (ARMS) protein. KIDINS220 has been shown to play a substantial role in the nervous system and vascular system development as well as in neuronal survival and differentiation. It serves as a downstream regulator for many important neuronal and vascular growth factors such as vascular endothelial growth factor (VEGF), the neurotrophin family, glutamate receptors and ephrin receptors. Moreover, activation and differentiation of B- and T-cells, as well as tumour cell proliferation has also shown to be related to KIDINS220. This review comprehensively summarises the existing research data on this protein, with a particular interest in its role in cancer and in other pathologies.
Collapse
Affiliation(s)
- Muhammad-Zawwad Raza
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| | - Simone Allegrini
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
| | - Charles Dumontet
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| | - Lars Petter Jordheim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| |
Collapse
|
33
|
SNX27 links DGKζ to the control of transcriptional and metabolic programs in T lymphocytes. Sci Rep 2017; 7:16361. [PMID: 29180720 PMCID: PMC5703713 DOI: 10.1038/s41598-017-16370-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/10/2017] [Indexed: 01/10/2023] Open
Abstract
Sorting nexin 27 (SNX27) recycles PSD-95, Dlg1, ZO-1 (PDZ) domain-interacting membrane proteins and is essential to sustain adequate brain functions. Here we define a fundamental SNX27 function in T lymphocytes controlling antigen-induced transcriptional activation and metabolic reprogramming. SNX27 limits the activation of diacylglycerol (DAG)-based signals through its high affinity PDZ-interacting cargo DAG kinase ζ (DGKζ). SNX27 silencing in human T cells enhanced T cell receptor (TCR)-stimulated activator protein 1 (AP-1)- and nuclear factor κB (NF-κB)-mediated transcription. Transcription did not increase upon DGKζ silencing, suggesting that DGKζ function is dependent on SNX27. The enhanced transcriptional activation in SNX27-silenced cells contrasted with defective activation of the mammalian target of rapamycin (mTOR) pathway. The analysis of Snx27−/− mice supported a role for SNX27 in the control of T cell growth. This study broadens our understanding of SNX27 as an integrator of lipid-based signals with the control of transcription and metabolic pathways.
Collapse
|
34
|
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
|
35
|
Peter J, Kasper C, Kaufholz M, Buschow R, Isensee J, Hucho T, Herberg FW, Schwede F, Stein C, Jordt SE, Brackmann M, Spahn V. Ankyrin-rich membrane spanning protein as a novel modulator of transient receptor potential vanilloid 1-function in nociceptive neurons. Eur J Pain 2017; 21:1072-1086. [PMID: 28182310 DOI: 10.1002/ejp.1008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2016] [Indexed: 02/01/2023]
Abstract
BACKGROUND The ion channel TRPV1 is mainly expressed in small diameter dorsal root ganglion (DRG) neurons, which are involved in the sensation of acute noxious thermal and chemical stimuli. Direct modifications of the channel by diverse signalling events have been intensively investigated, but little is known about the composition of modulating macromolecular TRPV1 signalling complexes. Here, we hypothesize that the novel adaptor protein ankyrin-rich membrane spanning protein/kinase D interacting substrate (ARMS) interacts with TRPV1 and modulates its function in rodent DRG neurons. METHODS We used immunohistochemistry, electrophysiology, microfluorimetry and immunoprecipitation experiments to investigate TRPV1 and ARMS interactions in DRG neurons and transfected cells. RESULTS We found that TRPV1 and ARMS are co-expressed in a subpopulation of DRG neurons. ARMS sensitizes TRPV1 towards capsaicin in transfected HEK 293 cells and in mouse DRG neurons in a PKA-dependent manner. Using a combination of functional imaging and immunocytochemistry, we show that the magnitude of the capsaicin response in DRG neurons depends not only on TRPV1 expression, but on the co-expression of ARMS alongside TRPV1. CONCLUSION These data indicate that ARMS is an important component of the signalling complex regulating the sensitivity of TRPV1. SIGNIFICANCE The study identifies ARMS as an important component of the signalling complex regulating the sensitivity of excitatory ion channels (TRPV1) in peripheral sensory neurons (DRG neurons) and transfected cells.
Collapse
Affiliation(s)
- J Peter
- Department of Anesthesiology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Germany
| | - C Kasper
- Department of Anesthesiology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Germany
| | - M Kaufholz
- Department of Biochemistry, University of Kassel, Germany
| | - R Buschow
- Department Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - J Isensee
- Department Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Germany
| | - T Hucho
- Department Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Germany
| | - F W Herberg
- Department of Biochemistry, University of Kassel, Germany
| | - F Schwede
- Biolog Life Science Institute, Bremen, Germany
| | - C Stein
- Department of Anesthesiology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Germany
| | - S-E Jordt
- Department of Pharmacology, Yale Medical School, New Haven, CT, USA
- Department of Anesthesiology, Clinical Science Department, Duke University, Durham, NC, USA
| | - M Brackmann
- Department of Anesthesiology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Germany
| | - V Spahn
- Department of Anesthesiology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
36
|
Sakamoto K, Imamura T, Kanayama T, Yano M, Asai D, Deguchi T, Hashii Y, Tanizawa A, Ohshima Y, Kiyokawa N, Horibe K, Sato A. Ph-like acute lymphoblastic leukemia with a novel PAX5-KIDINS220 fusion transcript. Genes Chromosomes Cancer 2016; 56:278-284. [PMID: 27870151 DOI: 10.1002/gcc.22433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 01/19/2023] Open
Abstract
Although "paired box 5" (PAX5)-related fusion genes are well documented in childhood B-cell precursor acute lymphoblastic leukemia (ALL), these types of fusion with the exception of PAX5-JAK2 are rarely seen in patients with gene expression profiles similar to those of BCR-ABL1 (Philadelphia)-positive ALL (Ph-like ALL). We report a novel fusion of the genes PAX5 and "kinase D-interacting substrate of 220 kDa" (KIDINS220, also known as ARMS) in a Ph-like ALL. As PAX5 is a master regulator of B-lymphocyte differentiation, PAX5 rearrangements induce a differentiation block in B lymphocytes. KIDINS220 is a mediator of multiple receptor signaling pathways, interacts with both T- and B-cell receptors, and is necessary for sustained extracellular signal-regulated kinase (ERK) signaling. Although functional studies are needed, the PAX5-KIDINS220 fusion protein might not only inhibit wild-type PAX5 function, but also promote sustained activation of the ERK signaling pathway through upregulation of KIDINS220. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Japan
| | - Takuyo Kanayama
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Japan
| | - Mio Yano
- Department of Pediatrics, National Hospital Organization Maizuru Medical Center, Japan
| | - Daisuke Asai
- Department of Pediatrics, Japanese Red Cross Kyoto Daini Hospital, Japan
| | | | | | | | - Yusei Ohshima
- Department of Pediatrics, University of Fukui, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Japan
| | - Atsushi Sato
- Department of Hematology/Oncology, Miyagi Children's Hospital, Japan
| |
Collapse
|
37
|
Curcio M, Salazar IL, Mele M, Canzoniero LMT, Duarte CB. Calpains and neuronal damage in the ischemic brain: The swiss knife in synaptic injury. Prog Neurobiol 2016; 143:1-35. [PMID: 27283248 DOI: 10.1016/j.pneurobio.2016.06.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022]
Abstract
The excessive extracellular accumulation of glutamate in the ischemic brain leads to an overactivation of glutamate receptors with consequent excitotoxic neuronal death. Neuronal demise is largely due to a sustained activation of NMDA receptors for glutamate, with a consequent increase in the intracellular Ca(2+) concentration and activation of calcium- dependent mechanisms. Calpains are a group of Ca(2+)-dependent proteases that truncate specific proteins, and some of the cleavage products remain in the cell, although with a distinct function. Numerous studies have shown pre- and post-synaptic effects of calpains on glutamatergic and GABAergic synapses, targeting membrane- associated proteins as well as intracellular proteins. The resulting changes in the presynaptic proteome alter neurotransmitter release, while the cleavage of postsynaptic proteins affects directly or indirectly the activity of neurotransmitter receptors and downstream mechanisms. These alterations also disturb the balance between excitatory and inhibitory neurotransmission in the brain, with an impact in neuronal demise. In this review we discuss the evidence pointing to a role for calpains in the dysregulation of excitatory and inhibitory synapses in brain ischemia, at the pre- and post-synaptic levels, as well as the functional consequences. Although targeting calpain-dependent mechanisms may constitute a good therapeutic approach for stroke, specific strategies should be developed to avoid non-specific effects given the important regulatory role played by these proteases under normal physiological conditions.
Collapse
Affiliation(s)
- Michele Curcio
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ivan L Salazar
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Miranda Mele
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal.
| |
Collapse
|
38
|
Josifova DJ, Monroe GR, Tessadori F, de Graaff E, van der Zwaag B, Mehta SG, Harakalova M, Duran KJ, Savelberg SMC, Nijman IJ, Jungbluth H, Hoogenraad CC, Bakkers J, Knoers NV, Firth HV, Beales PL, van Haaften G, van Haelst MM. Heterozygous KIDINS220/ARMS nonsense variants cause spastic paraplegia, intellectual disability, nystagmus, and obesity. Hum Mol Genet 2016; 25:2158-2167. [PMID: 27005418 DOI: 10.1093/hmg/ddw082] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/10/2016] [Indexed: 12/13/2022] Open
Abstract
We identified de novo nonsense variants in KIDINS220/ARMS in three unrelated patients with spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO). KIDINS220 is an essential scaffold protein coordinating neurotrophin signal pathways in neurites and is spatially and temporally regulated in the brain. Molecular analysis of patients' variants confirmed expression and translation of truncated transcripts similar to recently characterized alternative terminal exon splice isoforms of KIDINS220 KIDINS220 undergoes extensive alternative splicing in specific neuronal populations and developmental time points, reflecting its complex role in neuronal maturation. In mice and humans, KIDINS220 is alternative spliced in the middle region as well as in the last exon. These full-length and KIDINS220 splice variants occur at precise moments in cortical, hippocampal, and motor neuron development, with splice variants similar to the variants seen in our patients and lacking the last exon of KIDINS220 occurring in adult rather than in embryonic brain. We conducted tissue-specific expression studies in zebrafish that resulted in spasms, confirming a functional link with disruption of the KIDINS220 levels in developing neurites. This work reveals a crucial physiological role of KIDINS220 in development and provides insight into how perturbation of the complex interplay of KIDINS220 isoforms and their relative expression can affect neuron control and human metabolism. Altogether, we here show that de novo protein-truncating KIDINS220 variants cause a new syndrome, SINO. This is the first report of KIDINS220 variants causing a human disease.
Collapse
Affiliation(s)
- Dragana J Josifova
- Department of Clinical Genetics, Guys' and St. Thomas' Hospital, London SE1 7EH, UK
| | - Glen R Monroe
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Federico Tessadori
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands
| | - Esther de Graaff
- Division of Cell Biology, Faculty of Science, University of Utrecht, Utrecht 3584 CH, The Netherlands
| | | | - Sarju G Mehta
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | | | | | - Karen J Duran
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Sanne M C Savelberg
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Isaäc J Nijman
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Heinz Jungbluth
- Department of Paediatric Neurology, Evelina Children's Hospital, Guy's & St Thomas' Hospital NHS Foundation Trust, London SE1 7EH, UK Randall Division of Cell and Molecular Biophysics, Muscle Signalling Section, Department of Basic and Clinical Neuroscience, IoPPN, King's College, London WC2R 2LS, UK
| | - Casper C Hoogenraad
- Division of Cell Biology, Faculty of Science, University of Utrecht, Utrecht 3584 CH, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands Department of Medical Physiology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Nine V Knoers
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Helen V Firth
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1RQ, UK
| | - Philip L Beales
- Genetics and Genomics Medicine Program, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Gijs van Haaften
- Department of Genetics Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | | |
Collapse
|
39
|
Scholz-Starke J, Cesca F. Stepping Out of the Shade: Control of Neuronal Activity by the Scaffold Protein Kidins220/ARMS. Front Cell Neurosci 2016; 10:68. [PMID: 27013979 PMCID: PMC4789535 DOI: 10.3389/fncel.2016.00068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/01/2016] [Indexed: 12/31/2022] Open
Abstract
The correct functioning of the nervous system depends on the exquisitely fine control of neuronal excitability and synaptic plasticity, which relies on an intricate network of protein-protein interactions and signaling that shapes neuronal homeostasis during development and in adulthood. In this complex scenario, Kinase D interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning (Kidins220/ARMS) acts as a multi-functional scaffold protein with preferential expression in the nervous system. Engaged in a plethora of interactions with membrane receptors, cytosolic signaling components and cytoskeletal proteins, Kidins220/ARMS is implicated in numerous cellular functions including neuronal survival, neurite outgrowth and maturation and neuronal activity, often in the context of neurotrophin (NT) signaling pathways. Recent studies have highlighted a number of cell- and context-specific roles for this protein in the control of synaptic transmission and neuronal excitability, which are at present far from being completely understood. In addition, some evidence has began to emerge, linking alterations of Kidins220 expression to the onset of various neurodegenerative diseases and neuropsychiatric disorders. In this review, we present a concise summary of our fragmentary knowledge of Kidins220/ARMS biological functions, focusing on the mechanism(s) by which it controls various aspects of neuronal activity. We have tried, where possible, to discuss the available evidence in the wider context of NT-mediated regulation, and to outline emerging roles of Kidins220/ARMS in human pathologies.
Collapse
Affiliation(s)
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
40
|
López-Benito S, Lillo C, Hernández-Hernández Á, Chao MV, Arévalo JC. ARMS/Kidins220 and synembryn-B levels regulate NGF-mediated secretion. J Cell Sci 2016; 129:1866-77. [PMID: 26966186 DOI: 10.1242/jcs.184168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 03/05/2016] [Indexed: 01/22/2023] Open
Abstract
Proper development of the nervous system requires a temporally and spatially orchestrated set of events including differentiation, synapse formation and neurotransmission. Nerve growth factor (NGF) acting through the TrkA neurotrophin receptor (also known as NTRK1) regulates many of these events. However, the molecular mechanisms responsible for NGF-regulated secretion are not completely understood. Here, we describe a new signaling pathway involving TrkA, ARMS (also known as Kidins220), synembryn-B and Rac1 in NGF-mediated secretion in PC12 cells. Whereas overexpression of ARMS blocked NGF-mediated secretion, without affecting basal secretion, a decrease in ARMS resulted in potentiation. Similar effects were observed with synembryn-B, a protein that interacts directly with ARMS. Downstream of ARMS and synembryn-B are Gαq and Trio proteins, which modulate the activity of Rac1 in response to NGF. Expression of dominant-negative Rac1 rescued the secretion defects of cells overexpressing ARMS or synembryn-B. Thus, this neurotrophin pathway represents a new mechanism responsible for NGF-regulated secretion.
Collapse
Affiliation(s)
- Saray López-Benito
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), University of Salamanca, Salamanca 37007, Spain Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Concepción Lillo
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), University of Salamanca, Salamanca 37007, Spain Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Ángel Hernández-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| | - Moses V Chao
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology and Neuroscience, Psychiatry, and Neural Sciences, New York University School of Medicine, New York, NY 10016, USA
| | - Juan C Arévalo
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), University of Salamanca, Salamanca 37007, Spain Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| |
Collapse
|
41
|
Kranz TM, Berns A, Shields J, Rothman K, Walsh-Messinger J, Goetz RR, Chao MV, Malaspina D. Phenotypically distinct subtypes of psychosis accompany novel or rare variants in four different signaling genes. EBioMedicine 2016; 6:206-214. [PMID: 27211562 PMCID: PMC4856793 DOI: 10.1016/j.ebiom.2016.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/03/2016] [Accepted: 03/06/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Rare gene variants are important sources of schizophrenia vulnerability that likely interact with polygenic susceptibility loci. This study examined if novel or rare missense coding variants in any of four different signaling genes in sporadic schizophrenia cases were associated with clinical phenotypes in an exceptionally well-characterized sample. METHOD Structured interviews, cognition, symptoms and life course features were assessed in 48 ethnically-diverse cases with psychosis who underwent targeted exome sequencing of PTPRG (Protein Tyrosine Phosphatase, Receptor Type G), SLC39A13 (Solute Carrier Family 39 (Zinc Transporter) Member 13), TGM5 (transglutaminase 5) and ARMS/KIDINS220 (Ankyrin repeat-rich membrane spanning protein or Kinase D-Interacting Substrate of 220kDa). Cases harboring rare missense coding polymorphisms or novel mutations in one or more of these genes were compared to other cases not carrying any rare missense coding polymorphisms or novel mutations in these genes and healthy controls. FINDINGS Fifteen of 48 cases (31.25%) carried rare or novel missense coding variants in one or more of these genes. The subgroups significantly differed in important features, including specific working memory deficits for PTPRG (n=5); severe negative symptoms, global cognitive deficits and poor educational attainment, suggesting a developmental disorder, for SLC39A13 (n=4); slow processing speed, childhood attention deficit disorder and milder symptoms for TGM5 (n=4); and global cognitive deficits with good educational attainment suggesting neurodegeneration for ARMS/KIDINS220 (n=5). Case vignettes are included in the appendix. INTERPRETATION Genes prone to missense coding polymorphisms and/or mutations in sporadic cases may highlight influential genes for psychosis and illuminate heterogeneous pathways to schizophrenia. Ethnicity appears less important at the level of genetic variability. The sequence variations that potentially alter the function of specific genes or their signaling partners may contribute to particular subtypes of psychosis. This approach may be applicable to other complex disorders.
Collapse
Affiliation(s)
- Thorsten M Kranz
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, NY, NY 10016, USA.
| | - Adam Berns
- Institute for Social and Psychiatric Initiatives (InSPIRES), Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| | - Jerry Shields
- Institute for Social and Psychiatric Initiatives (InSPIRES), Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| | - Karen Rothman
- Institute for Social and Psychiatric Initiatives (InSPIRES), Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| | | | - Raymond R Goetz
- New York State Psychiatric Institute, Division of Clinical Phenomenology, 1051 Riverside Drive, New York, NY 10032, USA.
| | - Moses V Chao
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, NY, NY 10016, USA.
| | - Dolores Malaspina
- Institute for Social and Psychiatric Initiatives (InSPIRES), Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
42
|
Development of a neuroprotective peptide that preserves survival pathways by preventing Kidins220/ARMS calpain processing induced by excitotoxicity. Cell Death Dis 2015; 6:e1939. [PMID: 26492372 PMCID: PMC4632323 DOI: 10.1038/cddis.2015.307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/19/2015] [Accepted: 09/14/2015] [Indexed: 11/23/2022]
Abstract
Kinase D-interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), has a central role in the coordination of receptor crosstalk and the integration of signaling pathways essential for neuronal differentiation, survival and function. This protein is a shared downstream effector for neurotrophin- and ephrin-receptors signaling that also interacts with the N-methyl-d-aspartate type of glutamate receptors (NMDARs). Failures in neurotrophic support and glutamate signaling are involved in pathologies related to excitotoxicity and/or neurodegeneration, where different components of these dynamic protein complexes result altered by a combination of mechanisms. In the case of Kidins220/ARMS, overactivation of NMDARs in excitotoxicity and cerebral ischemia triggers its downregulation, which contributes to neuronal death. This key role in neuronal life/death decisions encouraged us to investigate Kidins220/ARMS as a novel therapeutic target for neuroprotection. As the main mechanism of Kidins220/ARMS downregulation in excitotoxicity is proteolysis by calpain, we decided to develop cell-penetrating peptides (CPPs) that could result in neuroprotection by interference of this processing. To this aim, we first analyzed in detail Kidins220/ARMS cleavage produced in vitro and in vivo, identifying a major calpain processing site in its C-terminal region (between amino acids 1669 and 1670) within a sequence motif highly conserved in vertebrates. Then, we designed a 25-amino acids CPP (Tat-K) containing a short Kidins220/ARMS sequence enclosing the identified calpain site (amino acids 1668–1681) fused to the HIV-1 Tat protein basic domain, able to confer membrane permeability to attached cargoes. Transduction of cortical neurons with Tat-K reduced Kidins220/ARMS calpain processing in a dose- and time-dependent manner upon excitotoxic damage and allowed preservation of the activity of pERK1/2 and pCREB, signaling molecules central to neuronal survival and functioning. Importantly, these effects were associated to a significant increase in neuronal viability. This Kidins220/ARMS-derived peptide merits further research to develop novel neuroprotective therapies for excitotoxicity-associated pathologies.
Collapse
|
43
|
Kranz TM, Goetz RR, Walsh-Messinger J, Goetz D, Antonius D, Dolgalev I, Heguy A, Seandel M, Malaspina D, Chao MV. Rare variants in the neurotrophin signaling pathway implicated in schizophrenia risk. Schizophr Res 2015; 168. [PMID: 26215504 PMCID: PMC4591185 DOI: 10.1016/j.schres.2015.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Multiple lines of evidence corroborate impaired signaling pathways as relevant to the underpinnings of schizophrenia. There has been an interest in neurotrophins, since they are crucial mediators of neurodevelopment and in synaptic connectivity in the adult brain. Neurotrophins and their receptors demonstrate aberrant expression patterns in cortical areas for schizophrenia cases in comparison to control subjects. There is little known about the contribution of neurotrophin genes in psychiatric disorders. To begin to address this issue, we conducted high-coverage targeted exome capture in a subset of neurotrophin genes in 48 comprehensively characterized cases with schizophrenia-related psychosis. We herein report rare missense polymorphisms and novel missense mutations in neurotrophin receptor signaling pathway genes. Furthermore, we observed that several genes have a higher propensity to harbor missense coding variants than others. Based on this initial analysis we suggest that rare variants and missense mutations in neurotrophin genes might represent genetic contributions involved across psychiatric disorders.
Collapse
Affiliation(s)
- Thorsten M. Kranz
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, New York, NY 10016, USA
| | - Ray R. Goetz
- New York State Psychiatric Institute, Division of Clinical Phenomenology, 1051 Riverside Drive, New York, NY 10032, USA and Columbia University, Department of Psychiatry, New York, NY 10032, USA
| | - Julie Walsh-Messinger
- Mental Illness, Research, Education, and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY 10468, USA and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deborah Goetz
- Department of Psychiatry, Social and Psychiatric Initiatives, New York University. 1 Park Avenue, 8th Floor Room 222, New York, NY 10016, USA
| | - Daniel Antonius
- University at Buffalo, Department of Psychiatry, Buffalo, NY, 14215, USA
| | - Igor Dolgalev
- Genome Technology Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Marco Seandel
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Dolores Malaspina
- Department of Psychiatry, Social and Psychiatric Initiatives, New York University. 1 Park Avenue, 8th Floor Room 222, New York, NY 10016, USA
| | - Moses V. Chao
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, New York, NY 10016, USA
| |
Collapse
|
44
|
Meta-Analysis of Microarray Data of Rainbow Trout Fry Gonad Differentiation Modulated by Ethynylestradiol. PLoS One 2015; 10:e0135799. [PMID: 26379055 PMCID: PMC4574709 DOI: 10.1371/journal.pone.0135799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 07/27/2015] [Indexed: 01/25/2023] Open
Abstract
Sex differentiation in fish is a highly labile process easily reversed by the use of exogenous hormonal treatment and has led to environmental concerns since low doses of estrogenic molecules can adversely impact fish reproduction. The goal of this study was to identify pathways altered by treatment with ethynylestradiol (EE2) in developing fish and to find new target genes to be tested further for their possible role in male-to-female sex transdifferentiation. To this end, we have successfully adapted a previously developed bioinformatics workflow to a meta-analysis of two datasets studying sex reversal following exposure to EE2 in juvenile rainbow trout. The meta-analysis consisted of retrieving the intersection of the top gene lists generated for both datasets, performed at different levels of stringency. The intersecting gene lists, enriched in true positive differentially expressed genes (DEGs), were subjected to over-representation analysis (ORA) which allowed identifying several statistically significant enriched pathways altered by EE2 treatment and several new candidate pathways, such as progesterone-mediated oocyte maturation and PPAR signalling. Moreover, several relevant key genes potentially implicated in the early transdifferentiation process were selected. Altogether, the results show that EE2 has a great effect on gene expression in juvenile rainbow trout. The feminization process seems to result from the altered transcription of genes implicated in normal female gonad differentiation, resulting in expression similar to that observed in normal females (i.e. the repression of key testicular markers cyp17a1, cyp11b, tbx1), as well as from other genes (including transcription factors) that respond specifically to the EE2 treatment. The results also showed that the bioinformatics workflow can be applied to different types of microarray platforms and could be generalized to (eco)toxicogenomics studies for environmental risk assessment purposes.
Collapse
|
45
|
Fiala GJ, Janowska I, Prutek F, Hobeika E, Satapathy A, Sprenger A, Plum T, Seidl M, Dengjel J, Reth M, Cesca F, Brummer T, Minguet S, Schamel WWA. Kidins220/ARMS binds to the B cell antigen receptor and regulates B cell development and activation. ACTA ACUST UNITED AC 2015; 212:1693-708. [PMID: 26324445 PMCID: PMC4577850 DOI: 10.1084/jem.20141271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/14/2015] [Indexed: 01/04/2023]
Abstract
Fiala et al. report that Kidins220/ARMS is a novel interactor of the B cell antigen receptor (BCR) and its deletion impairs B cell development and B cell functioning. B cell antigen receptor (BCR) signaling is critical for B cell development and activation. Using mass spectrometry, we identified a protein kinase D–interacting substrate of 220 kD (Kidins220)/ankyrin repeat–rich membrane-spanning protein (ARMS) as a novel interaction partner of resting and stimulated BCR. Upon BCR stimulation, the interaction increases in a Src kinase–independent manner. By knocking down Kidins220 in a B cell line and generating a conditional B cell–specific Kidins220 knockout (B-KO) mouse strain, we show that Kidins220 couples the BCR to PLCγ2, Ca2+, and extracellular signal-regulated kinase (Erk) signaling. Consequently, BCR-mediated B cell activation was reduced in vitro and in vivo upon Kidins220 deletion. Furthermore, B cell development was impaired at stages where pre-BCR or BCR signaling is required. Most strikingly, λ light chain–positive B cells were reduced sixfold in the B-KO mice, genetically placing Kidins220 in the PLCγ2 pathway. Thus, our data indicate that Kidins220 positively regulates pre-BCR and BCR functioning.
Collapse
Affiliation(s)
- Gina J Fiala
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Iga Janowska
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabiola Prutek
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Elias Hobeika
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Institute of Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Annyesha Satapathy
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Adrian Sprenger
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Plum
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Seidl
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Jörn Dengjel
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Michael Reth
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabrizia Cesca
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Tilman Brummer
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Susana Minguet
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang W A Schamel
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
46
|
Schmieg N, Thomas C, Yabe A, Lynch DS, Iglesias T, Chakravarty P, Schiavo G. Novel Kidins220/ARMS Splice Isoforms: Potential Specific Regulators of Neuronal and Cardiovascular Development. PLoS One 2015; 10:e0129944. [PMID: 26083449 PMCID: PMC4470590 DOI: 10.1371/journal.pone.0129944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/14/2015] [Indexed: 01/19/2023] Open
Abstract
Kidins220/ARMS is a transmembrane protein playing a crucial role in neuronal and cardiovascular development. Kidins220/ARMS is a downstream target of neurotrophin receptors and interacts with several signalling and trafficking factors. Through computational modelling, we found two potential sites for alternative splicing of Kidins220/ARMS. The first is located between exon 24 and exon 29, while the second site replaces exon 32 by a short alternative terminal exon 33. Here we describe the conserved occurrence of several Kidins220/ARMS splice isoforms at RNA and protein levels. Kidins220/ARMS splice isoforms display spatio-temporal regulation during development with distinct patterns in different neuronal populations. Neurotrophin receptor stimulation in cortical and hippocampal neurons and neuroendocrine cells induces specific Kidins220/ARMS splice isoforms and alters the appearance kinetics of the full-length transcript. Remarkably, alternative terminal exon splicing generates Kidins220/ARMS variants with distinct cellular localisation: Kidins220/ARMS containing exon 32 is targeted to the plasma membrane and neurite tips, whereas Kidins220/ARMS without exon 33 mainly clusters the full-length protein in a perinuclear intracellular compartment in PC12 cells and primary neurons, leading to a change in neurotrophin receptor expression. Overall, this study demonstrates the existence of novel Kidins220/ARMS splice isoforms with unique properties, revealing additional complexity in the functional regulation of neurotrophin receptors, and potentially other signalling pathways involved in neuronal and cardiovascular development.
Collapse
Affiliation(s)
- Nathalie Schmieg
- Molecular Neuropathobiology Laboratory, Sobell Department of Motor Neuroscience & Movement Disorders, UCL Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- The Francis Crick Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Claire Thomas
- The Francis Crick Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Arisa Yabe
- The Francis Crick Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - David S. Lynch
- Molecular Neuropathobiology Laboratory, Sobell Department of Motor Neuroscience & Movement Disorders, UCL Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- Leonard Wolfson Centre for Experimental Neurology, University College London, 8 Queen Anne Street, London W1G 9LD, United Kingdom
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid 28029, Spain
- CIBERNED (ISCIII), C/ Valderrebollo 5, Madrid 28031, Spain
| | - Probir Chakravarty
- The Francis Crick Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Giampietro Schiavo
- Molecular Neuropathobiology Laboratory, Sobell Department of Motor Neuroscience & Movement Disorders, UCL Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- * E-mail:
| |
Collapse
|
47
|
Cesca F, Satapathy A, Ferrea E, Nieus T, Benfenati F, Scholz-Starke J. Functional Interaction between the Scaffold Protein Kidins220/ARMS and Neuronal Voltage-Gated Na+ Channels. J Biol Chem 2015; 290:18045-18055. [PMID: 26037926 DOI: 10.1074/jbc.m115.654699] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Indexed: 12/19/2022] Open
Abstract
Kidins220 (kinase D-interacting substrate of 220 kDa)/ankyrin repeat-rich membrane spanning (ARMS) acts as a signaling platform at the plasma membrane and is implicated in a multitude of neuronal functions, including the control of neuronal activity. Here, we used the Kidins220(-/-) mouse model to study the effects of Kidins220 ablation on neuronal excitability. Multielectrode array recordings showed reduced evoked spiking activity in Kidins220(-/-) hippocampal networks, which was compatible with the increased excitability of GABAergic neurons determined by current-clamp recordings. Spike waveform analysis further indicated an increased sodium conductance in this neuronal subpopulation. Kidins220 association with brain voltage-gated sodium channels was shown by co-immunoprecipitation experiments and Na(+) current recordings in transfected HEK293 cells, which revealed dramatic alterations of kinetics and voltage dependence. Finally, an in silico interneuronal model incorporating the Kidins220-induced Na(+) current alterations reproduced the firing phenotype observed in Kidins220(-/-) neurons. These results identify Kidins220 as a novel modulator of Nav channel activity, broadening our understanding of the molecular mechanisms regulating network excitability.
Collapse
Affiliation(s)
- Fabrizia Cesca
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, 16163 Genova, Italy.
| | - Annyesha Satapathy
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Enrico Ferrea
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Sensorimotor Group, German Primate Center, 37077 Göttingen, Germany
| | - Thierry Nieus
- Neuro Technology Department, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Joachim Scholz-Starke
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Institute of Biophysics, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy.
| |
Collapse
|
48
|
Singh VB, Wooten AK, Jackson JW, Maggirwar SB, Kiebala M. Investigating the role of ankyrin-rich membrane spanning protein in human immunodeficiency virus type-1 Tat-induced microglia activation. J Neurovirol 2015; 21:186-98. [PMID: 25636783 DOI: 10.1007/s13365-015-0318-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/30/2014] [Accepted: 01/09/2015] [Indexed: 12/12/2022]
Abstract
Long-term persistence of human immunodeficiency virus type-1 (HIV) in the central nervous system (CNS) results in mild to severe neurocognitive impairment in a significant proportion of the HIV-infected population. These neurological deficits are known as HIV-associated neurocognitive disorders (HAND). Microglia are CNS-resident immune cells that are directly infected by HIV and consequently secrete proinflammatory molecules that contribute to HIV-induced neuroinflammation. Indeed, the number of activated macrophage and microglia in the brain is more highly correlated with cognitive impairment than the amount of neuronal apoptosis. Ankyrin-rich membrane spanning protein (ARMS/Kidins220) is a multidomain transmembrane protein that is involved with neurotrophin signaling in the CNS. We have previously established the role of ARMS in mediating neuronal survival via a neurotrophin-dependent mechanism. Recent reports also have suggested that ARMS is involved with cell signaling in multiple immune cell types. In this study, we aim to investigate the role of ARMS in HIV Tat-mediated microglial cell activation by employing in vitro methods. Following ARMS depletion by a lentivirus encoding ARMS-specific short hairpin RNA (shRNA), we observed a marked reduction in the HIV Tat-induced proinflammatory response, associated with loss of tumor necrosis factor alpha production and nuclear factor-kappa B (NF-κB) activation. Furthermore, co-immunoprecipitation studies suggested that ARMS physically interacts with inhibitory kappa B kinase subunits in order to facilitate NF-κB activation. Our results establish the role of ARMS in microglial activation by HIV Tat and warrant additional studies to better understand these molecular mechanisms, which may uncover novel therapeutic targets for the treatment of HAND.
Collapse
Affiliation(s)
- Vir B Singh
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 672, Rochester, NY, 14642, USA,
| | | | | | | | | |
Collapse
|
49
|
Avriyanti E, Atik N, Kunii M, Furumoto N, Iwano T, Yoshimura SI, Harada R, Harada A. Functional redundancy of protein kinase D1 and protein kinase D2 in neuronal polarity. Neurosci Res 2015; 95:12-20. [PMID: 25639845 DOI: 10.1016/j.neures.2015.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 11/25/2022]
Abstract
Mammalian protein kinase D (PKD) isoforms have been proposed to regulate diverse biological processes, including the establishment and maintenance of neuronal polarity. To investigate the function of PKD in neuronal polarization in vivo, we generated PKD knockout (KO) mice. Here, we show that the brain, particularly the hippocampus, of both PKD1 KO and PKD2 KO mice was similar to that of control animals. Neurite length in cultured PKD1 KO and PKD2 KO hippocampal neurons was similar to that of wild-type neurons. However, hippocampal neurons deficient in both PKD1 and PKD2 genes showed a reduction in axonal elongation and an increase in the percentage of neurons with multiple axons relative to control neurons. These results reveal that whereas PKD1 and PKD2 are essential for neuronal polarity, there exists a functional redundancy between the two proteins.
Collapse
Affiliation(s)
- Erda Avriyanti
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan
| | - Nur Atik
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Department of Anatomy and Cell Biology, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan
| | - Naomi Furumoto
- Laboratory for Molecular Traffic, Department of Cellular and Molecular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Tomohiko Iwano
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan
| | - Shin-Ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan
| | - Reiko Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Department of Judo Therapy, Takarazuka University of Medical and Health Care, Takarazuka, 666-0162, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Laboratory for Molecular Traffic, Department of Cellular and Molecular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| |
Collapse
|
50
|
Jung H, Shin JH, Park YS, Chang MS. Ankyrin repeat-rich membrane spanning (ARMS)/Kidins220 scaffold protein regulates neuroblastoma cell proliferation through p21. Mol Cells 2014; 37:881-7. [PMID: 25410904 PMCID: PMC4275705 DOI: 10.14348/molcells.2014.0182] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 12/13/2022] Open
Abstract
Cell proliferation is tightly controlled by the cell-cycle regulatory proteins, primarily by cyclins and cyclin-dependent kinases (CDKs) in the G1 phase. The ankyrin repeat-rich membrane spanning (ARMS) scaffold protein, also known as kinase D-interacting substrate of 220 kDa (Kidins 220), has been previously identified as a prominent downstream target of neurotrophin and ephrin receptors. Many studies have reported that ARMS/Kidins220 acts as a major signaling platform in organizing the signaling complex to regulate various cellular responses in the nervous and vascular systems. However, the role of ARMS/Kidins220 in cell proliferation and cell-cycle progression has never been investigated. Here we report that knockdown of ARMS/Kidins220 inhibits mouse neuroblastoma cell proliferation by inducing slowdown of cell cycle in the G1 phase. This effect is mediated by the upregulation of a CDK inhibitor p21, which causes the decrease in cyclin D1 and CDK4 protein levels and subsequent reduction of pRb hyperphosphorylation. Our results suggest a new role of ARMS/Kidins220 as a signaling platform to regulate tumor cell proliferation in response to the extracellular stimuli.
Collapse
Affiliation(s)
- Heekyung Jung
- Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749,
Korea
| | - Joo-Hyun Shin
- Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749,
Korea
| | - Young-Seok Park
- Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749,
Korea
| | - Mi-Sook Chang
- Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749,
Korea
| |
Collapse
|