1
|
Muir AM, Reich A, Zou F, Carere DA, Harasink SM, Tran L, McGivern B. A recurrent variant in PPP2R5C identified in individuals with macrocephaly, intellectual disability, and seizures. HGG ADVANCES 2025; 6:100394. [PMID: 39696819 PMCID: PMC11773225 DOI: 10.1016/j.xhgg.2024.100394] [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: 10/01/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024] Open
Abstract
PPP2R5C encodes a B-type regulatory subunit of protein phosphatase 2A (PP2A). This protein serine/threonine phosphatase is a component of multiple signaling pathways and is an established negative regulator of cell division, growth, and proliferation. De novo variants in other subunits of PP2A are associated with neurodevelopment disorders and intellectual disability (ID). We report two unrelated affected individuals with a recurrent variant in PPP2R5C (c.457G>A: p.(Glu153Lys)). Core features in affected individuals include macrocephaly, ID, hypotonia, and seizures. The Glu153 residue is part of a highly conserved acidic loop and directly interacts with the PP2A catalytic subunit. Our results support heterozygous PPP2R5C missense variants as a potential cause of macrocephaly and neurodevelopmental disorder.
Collapse
Affiliation(s)
| | - Adi Reich
- GeneDx, LLC, Gaithersburg, MD 20877, USA
| | | | | | - Sue Moyer Harasink
- Nemours Children's Health-Delaware, Division of Neurology, Wilmington, DE 19803, USA
| | - Lily Tran
- Nemours Children's Health-Delaware, Division of Neurology, Wilmington, DE 19803, USA
| | | |
Collapse
|
2
|
Peti W, Padi SKR, Page R. Combining cryo-electron microscopy (cryo-EM) with orthogonal solution state methods to define the molecular basis of the phosphoprotein phosphatase family regulation and substrate specificity. Curr Opin Struct Biol 2025; 91:102992. [PMID: 39951957 PMCID: PMC11885005 DOI: 10.1016/j.sbi.2025.102992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/17/2025]
Abstract
Protein phosphatases are dynamic enzymes that exhibit complex regulatory mechanisms, with disruptions in these regulatory processes associated with disease. It is now clear that many phosphatases assemble into large macromolecular complexes via the interaction of phosphatase-specific regulatory proteins and substrates containing short linear motifs (SLiMs) or short helical motifs (SHelMs). Here, we review how cryo-electron microscopy (cryo-EM) integrated with orthogonal methods to study dynamic protein-protein interactions (NMR spectroscopy, hydrogen-deuterium exchange mass spectrometry, among others) is leading to new discoveries about the mechanisms controlling phosphatase assembly, substrate recruitment and dephosphorylation and, in turn, are providing novel strategies for targeting phosphatase-related diseases. This review focuses on the recently determined structures and regulation of the phosphoprotein phosphatase (PPP) family of ser/thr phosphatases-PP1, PP2A, Calcineurin and PP5.
Collapse
Affiliation(s)
- Wolfgang Peti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, USA.
| | - Sathish K R Padi
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, USA
| | - Rebecca Page
- Department of Cell Biology, University of Connecticut Health Center, Farmington, USA.
| |
Collapse
|
3
|
Verbinnen I, Douzgou Houge S, Hsieh TC, Lesmann H, Kirchhoff A, Geneviève D, Brimble E, Lenaerts L, Haesen D, Levy RJ, Thevenon J, Faivre L, Marco E, Chong JX, Bamshad M, Patterson K, Mirzaa GM, Foss K, Dobyns W, White SM, Pais L, O'Heir E, Itzikowitz R, Donald KA, Van der Merwe C, Mussa A, Cervini R, Giorgio E, Roscioli T, Dias KR, Evans CA, Brown NJ, Ruiz A, Trujillo Quintero JP, Rabin R, Pappas J, Yuan H, Lachlan K, Thomas S, Devlin A, Wright M, Martin R, Karwowska J, Posmyk R, Chatron N, Stark Z, Heath O, Delatycki M, Buchert R, Korenke GC, Ramsey K, Narayanan V, Grange DK, Weisenberg JL, Haack TB, Karch S, Kipkemoi P, Mangi M, Bindels de Heus KGCB, de Wit MCY, Barakat TS, Lim D, Van Winckel G, Spillmann RC, Shashi V, Jacob M, Stehr AM, Krawitz P, Douzgos Houge G, Janssens V. Pathogenic de novo variants in PPP2R5C cause a neurodevelopmental disorder within the Houge-Janssens syndrome spectrum. Am J Hum Genet 2025; 112:554-571. [PMID: 39978342 PMCID: PMC11947181 DOI: 10.1016/j.ajhg.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Pathogenic variants resulting in protein phosphatase 2A (PP2A) dysfunction result in mild to severe neurodevelopmental delay. PP2A is a trimer of a catalytic (C) subunit, scaffolding (A) subunit, and substrate binding/regulatory (B) subunit, encoded by 19 different genes. De novo missense variants in PPP2R5D (B56δ) or PPP2R1A (Aα) and de novo missense and loss-of-function variants in PPP2CA (Cα) lead to syndromes with overlapping phenotypic features, known as Houge-Janssens syndrome (HJS) types 1, 2, and 3, respectively. Here, we describe an additional condition in the HJS spectrum in 26 individuals with variants in PPP2R5C, encoding the regulatory B56γ subunit. Most changes were de novo and of the missense type. The clinical features were well within the HJS spectrum with strongest resemblance to HJS type 1, caused by B56δ variants. Common features were neurodevelopmental delay and hypotonia, with a high risk of epilepsy, behavioral problems, and mildly dysmorphic facial features. Head circumferences were above average or macrocephalic. The degree of intellectual disability was, on average, milder than in other HJS types. All variants affected either substrate binding (2/19), C-subunit binding (2/19), or both (15/19). Five variants were recurrent. Catalytic activity of the phosphatase was variably affected by the variants. Of note, PPP2R5C total loss-of-function variants could be inherited from a non-symptomatic parent. This implies that a dominant-negative mechanism on substrate dephosphorylation or general PP2A function is the most likely pathogenic mechanism.
Collapse
Affiliation(s)
- Iris Verbinnen
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium; KU Leuven Institute for Rare Diseases (Leuven.IRD), Leuven, Belgium
| | - Sofia Douzgou Houge
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Hellen Lesmann
- Institute of Human Genetics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Aron Kirchhoff
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - David Geneviève
- Montpellier University, INSERM U1183, Centre de Référence Anomalies du développement et syndromes malformatifs, ERN ITHACA, Génétique clinique, CHU Montpellier, Montpellier, France
| | | | - Lisa Lenaerts
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Dorien Haesen
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Rebecca J Levy
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, CA, USA
| | - Julien Thevenon
- CNRS UMR 5309, INSERM U1209, Institute of Advanced Biosciences, Université Grenoble-Alpes, Service Génomique et Procréation, Centre Hospitalo-Universitaire Grenoble Alpes, Cedex Grenoble, France
| | - Laurence Faivre
- Centre de génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'enfants, CHU Dijon Bourgogne, Dijon, France; UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | | | - Jessica X Chong
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Mike Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Karynne Patterson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Ghayda M Mirzaa
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kimberly Foss
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William Dobyns
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Susan M White
- Victorian Clinical Genetics Services (VCGS), Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Lynn Pais
- Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emily O'Heir
- Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Raphaela Itzikowitz
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Celia Van der Merwe
- Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Alessandro Mussa
- Department of Public Health and Pediatric Sciences, University of Torino, Regina Margherita Children's Hospital, Torino, Italy
| | - Raffaela Cervini
- Child Neuropsychiatry Department, Maria Vittoria Hospital, Torino, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS Mondino Foundation, Neurogenetics Research Centre, Pavia, Italy
| | - Tony Roscioli
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; Centre for Clinical Genetics, Sydney Children's Hospital, Sydney, NSW, Australia; New South Wales Health Pathology Randwick Genomics, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Kerith-Rae Dias
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2031, Australia
| | - Carey-Anne Evans
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; New South Wales Health Pathology Randwick Genomics, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Natasha J Brown
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Anna Ruiz
- Genetics Laboratory, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, 08208 Sabadell, Spain
| | - Juan Pablo Trujillo Quintero
- Unitat de Genètica Clínica, Servei de Medicina Pediàtrica, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, 08208 Sabadell, Spain
| | - Rachel Rabin
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - John Pappas
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Hai Yuan
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Katherine Lachlan
- Wessex Clinical Genetics Service, University Hospital Southampton, Princess Anne Hospital, Southampton SO16 5YA, UK
| | - Simon Thomas
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; Wessex Regional Genetics Laboratory, Salisbury NSF Foundation Trust, Salisbury District Hospital, Salisbury, UK
| | - Anita Devlin
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Richard Martin
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Genetic Medicine, Newcastle upon Tyne, UK
| | - Joanna Karwowska
- Department of Clinical Genetics, Medical University in Bialystok, Bialystok, Poland
| | - Renata Posmyk
- Department of Clinical Genetics, Medical University in Bialystok, Bialystok, Poland
| | - Nicolas Chatron
- Hospices Civils de Lyon, Groupe Hospitalier Est, Service de génétique, Bron, France; Université de Lyon, University Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, Lyon, France
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Australian Genomics Health Alliance, Melbourne, VIC, Australia; Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Oliver Heath
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia
| | - Martin Delatycki
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia; Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Rebecca Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Georg-Christoph Korenke
- Klinik für Neuropädiatrie und angeborene Stoffwechselerkrankungen, Klinikum Oldenburg, Oldenburg, Germany
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Dorothy K Grange
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, One Children's Place, St. Louis, MO, USA
| | - Judith L Weisenberg
- Department of Pediatric Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Stephanie Karch
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Patricia Kipkemoi
- Neuroscience Unit, KEMRI-Wellcome Trust, Center for Geographic Medicine Research Coast, Kilifi, Kenya
| | - Moses Mangi
- Neuroscience Unit, KEMRI-Wellcome Trust, Center for Geographic Medicine Research Coast, Kilifi, Kenya
| | - Karen G C B Bindels de Heus
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Marie-Claire Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Neurology and Pediatric Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Tahsin Stefan Barakat
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Derek Lim
- Department of Clinical Genetics, Lavender House, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | | | - Rebecca C Spillmann
- Department of Pediatrics-Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Vandana Shashi
- Department of Pediatrics-Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Maureen Jacob
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Antonia M Stehr
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium; KU Leuven Institute for Rare Diseases (Leuven.IRD), Leuven, Belgium.
| |
Collapse
|
4
|
Khan AA, Dewald HD. Nitric oxide and peroxynitrite as new biomarkers for early diagnosis of autism. Brain Res 2025; 1850:149438. [PMID: 39793916 DOI: 10.1016/j.brainres.2024.149438] [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: 07/15/2024] [Revised: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Autism spectrum disorder, or autism, is a neurodevelopmental disorder of the developing child's brain with a genetic causality. It can be diagnosed at about three years after birth when it begins to present itself via a range of neuropsychiatric symptoms. Nitric oxide is a crucial small molecule of life synthesized within cells of our body systems, including cells of our brain. Peroxynitrite is the product of reaction between superoxide anion and nitric oxide. It normally isomerizes into harmless nitrates or nitrites. However, when excessive superoxide anion is present, the cellular concentration of peroxynitrite can increase to a toxic level. Autism has been suggested to cause oxidative damage to brain cells. Until now, it is impossible to sample tissue from a live brain. Instead, stem cells can be derived (from an autism patient's somatic cells) which can then be differentiated and chemically directed to grow into miniature 3-dimensional tissue masses resembling specific brain regions (e.g., the cortex) called brain organoids. This review discusses utilizing nitric oxide and peroxynitrite as biomarkers and comparing their relative concentrations in stem cells and stem cell derived brain organoids of healthy and autistic individuals to develop a bioanalytical process for early diagnosis of autism.
Collapse
Affiliation(s)
- Abdullah Asif Khan
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States.
| |
Collapse
|
5
|
Carter JL, Halmai JANM, Waldo JJ, Vij PA, Anguiano M, Villegas IJ, Du YX, Nolta J, Fink KD. A de novo missense mutation in PPP2R5D alters dopamine pathways and morphology of iPSC-derived midbrain neurons. Stem Cells 2025; 43:sxae068. [PMID: 39460716 PMCID: PMC11811633 DOI: 10.1093/stmcls/sxae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 08/27/2024] [Indexed: 10/28/2024]
Abstract
Induced pluripotent stem cell (iPSC) models of neurodevelopmental disorders (NDDs) have promoted an understanding of commonalities and differences within or across patient populations by revealing the underlying molecular and cellular mechanisms contributing to disease pathology. Here, we focus on developing a human model for PPP2R5D-related NDD, called Jordan syndrome, which has been linked to Early-Onset Parkinson's Disease (EOPD). Here we sought to understand the underlying molecular and cellular phenotypes across multiple cell states and neuronal subtypes in order to gain insight into Jordan syndrome pathology. Our work revealed that iPSC-derived midbrain neurons from Jordan syndrome patients display significant differences in dopamine-associated pathways and neuronal architecture. We then evaluated a CRISPR-based approach for editing heterozygous dominant G-to-A mutations at the transcript level in patient-derived neural stem cells. Our findings show that site-directed RNA editing is influenced by sgRNA length and cell type. These studies support the potential for a CRISPR RNA editor system to selectively edit mutant transcripts harboring G-to-A mutations in neural stem cells while providing an alternative editing technology for those suffering from NDDs.
Collapse
Affiliation(s)
- Jasmine L Carter
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Julian A N M Halmai
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Jennifer J Waldo
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Paula A Vij
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Maribel Anguiano
- Center for Neuroscience, University of California Davis, Sacramento, CA 95817, United States
| | - Isaac J Villegas
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Yu Xin Du
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| | - Jan Nolta
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
| | - Kyle D Fink
- Center for Interventional Genetics, University of California, Davis, Sacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA 95817, United States
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, United States
- Department of Neurology, University of California Davis Health Systems, Sacramento, CA, United States
| |
Collapse
|
6
|
Musumeci A, Vinci M, Verbinnen I, Treccarichi S, Nigliato E, Chiavetta V, Greco D, Vitello GA, Federico C, Janssens V, Saccone S, Calì F. PPP2R5E: New gene potentially involved in specific learning disorders and myopathy. Gene 2025; 933:148945. [PMID: 39284558 DOI: 10.1016/j.gene.2024.148945] [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: 05/14/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/21/2024]
Abstract
Protein phosphatase 2A (PP2A) is a family of multifunctional enzymatic complexes crucial for cellular signalling, playing a pivotal role in brain function and development. Mutations in specific genes encoding PP2A complexes have been associated with neurodevelopmental disorders with hypotonia and high risk of seizures. In the current work, we present an individual with specific learning problems, motor coordination disorders, hypotonia and behavioural issues. Although whole exome sequencing (WES) did not unveil pathogenic variants in known genes related to these symptoms, a de novo heterozygous variant Glu191Lys was identified within PPP2R5E, encoding the PP2A regulatory subunit B56ε. The novel variant was not observed in the four healthy brothers and was not detected as parental somatic mosaicism. The mutation predicted a change of charge of the mutated amino acid within a conserved LFDSEDPRER motif common to all PPP2R5 B-subunits. Biochemical assays demonstrated a decreased interaction with the PP2A A and C subunits, leading to disturbances in holoenzyme formation, and thus likely, function. For the first time, we report a potential causal link between the observed variant within the PPP2R5E gene and the symptoms manifested in the subject, spanning specific learning problems and motor coordination disorders potentially associated with myopathy.
Collapse
Affiliation(s)
- Antonino Musumeci
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| | - Mirella Vinci
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Herestraat 49, PO-box 901, B-3000 Leuven, Belgium
| | - Simone Treccarichi
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| | - Eleonora Nigliato
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Herestraat 49, PO-box 901, B-3000 Leuven, Belgium
| | - Valeria Chiavetta
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| | - Donatella Greco
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| | | | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Herestraat 49, PO-box 901, B-3000 Leuven, Belgium
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy.
| | - Francesco Calì
- Oasi Research Institute-IRCCS, via Conte Ruggero 73, 94018, Troina, EN, Italy
| |
Collapse
|
7
|
Comisi F, Soddu C, Lai F, Marica M, Lorrai M, Mancuso G, Giglio S, Savasta S. PPP2R5D-Related Neurodevelopmental Disorder and Multiple Haemangiomas: A Novel Phenotypic Trait? Pediatr Rep 2024; 16:1200-1206. [PMID: 39728742 DOI: 10.3390/pediatric16040101] [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: 10/16/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Houge-Janssens syndrome 1 is a condition with onset in early childhood caused by heterozygous pathogenic variants in the PPP2R5D gene, which encodes a B56 regulatory subunit of the serine/threonine protein phosphatase 2A (PP2A). There is evidence that the PP2A-PPP2R5D complex is involved in regulating the phosphatidylinositol 3-kinase (PI3K)/AKT signalling pathway, which is crucial for several cellular processes, including the pathogenesis and progression of haemangiomas. CASE PRESENTATION We report the first PPP2R5D-related neurodevelopmental disorder case from Sardinia, a child with transient hypoglycaemia, facial dysmorphisms, and multiple haemangiomas. Whole Exome Sequencing analysis confirmed the clinical suspicion, detecting the presence of the de novo missense variant c.592G>A in the PPP2R5D gene. CONCLUSIONS Haemangiomas have never been linked to the syndromic phenotype of the PPP2R5D-associated disorder. The close correlation between the PP2A enzyme and the PI3K/AKT signalling pathway suggests the possible correlation between its dysfunction and activation of haemangiogenesis. Our report highlights a possible link between the PPP2R5D-related disorder and altered angiogenesis, characterizing diffuse haemangiomas as a possible novel phenotypic trait of this condition.
Collapse
Affiliation(s)
- Francesco Comisi
- Pediatrics Department, Microcitemico Hospital "A. Cao", University of Cagliari, 09124 Cagliari, Italy
| | - Consolata Soddu
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital "A. Cao", ASL 8 Cagliari, 09121 Cagliari, Italy
| | - Francesco Lai
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital "A. Cao", ASL 8 Cagliari, 09121 Cagliari, Italy
- Unit of Oncology and Molecular Pathology, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Monica Marica
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital "A. Cao", ASL 8 Cagliari, 09121 Cagliari, Italy
| | - Michela Lorrai
- Medical Genetics Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Giancarlo Mancuso
- Medical Genetics Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Sabrina Giglio
- Medical Genetics Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
- Centre for Research University Services, University of Cagliari, 09124 Cagliari, Italy
- Medical Genetics, "R. Binaghi" Hospital, ASL 8 Cagliari, 09126 Cagliari, Italy
| | - Salvatore Savasta
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital "A. Cao", Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
8
|
Lin F, Liang X, Meng Y, Zhu Y, Li C, Zhou X, Hu S, Yi N, Lin Q, He S, Sun Y, Sheng J, Fan H, Li L, Peng L. Unmasking Protein Phosphatase 2A Regulatory Subunit B as a Crucial Factor in the Progression of Dilated Cardiomyopathy. Biomedicines 2024; 12:1887. [PMID: 39200351 PMCID: PMC11352103 DOI: 10.3390/biomedicines12081887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure. Although significant progress has been made in elucidating the underlying mechanisms, further investigation is required for clarifying molecular diagnostic and therapeutic targets. In this study, we found that the mRNA level of protein phosphatase 2 regulatory subunit B' delta (Ppp2r5d) was altered in the peripheral blood plasma of DCM patients. Knockdown of Ppp2r5d in murine cardiomyocytes increased the intracellular levels of reactive oxygen species (ROS) and inhibited adenosine triphosphate (ATP) synthesis. In vivo knockdown of Ppp2r5d in an isoproterenol (ISO)-induced DCM mouse model aggravated the pathogenesis and ultimately led to heart failure. Mechanistically, Ppp2r5d-deficient cardiomyocytes showed an increase in phosphorylation of STAT3 at Y705 and a decrease in phosphorylation of STAT3 at S727. The elevated levels of phosphorylation at Y705 in STAT3 triggered the upregulation of interleukin 6 (IL6) expression. Moreover, the decreased phosphorylation at S727 in STAT3 disrupted mitochondrial electron transport chain function and dysregulated ATP synthesis and ROS levels. These results hereby reveal a novel role for Ppp2r5d in modulating STAT3 pathway in DCM, suggesting it as a potential target for the therapy of the disease.
Collapse
Affiliation(s)
- Fang Lin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yilei Meng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Yuping Zhu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Chenyu Li
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Sangyu Hu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Na Yi
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Qin Lin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Siyu He
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Yizhuo Sun
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Jie Sheng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Huimin Fan
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Li Li
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Luying Peng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| |
Collapse
|
9
|
Srivastava S, Koh HY, Smith L, Poduri A. Cerebral Palsy Phenotypes in Genetic Epilepsies. Pediatr Neurol 2024; 157:79-86. [PMID: 38901369 PMCID: PMC11418751 DOI: 10.1016/j.pediatrneurol.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/03/2024] [Accepted: 05/26/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Although there are established connections between genetic epilepsies and neurodevelopmental disorders like intellectual disability, the presence of cerebral palsy (CP) in genetic epilepsies is undercharacterized. We performed a retrospective chart review evaluating the motor phenotype of patients with genetic epilepsies. METHODS Patients were ascertained through a research exome sequencing study to identify genetic causes of epilepsy. We analyzed data from the first 100 individuals with molecular diagnoses. We determined motor phenotype by reviewing medical records for muscle tone and motor function data. We characterized patients according to CP subtypes: spastic diplegic, spastic quadriplegic, spastic hemiplegic, dyskinetic, hypotonic-ataxic. RESULTS Of 100 individuals with genetic epilepsies, 14% had evidence of possible CP, including 5% characterized as hypotonic-ataxic CP, 5% spastic quadriplegic CP, 3% spastic diplegic CP, and 1% hemiplegic CP. Presence of CP did not correlate with seizure onset age (P = 0.63) or seizure control (P = 0.07). CP occurred in 11% (n = 3 of 27) with focal epilepsy, 9% (n = 5 of 54) with generalized epilepsy, and 32% (n = 6 of 19) with combined focal/generalized epilepsy (P = 0.06). CONCLUSIONS In this retrospective analysis of patients with genetic epilepsies, we identified a substantial portion with CP phenotypes, representing an under-recognized comorbidity. These findings underscore the many neurodevelopmental features associated with neurogenetic conditions, regardless of the feature for which they were ascertained for sequencing. Detailed motor phenotyping is needed to determine the prevalence of CP and its subtypes among genetic epilepsies. These motor phenotypes require clinical management and represent important targeted outcomes in trials for patients with genetic epilepsies.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts; Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts; Cerebral Palsy and Spasticity Center, Boston Children's Hospital, Boston, Massachusetts
| | - Hyun Yong Koh
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Neurogenetics Program and Epilepsy Genetics Program, Boston Children's Hospital, Boston, Massachusetts
| | - Lacey Smith
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Neurogenetics Program and Epilepsy Genetics Program, Boston Children's Hospital, Boston, Massachusetts
| | - Annapurna Poduri
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts; Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts; Neurogenetics Program and Epilepsy Genetics Program, Boston Children's Hospital, Boston, Massachusetts.
| |
Collapse
|
10
|
Jiang Y, Wu B, Zhang X, Yang L, Wang S, Li H, Zhou S, Qian Y, Wang H. Thirteen New Patients of PPP2R5D Gene Mutation and the Fine Profile of Genotype-Phenotype Correlation Unraveling the Pathogenic Mechanism Underlying Macrocephaly Phenotype. CHILDREN (BASEL, SWITZERLAND) 2024; 11:897. [PMID: 39201832 PMCID: PMC11352527 DOI: 10.3390/children11080897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024]
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) are a group of diseases that severely affect the physical and mental health of children. The PPP2R5D gene encodes B56δ, the regulatory subunit of protein phosphatase 2A (PP2A). NDDs related to the PPP2R5D gene have recently been defined as Houge-Janssens syndrome 1. METHODS Clinical/whole exome sequencing was performed on approximately 3000 patients with NDDs from 2017 to 2023. In vitro experiments were performed to assess the impairment of variants to protein expression and the assembly of PP2A holoenzyme. The genetic information and phenotypes of the reported patients, as well as patients in this study, were summarized, and the genotype-phenotype relationship was analyzed. The probability of pathogenic missense variants in PPP2R5D was predicted using AlphaMissense (AM), and the relationship between certain phenotype and 3D protein structural features were analyzed. RESULTS Thirteen new patients carrying twelve PPP2R5D gene variants were detected, including five novel missense variants and one novel frameshift variant. In vitro experiments revealed that the frameshift variant p.H463Mfs*3 resulted in a ~50 kDa truncated protein with lower expression level. Except for E420K and T536R, other missense variants impaired holoenzyme assembly. Furthermore, we found that pathogenic/likely pathogenic (P/LP) variants that have been reported so far were all missense variants and clustered in three conserved regions, and the likelihood of P/LP mutations located in these conserved regions was extremely high. In addition, the macrocephaly phenotype was related to negatively charged residues involved in substrate recruitment. CONCLUSIONS We reported thirteen new patients with PPP2R5D gene variants and expanded the PPP2R5D variant spectrum. We confirmed the pathogenicity of novel variants through in vitro experiments. Our findings in genotype-phenotype relationship provide inspiration for genetic counseling and interpretation of variants. We also provide directions for further research on the mechanism of macrocephaly phenotype.
Collapse
Affiliation(s)
- Yinmo Jiang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| | - Bingbing Wu
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| | - Xi Zhang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| | - Lin Yang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| | - Sujuan Wang
- Department of Rehabilitation, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Huiping Li
- Department of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Shuizhen Zhou
- Neurology Department, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Yanyan Qian
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| | - Huijun Wang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.J.); (X.Z.)
| |
Collapse
|
11
|
Ayoubi R, Fotouhi M, Alende C, Southern K, Laflamme C, NeuroSGC/YCharOS/EDDU collaborative group, ABIF consortium. A guide to selecting high-performing antibodies for Serine/threonine-protein phosphatase 2A 56 kDa regulatory subunit delta isoform (PPP2R5D) for use in Western Blot, immunoprecipitation and immunofluorescence. F1000Res 2024; 13:1. [PMID: 39935523 PMCID: PMC11811605 DOI: 10.12688/f1000research.145146.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 02/13/2025] Open
Abstract
Protein phosphatase 2A is a serine/threonine phosphatase with activity dependent on an associated regulatory subunit, serine/threonine-protein phosphatase 2A 56 kDa regulatory subunit delta (δ) isoform (PPP2R5D). PPP2R5D is the δ isoform in the B56 family of regulatory subunits. Abundantly expressed in the brain and involved in a broad range of cellular processes, PPP2R5D plays an essential role in modulating key neuronal pathways and signalling. Pathogenic mutations in the PPP2R5D gene are linked to clinical symptoms characterized by neurodevelopmental delay, intellectual disability, and autism spectrum disorders. The etiology of these genetic disorders remains unknown, which can partly be due to the lack of independently characterized antibodies. Here we have characterized six PPP2R5D commercial antibodies for Western Blot, immunoprecipitation, and immunofluorescence using a standardized experimental protocol based on comparing read-outs in knockout cell lines and isogenic parental controls. These studies are part of a larger, collaborative initiative seeking to address antibody reproducibility by characterizing commercially available antibodies for human proteins and publishing the results openly as a resource for the scientific community. While use of antibodies and protocols vary between laboratories, we encourage readers to use this report as a guide to select the most appropriate antibodies for their specific needs.
Collapse
Affiliation(s)
- Riham Ayoubi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Maryam Fotouhi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Charles Alende
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Kathleen Southern
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Carl Laflamme
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | | | - ABIF consortium
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| |
Collapse
|
12
|
Mayer A, Derua R, Spahn E, Verbinnen I, Zhang Y, Wadzinski B, Swingle MR, Honkanen R, Janssens V, Xia H. The role of liprin-α1 phosphorylation in its liquid-liquid phase separation: regulation by PPP2R5D/PP2A holoenzyme. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599485. [PMID: 38948786 PMCID: PMC11213027 DOI: 10.1101/2024.06.18.599485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Liprin-α1 is a widely expressed scaffolding protein responsible for regulating cellular processes such as focal adhesion, cell motility, and synaptic transmission. Liprin-α1 interacts with many proteins including ELKS, GIT1, liprin-β, and LAR-family receptor tyrosine protein phosphatase. Through these protein-protein interactions, liprin-α1 assembles large higher-order molecular complexes; however, the regulation of this complex assembly/disassembly is unknown. Liquid-liquid phase separation (LLPS) is a process that concentrates proteins within cellular nano-domains to facilitate efficient spatiotemporal signaling in response to signaling cascades. While there is no report that liprin-α1 spontaneously undergoes LLPS, we found that GFP-liprin-α1 expressed in HEK293 cells occasionally forms droplet-like condensates. MS-based interactomics identified Protein Phosphatase 2A (PP2A)/B56δ (PPP2R5D) trimers as specific interaction partners of liprin-α1 through a canonical Short Linear Interaction Motif (SLiM) in its N-terminal dimerization domain. Mutation of this SLiM nearly abolished PP2A interaction, and resulted in significantly increased LLPS. GFP-liprin-α1 showed significantly increased droplet formation in HEK293 cells devoid of B56δ (PPP2R5D knockout), suggesting that PPP2R5D/PP2A holoenzyme inhibits liprin-α1 LLPS. Guided by reported liprin-α1 Ser/Thr phosphorylation sites, we found liprin-α1 phospho-mimetic mutant at serine 763 (S763E) is sufficient to drive its LLPS. Domain mapping studies of liprin-α1 indicated that the intrinsically disordered region, the N-terminal dimerization domain, and the SAM domains are all necessary for liprin-α1 LLPS. Finally, expression of p.E420K, a human PPP2R5D variant causing Houge-Janssens Syndrome type 1 (also known as Jordan's Syndrome), significantly compromised suppression of liprin-α1 LLPS. Our work identified B56δ-PP2A holoenzyme as an inhibitor of liprin-α1 LLPS via regulation at multiple phosphorylation sites.
Collapse
|
13
|
Mahale R, Arunachal G, Chadha D, Padmanabha H, M P, Pavagada M. Early-onset levodopa responsive parkinsonism in PPP2R5D mutation. Parkinsonism Relat Disord 2024; 123:106952. [PMID: 38582018 DOI: 10.1016/j.parkreldis.2024.106952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Affiliation(s)
- Rohan Mahale
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India.
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Deepak Chadha
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Hansashree Padmanabha
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Pooja M
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Mathuranath Pavagada
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| |
Collapse
|
14
|
Zreibe K, Kanner CH, Uher D, Beard G, Patterson M, Harris M, Doerger J, Calamia S, Chung WK, Montes J. Characterizing ambulatory function in children with PPP2R5D-related neurodevelopmental disorder. Gait Posture 2024; 110:77-83. [PMID: 38547676 PMCID: PMC11056288 DOI: 10.1016/j.gaitpost.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/29/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Individuals with PPP2R5D-related neurodevelopmental disorder have an atypical gait pattern characterized by ataxia and incoordination. Structured, quantitative assessments are needed to further understand the impact of these impairments on function. RESEARCH QUESTION How do gait parameters and ambulatory function of individuals with PPP2R5D-related neurodevelopmental disorder compare to age and sex matched healthy norms? METHODS Twenty-six individuals with PPP2R5D pathogenic genetic variants participated in this observational, single visit study. Participants completed at least one of the following gait assessments: quantitative gait analysis at three different speeds (preferred pace walking (PPW), fast paced walking (FPW) and running, six-minute walk test (6MWT), 10-meter walk run (10MWR), and timed up and go (TUG). Descriptive statistics were used to summarize gait variables. Percent of predicted values were calculated using published norms. Paired t-tests and regression analyses were used to compare gait variables. RESULTS The median age of the participants was 8 years (range 4-27) and eighteen (69.2 %) were female. Individuals with PPP2R5D-related neurodevelopmental disorder walked slower and with a wider base of support than predicted for their age and sex. Stride velocity ranged from 48.9 % to 70.1 % and stride distance from 58.5 % to 81.9 % of predicted during PPW. Percent of predicted distance walked on the 6MWT ranged from 30.6 % to 71.1 % representing varied walking impairment. Increases in stride distance, not cadence, were associated with changes in stride velocity in FPW (R2 = 0.675, p =< 0.001) and running conditions (R2 = 0.918, p =< 0.001). SIGNIFICANCE We quantitatively assessed the abnormal gait in individuals with PPP2R5D-related neurodevelopmental disorder. These impairments may affect ability to adapt to environmental changes and participation in daily life. Rehabilitative interventions targeting gait speed and balance may improve function and safety for individuals with PPP2R5D-related neurodevelopmental disorder.
Collapse
Affiliation(s)
- Kyle Zreibe
- Department of Rehabilitation, UHealth-Jackson Holtz Children's Hospital, Miami, FL, USA; Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Cara H Kanner
- Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David Uher
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY, USA
| | - Gabriella Beard
- Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Madison Patterson
- Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Matthew Harris
- Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jerome Doerger
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sean Calamia
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA USA
| | - Jacqueline Montes
- Department of Rehabilitation & Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
15
|
Sudnawa KK, Chung WK. SPARKing New Insight Into Autism Across the Lifespan. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2024; 129:91-95. [PMID: 38411241 DOI: 10.1352/1944-7558-129.2.91] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Autism is heterogeneous at many levels, including clinical symptoms and etiology. A key strategy in studying heterogeneous conditions is having large enough sample sizes to stratify into smaller groups that are more homogeneous. SPARK and Simons Searchlight are large and growing research cohorts of individuals with autism in the United States and individuals with genetically defined neurodevelopmental conditions around the world, respectively. They both provide freely available phenotypic and genotypic data with the ability to re-contact participants through the research match program. Deep dives into each gene in Searchlight provide comprehensive natural history data to understand the differing clinical courses to inform proper clinical care, and work toward treatment for each condition. Moreover, pilots of genetically based newborn screening programs for neurogenetic disorders can provide opportunities for equitable and early diagnosis to try to improve outcomes with earlier interventions.
Collapse
Affiliation(s)
- Khemika K Sudnawa
- Khemika K. Sudnawa, Boston Children's Hospital, Harvard Medical School and Department of Pediatrics and Pramongkutklao Hospital and Pramongkutklao College of Medicine, Bangkok, Thailand
| | - Wendy K Chung
- Wendy K. Chung, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School
| |
Collapse
|
16
|
Kanner CH, Uher D, Zreibe K, Beard G, Patterson M, Harris M, Doerger J, Calamia S, Chung WK, Montes J. Validation of a modified version of the gross motor function measure in PPPR5D related neurodevelopmental disorder. Orphanet J Rare Dis 2024; 19:45. [PMID: 38326877 PMCID: PMC10848481 DOI: 10.1186/s13023-024-03067-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Protein phosphatase 2 regulatory subunit B' Delta (PPP2R5D)-related neurodevelopmental disorder is a rare genetic condition caused by pathogenic variants in the PPP2R5D gene. Clinical signs include hypotonia, gross motor delay, intellectual disability (ID), epilepsy, speech delays, and abnormal gait among other impairments. As this disorder was recognized within the last decade, there are only 103 people published diagnoses to date. A thorough understanding of the motor manifestations of this disorder has not yet been established. Knowledge of the natural history of PPP2R5D related neurodevelopmental disorder will lead to improved standard of care treatments as well as serve as a baseline foundation for future clinical trials. Appropriate outcome measures are necessary for use in clinical trials to uniformly measure function and monitor potential for change. The aim of this study was to validate the gross motor function measure (GMFM) in children and adults with PPP2R5D-related neurodevelopmental disorder in order to better characterize the disorder. RESULTS Thirty-eight individuals with PPP2R5D pathogenic variants, median age 8.0 years (range 1-27) were evaluated. Gross motor, upper limb and ambulatory function were assessed using the GMFM-66, six-minute walk test (6MWT), 10-meter walk run (10MWR), timed up and go (TUG), and revised upper limb module (RULM). The pediatric disability inventory computer adapted test (PEDI-CAT) captured caregiver reported assessment. Median GMFM-66 score was 60.6 (SD = 17.3, range 21.1-96.0). There were strong associations between the GMFM-66 and related mobility measures, 10MWR (rs = -0.733; p < 0.001), TUG (rs= -0.747; p = 0.003), 6MWT (r = 0.633; p = 0.006), RULM (r = 0.763; p < 0.001), PEDICAT-mobility (r = 0.855; p < 0.001), and daily activities (r = 0.822; p < 0.001) domains. CONCLUSIONS The GMFM is a valid measure for characterizing motor function in individuals with PPP2R5D related neurodevelopmental disorder. The GMFM-66 had strong associations with the RULM and timed function tests which characterized gross motor, upper limb and ambulatory function demonstrating concurrent validity. The GMFM-66 was also able to differentiate between functional levels in PPP2R5D related neurodevelopmental disorder demonstrating discriminant validity. Future studies should examine its sensitivity to change over time, ability to identify sub-phenotypes, and suitability as an outcome measure in future clinical trials in individuals with PPP2R5D variants.
Collapse
Affiliation(s)
- Cara H Kanner
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA.
| | - David Uher
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY, USA
| | - Kyle Zreibe
- Department of Rehabilitation, UHealth-Jackson Holtz Children's Hospital, Miami, FL, USA
| | - Gabriella Beard
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA
| | - Madison Patterson
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA
| | - Matthew Harris
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA
| | - Jerome Doerger
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sean Calamia
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jacqueline Montes
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, 617 West 168th Street, New York, NY, 10032, USA
| |
Collapse
|
17
|
Wu CG, Balakrishnan VK, Merrill RA, Parihar PS, Konovolov K, Chen YC, Xu Z, Wei H, Sundaresan R, Cui Q, Wadzinski BE, Swingle MR, Musiyenko A, Chung WK, Honkanen RE, Suzuki A, Huang X, Strack S, Xing Y. B56δ long-disordered arms form a dynamic PP2A regulation interface coupled with global allostery and Jordan's syndrome mutations. Proc Natl Acad Sci U S A 2024; 121:e2310727120. [PMID: 38150499 PMCID: PMC10769853 DOI: 10.1073/pnas.2310727120] [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: 07/01/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Intrinsically disordered regions (IDR) and short linear motifs (SLiMs) play pivotal roles in the intricate signaling networks governed by phosphatases and kinases. B56δ (encoded by PPP2R5D) is a regulatory subunit of protein phosphatase 2A (PP2A) with long IDRs that harbor a substrate-mimicking SLiM and multiple phosphorylation sites. De novo missense mutations in PPP2R5D cause intellectual disabilities (ID), macrocephaly, Parkinsonism, and a broad range of neurological symptoms. Our single-particle cryo-EM structures of the PP2A-B56δ holoenzyme reveal that the long, disordered arms at the B56δ termini fold against each other and the holoenzyme core. This architecture suppresses both the phosphatase active site and the substrate-binding protein groove, thereby stabilizing the enzyme in a closed latent form with dual autoinhibition. The resulting interface spans over 190 Å and harbors unfavorable contacts, activation phosphorylation sites, and nearly all residues with ID-associated mutations. Our studies suggest that this dynamic interface is coupled to an allosteric network responsive to phosphorylation and altered globally by mutations. Furthermore, we found that ID mutations increase the holoenzyme activity and perturb the phosphorylation rates, and the severe variants significantly increase the mitotic duration and error rates compared to the normal variant.
Collapse
Affiliation(s)
- Cheng-Guo Wu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
- Biophysics Program, University of Wisconsin at Madison, Madison, WI53706
| | - Vijaya K. Balakrishnan
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
| | - Ronald A. Merrill
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA52242
| | - Pankaj S. Parihar
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
| | - Kirill Konovolov
- Chemistry Department, University of Wisconsin at Madison, Madison, WI53706
| | - Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
- Molecular and Cellular Pharmacology Program, University of Wisconsin at Madison, Madison, WI53706
| | - Zhen Xu
- Protein and Crystallography Facility, University of Iowa, Iowa City, IA52242
| | - Hui Wei
- The Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY10027
| | - Ramya Sundaresan
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA02215
| | | | - Mark R. Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL36688
| | - Alla Musiyenko
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL36688
| | - Wendy K. Chung
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA02215
| | - Richard E. Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL36688
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
- Biophysics Program, University of Wisconsin at Madison, Madison, WI53706
- Molecular and Cellular Pharmacology Program, University of Wisconsin at Madison, Madison, WI53706
| | - Xuhui Huang
- Biophysics Program, University of Wisconsin at Madison, Madison, WI53706
- Chemistry Department, University of Wisconsin at Madison, Madison, WI53706
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA52242
| | - Yongna Xing
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI53705
- Biophysics Program, University of Wisconsin at Madison, Madison, WI53706
| |
Collapse
|
18
|
Sedlackova L, Sterbova K, Vlckova M, Seeman P, Zarubova J, Marusic P, Krsek P, Krijtova H, Musilova A, Lassuthova P. Yield of exome sequencing in patients with developmental and epileptic encephalopathies and inconclusive targeted gene panel. Eur J Paediatr Neurol 2024; 48:17-29. [PMID: 38008000 DOI: 10.1016/j.ejpn.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/28/2023]
Abstract
OBJECTIVE Developmental and epileptic encephalopathies (DEEs) are a group of severe, early-onset epilepsies characterised by refractory seizures, developmental delay, or regression and generally poor prognosis. DEE are now known to have an identifiable molecular genetic basis and are usually examined using a gene panel. However, for many patients, the genetic cause has still not been identified. The aims of this study were to identify causal variants for DEE in patients for whom the previous examination with a gene panel did not determine their genetic diagnosis. It also aims for a detailed description and broadening of the phenotypic spectrum of several rare DEEs. METHODS In the last five years (2015-2020), 141 patients from all over the Czech Republic were referred to our department for genetic testing in association with their diagnosis of epilepsy. All patients underwent custom-designed gene panel testing prior to enrolment into the study, and their results were inconclusive. We opted for whole exome sequencing (WES) to identify the cause of their disorder. If a causal or potentially causal variant was identified, we performed a detailed clinical evaluation and phenotype-genotype correlation study to better describe the specific rare subtypes. RESULTS Explanatory causative variants were detected in 20 patients (14%), likely pathogenic variants that explain the epilepsy in 5 patients (3.5%) and likely pathogenic variants that do not fully explain the epilepsy in 11 patients (7.5%), and variants in candidate genes in 4 patients (3%). Variants were mostly de novo 29/40 (72.5%). SIGNIFICANCE WES enables us to identify the cause of the disease in additional patients, even after gene panel testing. It is very important to perform a WES in DEE patients as soon as possible, since it will spare the patients and their families many years of a diagnostic odyssey. In particular, patients with rare epilepsies might significantly benefit from this approach, and we propose using WES as a new standard in the diagnosis of DEE instead of targeted gene panel testing.
Collapse
Affiliation(s)
- Lucie Sedlackova
- Neurogenetic Laboratory, Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Katalin Sterbova
- Department of Paediatric Neurology, Second Faculty of Medicine, Motol Epilepsy Center, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Marketa Vlckova
- Department of Biology and Medical Genetics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Pavel Seeman
- Neurogenetic Laboratory, Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic; Department of Medical Genetics, Masaryk Hospital, Ústí nad Labem, Czech Republic.
| | - Jana Zarubova
- Department of Neurology, Motol Epilepsy Center, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Petr Marusic
- Department of Neurology, Motol Epilepsy Center, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Pavel Krsek
- Department of Paediatric Neurology, Second Faculty of Medicine, Motol Epilepsy Center, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Hana Krijtova
- Department of Neurology, Motol Epilepsy Center, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Alena Musilova
- Neurogenetic Laboratory, Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| | - Petra Lassuthova
- Neurogenetic Laboratory, Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Full Member of the ERN EpiCARE, Czech Republic.
| |
Collapse
|
19
|
Alhajaj G, Lacroix C, Trakadis Y, Garfinkle J, Srour M. An in-frame deletion affecting the critical acid loop of PPP2R5D is associated with a neonatal lethal form of PPP2R5D-related neurodevelopmental disorder. Am J Med Genet A 2023; 191:2416-2421. [PMID: 37248744 DOI: 10.1002/ajmg.a.63307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/31/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023]
Abstract
Heterozygous pathogenic variants in PPP2R5D gene are associated with PPP2R5D-related neurodevelopmental disorder, a rare autosomal dominant condition, characterized by neurodevelopmental impairment in childhood, macrocephaly/megalencephaly, hypotonia, epilepsy, and dysmorphic features. Up-to-date, only approximately 100 cases have been published in the literature and the full phenotypic and genotypic spectrum have not yet been fully described. PPP2R5D gene encodes the B56δ subunit of the PP2A enzyme complex. We describe a neonatal form of PPP2R5D-related disorder with early infantile death, caused by a novel in-frame deletion causing loss of 8 amino acids and insertion of serine at position 201 (p.Phe194_Pro201delinsSer) of the B56δ subunit. This deletion is predicted to disrupt a critical acidic loop of amino acids important for binding other subunits of the PP2A enzyme complex, and harbors many of the residues previously reported to cause a mild-moderate form of this condition. This report describes a neonatal lethal presentation of the PPP2R5D-related neurodevelopmental disorder and provides additional evidence that disruption of the acidic loop is an important pathomechanism underlying PPP2R5D-related disorder.
Collapse
Affiliation(s)
- Ghadd Alhajaj
- Department of Pediatrics, Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia
- Department of Pediatrics, Division of Pediatric Neurology, McGill University Health Center, Montreal, Quebec, Canada
| | - Caroline Lacroix
- Department of Diagnostic Radiology, McGill University Health Center, Montreal, Quebec, Canada
| | - Yannis Trakadis
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal, Quebec, Canada
| | - Jarred Garfinkle
- Department of Pediatrics, Division of Neonatology, McGill University Health Center, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Myriam Srour
- Department of Pediatrics, Division of Pediatric Neurology, McGill University Health Center, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Qian Y, Jiang Y, Wang J, Li G, Wu B, Zhou Y, Xu X, Wang H. Novel Variants of PPP2R1A in Catalytic Subunit Binding Domain and Genotype-Phenotype Analysis in Neurodevelopmentally Delayed Patients. Genes (Basel) 2023; 14:1750. [PMID: 37761890 PMCID: PMC10531206 DOI: 10.3390/genes14091750] [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: 07/20/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of high-incidence rare diseases with genetic heterogeneity. PPP2R1A, the regulatory subunit of protein phosphatase 2A, is a recently discovered gene associated with NDDs. Whole/clinical exome sequencing was performed in five patients with a family with NDDs. In vitro experiments were performed to evaluate the mutants' expression and interactions with the complex. The genotype-phenotype correlations of reported cases as well as our patients with PPP2R1A variants were reviewed. We reported five unrelated individuals with PPP2R1A variants, including two novel missense variants and one frameshift variant. The protein expression of the Arg498Leu variant was less than that of the wild-type protein, the frameshift variant Asn282Argfs*14 was not decreased but truncated, and these two variants impaired the interactions with endogenous PPP25RD and PPP2CA. Furthermore, we found that pathogenic variants clustered in HEAT repeats V, VI and VII, and patients with the Met180Val/Thr variants had macrocephaly, severe ID and hypotonia, but no epilepsy, whereas those with Arg258 amino acid changes had microcephaly, while a few had epilepsy or feeding problems. In this study, we reported five NDD patients with PPP2R1A gene variants and expanded PPP2R1A pathogenic variant spectrum. The genotype and phenotype association findings provide reminders regarding the prognostication and evidence for genetic counseling.
Collapse
Affiliation(s)
- Yanyan Qian
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.Q.); (Y.J.); (G.L.); (B.W.)
| | - Yinmo Jiang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.Q.); (Y.J.); (G.L.); (B.W.)
| | - Ji Wang
- Neurology Department, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (J.W.); (Y.Z.)
| | - Gang Li
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.Q.); (Y.J.); (G.L.); (B.W.)
| | - Bingbing Wu
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.Q.); (Y.J.); (G.L.); (B.W.)
| | - Yuanfeng Zhou
- Neurology Department, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (J.W.); (Y.Z.)
| | - Xiu Xu
- Department of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Huijun Wang
- Center for Molecular Medicine, Pediatrics Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China; (Y.Q.); (Y.J.); (G.L.); (B.W.)
| |
Collapse
|
21
|
Smolen KA, Papke CM, Swingle MR, Musiyenko A, Li C, Salter EA, Camp AD, Honkanen RE, Kettenbach AN. Quantitative proteomics and phosphoproteomics of PP2A-PPP2R5D variants reveal deregulation of RPS6 phosphorylation via converging signaling cascades. J Biol Chem 2023; 299:105154. [PMID: 37572851 PMCID: PMC10485637 DOI: 10.1016/j.jbc.2023.105154] [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: 01/11/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/14/2023] Open
Abstract
Genetic germline variants of PPP2R5D (encoding: phosphoprotein phosphatase 2 regulatory protein 5D) result in PPP2R5D-related disorder (Jordan's Syndrome), which is characterized by intellectual disability, hypotonia, seizures, macrocephaly, autism spectrum disorder, and delayed motor skill development. The disorder originates from de novo single nucleotide mutations, generating missense variants that act in a dominant manner. Pathogenic mutations altering 13 different amino acids have been identified, with the E198K variant accounting for ∼40% of reported cases. However, the generation of a heterozygous E198K variant cell line to study the molecular effects of the pathogenic mutation has been challenging. Here, we use CRISPR-PRIME genomic editing to introduce a transition (c.592G>A) in a single PPP2R5D allele in HEK293 cells, generating E198K-heterozygous lines to complement existing E420K variant lines. We generate global protein and phosphorylation profiles of WT, E198K, and E420K cell lines and find unique and shared changes between variants and WT cells in kinase- and phosphatase-controlled signaling cascades. We observed ribosomal protein S6 (RPS6) hyperphosphorylation as a shared signaling alteration, indicative of increased ribosomal protein S6-kinase activity. Treatment with rapamycin or an RPS6-kinase inhibitor (LY2584702) suppressed RPS6 phosphorylation in both, suggesting upstream activation of mTORC1/p70S6K. Intriguingly, our data suggests ERK-dependent activation of mTORC1 in both E198K and E420K variant cells, with additional AKT-mediated mTORC1 activation in the E420K variant. Thus, although upstream activation of mTORC1 differs between PPP2R5D-related disorder genotypes, inhibition of mTORC1 or RPS6 kinases warrants further investigation as potential therapeutic strategies for patients.
Collapse
Affiliation(s)
- Kali A Smolen
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Cinta M Papke
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Mark R Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Alla Musiyenko
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Chenchen Li
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - E Alan Salter
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Ashley D Camp
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Richard E Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA.
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA.
| |
Collapse
|
22
|
Konovalov KA, Wu CG, Qiu Y, Balakrishnan VK, Parihar PS, O’Connor MS, Xing Y, Huang X. Disease mutations and phosphorylation alter the allosteric pathways involved in autoinhibition of protein phosphatase 2A. J Chem Phys 2023; 158:215101. [PMID: 37260014 PMCID: PMC10238128 DOI: 10.1063/5.0150272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023] Open
Abstract
Mutations in protein phosphatase 2A (PP2A) are connected to intellectual disability and cancer. It has been hypothesized that these mutations might disrupt the autoinhibition and phosphorylation-induced activation of PP2A. Since they are located far from both the active and substrate binding sites, it is unclear how they exert their effect. We performed allosteric pathway analysis based on molecular dynamics simulations and combined it with biochemical experiments to investigate the autoinhibition of PP2A. In the wild type (WT), the C-arm of the regulatory subunit B56δ obstructs the active and substrate binding sites exerting a dual autoinhibition effect. We find that the disease mutant, E198K, severely weakens the allosteric pathways that stabilize the C-arm in the WT. Instead, the strongest allosteric pathways in E198K take a different route that promotes exposure of the substrate binding site. To facilitate the allosteric pathway analysis, we introduce a path clustering algorithm for lumping pathways into channels. We reveal remarkable similarities between the allosteric channels of E198K and those in phosphorylation-activated WT, suggesting that the autoinhibition can be alleviated through a conserved mechanism. In contrast, we find that another disease mutant, E200K, which is in spatial proximity of E198, does not repartition the allosteric pathways leading to the substrate binding site; however, it may still induce exposure of the active site. This finding agrees with our biochemical data, allowing us to predict the activity of PP2A with the phosphorylated B56δ and provide insight into how disease mutations in spatial proximity alter the enzymatic activity in surprisingly different mechanisms.
Collapse
Affiliation(s)
- Kirill A. Konovalov
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | - Yunrui Qiu
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Vijaya Kumar Balakrishnan
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Pankaj Singh Parihar
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Michael S. O’Connor
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Yongna Xing
- Authors to whom correspondence should be addressed: and
| | - Xuhui Huang
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
23
|
Rihar N, Krgovic D, Kokalj-Vokač N, Stangler-Herodez S, Zorc M, Dovc P. Identification of potentially pathogenic variants for autism spectrum disorders using gene-burden analysis. PLoS One 2023; 18:e0273957. [PMID: 37167322 PMCID: PMC10174571 DOI: 10.1371/journal.pone.0273957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023] Open
Abstract
Gene- burden analyses have lately become a very successful way for the identification of genes carrying risk variants underlying the analysed disease. This approach is also suitable for complex disorders like autism spectrum disorder (ASD). The gene-burden analysis using Testing Rare Variants with Public Data (TRAPD) software was conducted on whole exome sequencing data of Slovenian patients with ASD to determine potentially novel disease risk variants in known ASD-associated genes as well as in others. To choose the right control group for testing, principal component analysis based on the 1000 Genomes and ASD cohort samples was conducted. The subsequent protein structure and ligand binding analysis usingI-TASSER package were performed to detect changes in protein structure and ligand binding to determine a potential pathogenic consequence of observed mutation. The obtained results demonstrate an association of two variants-p.Glu198Lys (PPP2R5D:c.592G>A) and p.Arg253Gln (PPP2R5D:c.758G>A) with the ASD. Substitution p.Glu198Lys (PPP2R5D:c.592G>A) is a variant, previously described as pathogenic in association with ASD combined with intellectual disability, whereas p.Arg253Gln (PPP2R5D:c.758G>A) has not been described as an ASD-associated pathogenic variant yet. The results indicate that the filtering process was suitable and could be used in the future for detection of novel pathogenic variants when analysing groups of ASD patients.
Collapse
Affiliation(s)
- Nika Rihar
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
| | - Danijela Krgovic
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
- Maribor Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Nadja Kokalj-Vokač
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
- Maribor Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Spela Stangler-Herodez
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
- Maribor Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Minja Zorc
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Dovc
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
24
|
Fevga C, Tesson C, Carreras Mascaro A, Courtin T, van Coller R, Sakka S, Ferraro F, Farhat N, Bardien S, Damak M, Carr J, Ferrien M, Boumeester V, Hundscheid J, Grillenzoni N, Kessissoglou IA, Kuipers DJS, Quadri M, Corvol JC, Mhiri C, Hassan BA, Breedveld GJ, Lesage S, Mandemakers W, Brice A, Bonifati V. PTPA variants and impaired PP2A activity in early-onset parkinsonism with intellectual disability. Brain 2023; 146:1496-1510. [PMID: 36073231 PMCID: PMC10115167 DOI: 10.1093/brain/awac326] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The protein phosphatase 2A complex (PP2A), the major Ser/Thr phosphatase in the brain, is involved in a number of signalling pathways and functions, including the regulation of crucial proteins for neurodegeneration, such as alpha-synuclein, tau and LRRK2. Here, we report the identification of variants in the PTPA/PPP2R4 gene, encoding a major PP2A activator, in two families with early-onset parkinsonism and intellectual disability. We carried out clinical studies and genetic analyses, including genome-wide linkage analysis, whole-exome sequencing, and Sanger sequencing of candidate variants. We next performed functional studies on the disease-associated variants in cultured cells and knock-down of ptpa in Drosophila melanogaster. We first identified a homozygous PTPA variant, c.893T>G (p.Met298Arg), in patients from a South African family with early-onset parkinsonism and intellectual disability. Screening of a large series of additional families yielded a second homozygous variant, c.512C>A (p.Ala171Asp), in a Libyan family with a similar phenotype. Both variants co-segregate with disease in the respective families. The affected subjects display juvenile-onset parkinsonism and intellectual disability. The motor symptoms were responsive to treatment with levodopa and deep brain stimulation of the subthalamic nucleus. In overexpression studies, both the PTPA p.Ala171Asp and p.Met298Arg variants were associated with decreased PTPA RNA stability and decreased PTPA protein levels; the p.Ala171Asp variant additionally displayed decreased PTPA protein stability. Crucially, expression of both variants was associated with decreased PP2A complex levels and impaired PP2A phosphatase activation. PTPA orthologue knock-down in Drosophila neurons induced a significant impairment of locomotion in the climbing test. This defect was age-dependent and fully reversed by L-DOPA treatment. We conclude that bi-allelic missense PTPA variants associated with impaired activation of the PP2A phosphatase cause autosomal recessive early-onset parkinsonism with intellectual disability. Our findings might also provide new insights for understanding the role of the PP2A complex in the pathogenesis of more common forms of neurodegeneration.
Collapse
Affiliation(s)
- Christina Fevga
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Christelle Tesson
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Ana Carreras Mascaro
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Thomas Courtin
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Génétique, DMU BioGeM, Paris, France
| | - Riaan van Coller
- Department of Neurology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Salma Sakka
- Research Unit in Neurogenetics, Clinical Investigation Center (CIC) at the CHU Habib Bourguiba, Sfax, Tunisia
| | - Federico Ferraro
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Nouha Farhat
- Research Unit in Neurogenetics, Clinical Investigation Center (CIC) at the CHU Habib Bourguiba, Sfax, Tunisia
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Mariem Damak
- Research Unit in Neurogenetics, Clinical Investigation Center (CIC) at the CHU Habib Bourguiba, Sfax, Tunisia
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Mélanie Ferrien
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Valerie Boumeester
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Jasmijn Hundscheid
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Nicola Grillenzoni
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Irini A Kessissoglou
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Demy J S Kuipers
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Marialuisa Quadri
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Jean-Christophe Corvol
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neurologie, Centre d'Investigation Clinique Neurosciences, DMU Neuroscience, Paris, France
| | - Chokri Mhiri
- Research Unit in Neurogenetics, Clinical Investigation Center (CIC) at the CHU Habib Bourguiba, Sfax, Tunisia
| | - Bassem A Hassan
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Guido J Breedveld
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Suzanne Lesage
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Wim Mandemakers
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Alexis Brice
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Génétique, DMU BioGeM, Paris, France
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
25
|
Wu CG, Balakrishnan VK, Parihar PS, Konovolov K, Chen YC, Merrill RA, Wei H, Carragher B, Sundaresan R, Cui Q, Wadzinski BE, Swingle MR, Musiyenko A, Honkanen R, Chung WK, Suzuki A, Strack S, Huang X, Xing Y. Extended regulation interface coupled to the allosteric network and disease mutations in the PP2A-B56δ holoenzyme. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.530109. [PMID: 37066309 PMCID: PMC10103954 DOI: 10.1101/2023.03.09.530109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
An increasing number of mutations associated with devastating human diseases are diagnosed by whole-genome/exon sequencing. Recurrent de novo missense mutations have been discovered in B56δ (encoded by PPP2R5D), a regulatory subunit of protein phosphatase 2A (PP2A), that cause intellectual disabilities (ID), macrocephaly, Parkinsonism, and a broad range of neurological symptoms. Single-particle cryo-EM structures show that the PP2A-B56δ holoenzyme possesses closed latent and open active forms. In the closed form, the long, disordered arms of B56δ termini fold against each other and the holoenzyme core, establishing dual autoinhibition of the phosphatase active site and the substrate-binding protein groove. The resulting interface spans over 190 Å and harbors unfavorable contacts, activation phosphorylation sites, and nearly all residues with ID-associated mutations. Our studies suggest that this dynamic interface is close to an allosteric network responsive to activation phosphorylation and altered globally by mutations. Furthermore, we found that ID mutations perturb the activation phosphorylation rates, and the severe variants significantly increase the mitotic duration and error rates compared to the wild variant.
Collapse
Affiliation(s)
- Cheng-Guo Wu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
- Biophysics program, University of Wisconsin at Madison, Wisconsin 53706, USA
| | - Vijaya K. Balakrishnan
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | - Pankaj S. Parihar
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | - Kirill Konovolov
- Chemistry Department, University of Wisconsin at Madison, Wisconsin 53706, USA
| | - Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
- Molecular and Cellular Pharmacology program, University of Wisconsin at Madison, Wisconsin 53706, USA
| | - Ronald A Merrill
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Hui Wei
- New York Structural biology Center, New York, NY 10027, USA
| | | | - Ramya Sundaresan
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | - Qiang Cui
- Department of Chemistry, Metcalf Center for Science & Engineering, Boston University, Boston, MA 02215, USA
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mark R. Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Alla Musiyenko
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Richard Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
- Biophysics program, University of Wisconsin at Madison, Wisconsin 53706, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Xuhui Huang
- Biophysics program, University of Wisconsin at Madison, Wisconsin 53706, USA
- Chemistry Department, University of Wisconsin at Madison, Wisconsin 53706, USA
| | - Yongna Xing
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, Wisconsin 53705, USA
- Biophysics program, University of Wisconsin at Madison, Wisconsin 53706, USA
| |
Collapse
|
26
|
KA S, CM P, Swingle MR, A M, C L, AD C, RE H, AN K. Quantitative proteomics and phosphoproteomics of PPP2R5D variants reveal deregulation of RPS6 phosphorylation through converging signaling cascades. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534397. [PMID: 37034727 PMCID: PMC10081281 DOI: 10.1101/2023.03.27.534397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Variants in the phosphoprotein phosphatase-2 regulatory protein-5D gene ( PPP2R5D ) cause the clinical phenotype of Jordan's Syndrome (PPP2R5D-related disorder), which includes intellectual disability, hypotonia, seizures, macrocephaly, autism spectrum disorder and delayed motor skill development. The disorder originates from de novo single nucleotide mutations, generating missense variants that act in a dominant manner. Pathogenic mutations altering 13 different amino acids have been identified, with the E198K variant accounting for ∼40% of reported cases. Here, we use CRISPR-PRIME genomic editing to introduce a transition (c.592G>A) in the PPP2R5D allele in a heterozygous manner in HEK293 cells, generating E198K-heterozygous lines to complement existing E420K variant lines. We generate global protein and phosphorylation profiles of wild-type, E198K, and E420K cell lines and find unique and shared changes between variants and wild-type cells in kinase- and phosphatase-controlled signaling cascades. As shared signaling alterations, we observed ribosomal protein S6 (RPS6) hyperphosphorylation, indicative of increased ribosomal protein S6-kinase activity. Rapamycin treatment suppressed RPS6 phosphorylation in both, suggesting activation of mTORC1. Intriguingly, our data suggest AKT-dependent (E420K) and -independent (E198K) activation of mTORC1. Thus, although upstream activation of mTORC1 differs between PPP2R5D-related disorder genotypes, treatment with rapamycin or a p70S6K inhibitor warrants further investigation as potential therapeutic strategies for patients.
Collapse
Affiliation(s)
- Smolen KA
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Papke CM
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - MR Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Musiyenko A
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Li C
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Camp AD
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Honkanen RE
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Kettenbach AN
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
27
|
Schwab K, Coronel L, Riege K, Sacramento EK, Rahnis N, Häckes D, Cirri E, Groth M, Hoffmann S, Fischer M. Multi-omics analysis identifies RFX7 targets involved in tumor suppression and neuronal processes. Cell Death Discov 2023; 9:80. [PMID: 36864036 PMCID: PMC9981735 DOI: 10.1038/s41420-023-01378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Recurrently mutated in lymphoid neoplasms, the transcription factor RFX7 is emerging as a tumor suppressor. Previous reports suggested that RFX7 may also have a role in neurological and metabolic disorders. We recently reported that RFX7 responds to p53 signaling and cellular stress. Furthermore, we found RFX7 target genes to be dysregulated in numerous cancer types also beyond the hematological system. However, our understanding of RFX7's target gene network and its role in health and disease remains limited. Here, we generated RFX7 knock-out cells and employed a multi-omics approach integrating transcriptome, cistrome, and proteome data to obtain a more comprehensive picture of RFX7 targets. We identify novel target genes linked to RFX7's tumor suppressor function and underscoring its potential role in neurological disorders. Importantly, our data reveal RFX7 as a mechanistic link that enables the activation of these genes in response to p53 signaling.
Collapse
Affiliation(s)
- Katjana Schwab
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Luis Coronel
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Konstantin Riege
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Erika K. Sacramento
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Norman Rahnis
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - David Häckes
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Emilio Cirri
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Marco Groth
- grid.418245.e0000 0000 9999 5706Core Facility for Next-Generation Sequencing, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Steve Hoffmann
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745, Jena, Germany.
| |
Collapse
|
28
|
Verbinnen I, Procknow SS, Lenaerts L, Reynhout S, Mehregan A, Ulens C, Janssens V, King KA. Clinical and molecular characteristics of a novel rare de novo variant in PPP2CA in a patient with a developmental disorder, autism, and epilepsy. Front Cell Dev Biol 2022; 10:1059938. [DOI: 10.3389/fcell.2022.1059938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
PP2A-related (neuro) developmental disorders are a family of genetic diseases caused by a heterozygous alteration in one of several genes encoding a subunit of type 2A protein phosphatases. Reported affected genes, so far, are PPP2R5D, encoding the PP2A regulatory B56δ subunit; PPP2R1A, encoding the scaffolding Aα subunit; and PPP2CA, encoding the catalytic Cα subunit—in that order of frequency. Patients with a pathogenic de novo mutation in one of these genes, in part, present with overlapping features, such as generalized hypotonia, intellectual and developmental delay, facial dysmorphologies, seizures, and autistic features, and, in part, with opposite features, e.g., smaller versus larger head sizes or normal versus absent corpus callosum. Molecular variant characterization has been consistent so far with loss-of-function or dominant-negative disease mechanisms for all three affected genes. Here, we present a case report of another PPP2CA-affected individual with a novel de novo missense variant, resulting in a one-amino acid substitution in the Cα subunit: p.Cys196Arg. Biochemical characterization of the variant revealed its pathogenicity, as it appeared severely catalytically impaired, showed mildly affected A subunit binding, and moderately decreased binding to B/B55, B”/PR72, and all B56 subunits, except B56γ1. Carboxy-terminal methylation appeared unaffected, as was binding to B”’/STRN3—all being consistent with a partial loss of function. Clinically, the girl presented with mild-to-moderate developmental delay, a full-scale IQ of 83, mild dysmorphic facial features, tonic–clonic seizures, and autistic behaviors. Brain MRI appeared normal. We conclude that this individual falls within the milder end of the clinical and molecular spectrum of previously reported PPP2CA cases.
Collapse
|
29
|
A novel nonsense mutation in PPP2R5D is associated with neurodevelopmental disorders and shows incomplete penetrance in a Chinese pedigree. Clin Neurol Neurosurg 2022; 223:107524. [DOI: 10.1016/j.clineuro.2022.107524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022]
|
30
|
Murtaza N, Cheng AA, Brown CO, Meka DP, Hong S, Uy JA, El-Hajjar J, Pipko N, Unda BK, Schwanke B, Xing S, Thiruvahindrapuram B, Engchuan W, Trost B, Deneault E, Calderon de Anda F, Doble BW, Ellis J, Anagnostou E, Bader GD, Scherer SW, Lu Y, Singh KK. Neuron-specific protein network mapping of autism risk genes identifies shared biological mechanisms and disease-relevant pathologies. Cell Rep 2022; 41:111678. [DOI: 10.1016/j.celrep.2022.111678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
|
31
|
Vaneynde P, Verbinnen I, Janssens V. The role of serine/threonine phosphatases in human development: Evidence from congenital disorders. Front Cell Dev Biol 2022; 10:1030119. [PMID: 36313552 PMCID: PMC9608770 DOI: 10.3389/fcell.2022.1030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022] Open
Abstract
Reversible protein phosphorylation is a fundamental regulation mechanism in eukaryotic cell and organismal physiology, and in human health and disease. Until recently, and unlike protein kinases, mutations in serine/threonine protein phosphatases (PSP) had not been commonly associated with disorders of human development. Here, we have summarized the current knowledge on congenital diseases caused by mutations, inherited or de novo, in one of 38 human PSP genes, encoding a monomeric phosphatase or a catalytic subunit of a multimeric phosphatase. In addition, we highlight similar pathogenic mutations in genes encoding a specific regulatory subunit of a multimeric PSP. Overall, we describe 19 affected genes, and find that most pathogenic variants are loss-of-function, with just a few examples of gain-of-function alterations. Moreover, despite their widespread tissue expression, the large majority of congenital PSP disorders are characterised by brain-specific abnormalities, suggesting a generalized, major role for PSPs in brain development and function. However, even if the pathogenic mechanisms are relatively well understood for a small number of PSP disorders, this knowledge is still incomplete for most of them, and the further identification of downstream targets and effectors of the affected PSPs is eagerly awaited through studies in appropriate in vitro and in vivo disease models. Such lacking studies could elucidate the exact mechanisms through which these diseases act, and possibly open up new therapeutic avenues.
Collapse
Affiliation(s)
- Pieter Vaneynde
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
- *Correspondence: Veerle Janssens,
| |
Collapse
|
32
|
Oyama N, Vaneynde P, Reynhout S, Pao EM, Timms A, Fan X, Foss K, Derua R, Janssens V, Chung W, Mirzaa GM. Clinical, neuroimaging and molecular characteristics of PPP2R5D-related neurodevelopmental disorders: an expanded series with functional characterisation and genotype-phenotype analysis. J Med Genet 2022; 60:511-522. [PMID: 36216457 DOI: 10.1136/jmg-2022-108713] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/11/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Variants in PPP2R5D, affecting the regulatory B56δ subunit of protein phosphatase 2A (PP2A), have been identified in individuals with neurodevelopmental abnormalities. However, the molecular and clinical spectra remain incompletely understood. METHODS Individuals with PPP2R5D variants were enrolled through Simons Variation in Individuals Project/Simons Searchlight. Data were collected from medical history interviews, medical record review, online validated instruments and neuroimaging review. Genetic variants were biochemically characterised. RESULTS We studied 76 individuals with PPP2R5D variants, including 68 with pathogenic de novo variants, four with a variant of uncertain significance (VUS) and four siblings with a novel dominantly inherited pathogenic variant. Among 13 pathogenic variants, eight were novel and two (p.Glu198Lys and p.Glu200Lys) were highly recurrent. Functional analysis revealed impaired PP2A A/C-subunit binding, decreased short linear interaction motif-dependent substrate binding or both-with the most severe phenotypes associated with variants that completely retained one of these binding characteristics and lost the other-further supporting a dominant-negative disease mechanism. p.Glu198Lys showed the highest C-binding defect and a more severe clinical phenotype. The inherited p.Glu197Gly variant had a mild substrate binding defect, and three of four VUS had no biochemical impact. Common clinical phenotypes were language, intellectual or learning disabilities (80.6%), hypotonia (75.0%), macrocephaly (66.7%), seizures (45.8%) and autism spectrum disorder (26.4%). The mean composite Vineland score was 59.8, and most participants were in the 'moderate to low' and 'low' adaptive levels in all domains. CONCLUSION Our study delineates the most common features of PPP2R5D-related neurodevelopmental disorders, expands the clinical and molecular spectrum and identifies genotype-phenotype correlations.
Collapse
Affiliation(s)
- Nora Oyama
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Pieter Vaneynde
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Sara Reynhout
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Emily M Pao
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Andrew Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Xiao Fan
- Department of Pediatrics, Columbia University, New York City, New York, USA
| | - Kimberly Foss
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,SyBioMa, University of Leuven (KU Leuven), Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Wendy Chung
- Department of Pediatrics, Columbia University, New York City, New York, USA.,Department of Medicine, Columbia University, New York City, New York, USA
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA .,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
33
|
Madaan P, Kaur A, Saini L, Paria P, Vyas S, Sharma AR, Sahu JK. PPP2R5D-Related Neurodevelopmental Disorder or Developmental and Epileptic Encephalopathy?: A Novel Phenotypic Description and Review of Published Cases. Neuropediatrics 2022; 53:20-25. [PMID: 34448180 DOI: 10.1055/s-0041-1733984] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Protein phosphatase 2 regulatory subunit B' delta (PPP2R5D)-related neurodevelopmental disorder is caused by pathogenic variations in the PPP2R5D gene, product of which is involved in dephosphorylation. This is a rare disorder with description limited to case reports. Its phenotypic spectrum has expanded over the last decade. METHODS We report a child with a developmental and epileptic encephalopathy phenotype with a pathogenic PPP2R5D variant. This phenotype has not been previously reported. We also reviewed the previously published reports of patients with this disorder. RESULTS Including the index child, 28 cases (15 girls) were identified from nine relevant research items for analysis. All patients had developmental delay. History of seizures was observed in seven patients while macrocephaly was seen in nearly 80% of patients. Nonneurological manifestations were observed in 13 patients with the most common one being ophthalmological manifestations. The most common genetic variation was c.G592A (p.E198K). The common phenotypic associations of this variation were developmental delay, macrocephaly (11/15), and epilepsy (6/15). CONCLUSION PPP2R5D gene variations should be suspected in children with developmental delay, autistic features, macrocephaly with or without epilepsy in the absence of any clear etiology. Dysmorphic features might provide a diagnostic clue. DEE phenotype may also be the presenting feature and might be an underreported entity.
Collapse
Affiliation(s)
- Priyanka Madaan
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amrit Kaur
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Lokesh Saini
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,Department of Pediatrics, All India Institute of Medical Sciences, Jodhpur, India
| | - Pradip Paria
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sameer Vyas
- Department of Radiodiagnosis and Imaging (Section of Neuroimaging and Interventional Radiology), Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amit R Sharma
- Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jitendra K Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
34
|
Lombardo MV, Busuoli EM, Schreibman L, Stahmer AC, Pramparo T, Landi I, Mandelli V, Bertelsen N, Barnes CC, Gazestani V, Lopez L, Bacon EC, Courchesne E, Pierce K. Pre-treatment clinical and gene expression patterns predict developmental change in early intervention in autism. Mol Psychiatry 2021; 26:7641-7651. [PMID: 34341515 PMCID: PMC8872998 DOI: 10.1038/s41380-021-01239-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/29/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
Early detection and intervention are believed to be key to facilitating better outcomes in children with autism, yet the impact of age at treatment start on the outcome is poorly understood. While clinical traits such as language ability have been shown to predict treatment outcome, whether or not and how information at the genomic level can predict treatment outcome is unknown. Leveraging a cohort of toddlers with autism who all received the same standardized intervention at a very young age and provided a blood sample, here we find that very early treatment engagement (i.e., <24 months) leads to greater gains while controlling for time in treatment. Pre-treatment clinical behavioral measures predict 21% of the variance in the rate of skill growth during early intervention. Pre-treatment blood leukocyte gene expression patterns also predict the rate of skill growth, accounting for 13% of the variance in treatment slopes. Results indicated that 295 genes can be prioritized as driving this effect. These treatment-relevant genes highly interact at the protein level, are enriched for differentially histone acetylated genes in autism postmortem cortical tissue, and are normatively highly expressed in a variety of subcortical and cortical areas important for social communication and language development. This work suggests that pre-treatment biological and clinical behavioral characteristics are important for predicting developmental change in the context of early intervention and that individualized pre-treatment biology related to histone acetylation may be key.
Collapse
Affiliation(s)
- Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.
- Department of Psychiatry, Autism Research Centre, University of Cambridge, Cambridge, UK.
| | - Elena Maria Busuoli
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Laura Schreibman
- Department of Psychology, University of California, San Diego, La Jolla, CA, USA
| | - Aubyn C Stahmer
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
| | - Tiziano Pramparo
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Isotta Landi
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Veronica Mandelli
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Natasha Bertelsen
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Cynthia Carter Barnes
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Vahid Gazestani
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Linda Lopez
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Elizabeth C Bacon
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Eric Courchesne
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Karen Pierce
- Department of Neurosciences, Autism Center of Excellence, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Maines E, Franceschi R, Martinelli D, Soli F, Lepri FR, Piccoli G, Soffiati M. Hypoglycemia due to PI3K/AKT/mTOR signaling pathway defects: two novel cases and review of the literature. Hormones (Athens) 2021; 20:623-640. [PMID: 33876391 DOI: 10.1007/s42000-021-00287-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/25/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The PI3K/AKT/mTOR signaling pathway is important for the regulation of multiple biological processes, including cellular growth and glucose metabolism. Defects of the PI3K/AKT/mTOR signaling pathway are not usually considered among the genetic causes of recurrent hypoglycemia in childhood. However, accumulating evidence links hypoglycemia with defects of this pathway. CASE REPORTS AND REVIEW We describe here two cases of macrocephaly and hypoglycemia bearing genetic defects in genes involved in the PI3K/AKT/mTOR pathway. The first patient was diagnosed with a PTEN hamartoma tumour syndrome (PTHS) due to the de novo germline missense mutation c.[492 + 1G > A] of the PTEN gene. The second patient presented the autosomal dominant mental retardation-35 (MDR35) due to the heterozygous missense mutation c.592G > A in the PPP2R5D gene. A review of the literature on hypoglycemia and PI3K/AKT/mTOR signaling pathway defects, with a special focus on the metabolic characterization of hypoglycemia, is included. CONCLUSIONS PI3K/AKT/mTOR pathway defects should be included in the differential diagnosis of patients with hypoglycemia and macrocephaly. Clinical suspicion and molecular confirmation are important, not just for an accurate genetic counselling but also for defining the follow-up management, including cancer surveillance. The biochemical profile of hypoglycemia varies among patients. While most patients are characterized by low plasmatic insulin levels, hyperinsulinemia has also been observed. Large patient cohorts are needed to gain a comprehensive profile of the biochemical patterns of hypoglycemia in such defects and eventually guide targeted therapeutic interventions.
Collapse
Affiliation(s)
- Evelina Maines
- Division of Pediatrics, S. Chiara General Hospital, Largo Medaglie d'oro, 9, 38122, Trento, Italy.
| | - Roberto Franceschi
- Division of Pediatrics, S. Chiara General Hospital, Largo Medaglie d'oro, 9, 38122, Trento, Italy
| | - Diego Martinelli
- Division of Metabolism and Research Unit of Metabolic Biochemistry, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fiorenza Soli
- Division of Medical Genetics, S. Chiara General Hospital, Trento, Italy
| | | | - Giovanni Piccoli
- CIBIO - Centre for Integrative Biology, Università Degli Studi Di Trento, Italy & Dulbecco Telethon Institute, Trento, Italy
| | - Massimo Soffiati
- Division of Pediatrics, S. Chiara General Hospital, Largo Medaglie d'oro, 9, 38122, Trento, Italy
| |
Collapse
|
36
|
Fazia T, Marzanati D, Carotenuto AL, Beecham A, Hadjixenofontos A, McCauley JL, Saddi V, Piras M, Bernardinelli L, Gentilini D. Homozygosity Haplotype and Whole-Exome Sequencing Analysis to Identify Potentially Functional Rare Variants Involved in Multiple Sclerosis among Sardinian Families. Curr Issues Mol Biol 2021; 43:1778-1793. [PMID: 34889895 PMCID: PMC8929092 DOI: 10.3390/cimb43030125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple Sclerosis (MS) is a complex multifactorial autoimmune disease, whose sex- and age-adjusted prevalence in Sardinia (Italy) is among the highest worldwide. To date, 233 loci were associated with MS and almost 20% of risk heritability is attributable to common genetic variants, but many low-frequency and rare variants remain to be discovered. Here, we aimed to contribute to the understanding of the genetic basis of MS by investigating potentially functional rare variants. To this end, we analyzed thirteen multiplex Sardinian families with Immunochip genotyping data. For five families, Whole Exome Sequencing (WES) data were also available. Firstly, we performed a non-parametric Homozygosity Haplotype analysis for identifying the Region from Common Ancestor (RCA). Then, on these potential disease-linked RCA, we searched for the presence of rare variants shared by the affected individuals by analyzing WES data. We found: (i) a variant (43181034 T > G) in the splicing region on exon 27 of CUL9; (ii) a variant (50245517 A > C) in the splicing region on exon 16 of ATP9A; (iii) a non-synonymous variant (43223539 A > C), on exon 9 of TTBK1; (iv) a non-synonymous variant (42976917 A > C) on exon 9 of PPP2R5D; and v) a variant (109859349-109859354) in 3'UTR of MYO16.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Daria Marzanati
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Anna Laura Carotenuto
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Athena Hadjixenofontos
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Jacob L. McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Valeria Saddi
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, 08100 Nuoro, Italy; (V.S.); (M.P.)
| | - Marialuisa Piras
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, 08100 Nuoro, Italy; (V.S.); (M.P.)
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, 20095 Cusano Milanino, Italy
| |
Collapse
|
37
|
Barski MS, Minnell JJ, Maertens GN. PP2A Phosphatase as an Emerging Viral Host Factor. Front Cell Infect Microbiol 2021; 11:725615. [PMID: 34422684 PMCID: PMC8371333 DOI: 10.3389/fcimb.2021.725615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is one of the most ubiquitous cellular proteins and is responsible for the vast majority of Ser/Thr phosphatase activity in eukaryotes. PP2A is a heterotrimer, and its assembly, intracellular localization, enzymatic activity, and substrate specificity are subject to dynamic regulation. Each of its subunits can be targeted by viral proteins to hijack and modulate its activity and downstream signaling to the advantage of the virus. Binding to PP2A is known to be essential to the life cycle of many viruses and seems to play a particularly crucial role for oncogenic viruses, which utilize PP2A to transform infected cells through controlling the cell cycle and apoptosis. Here we summarise the latest developments in the field of PP2A viral targeting; in particular recent discoveries of PP2A hijacking through molecular mimicry of a B56-specific motif by several different viruses. We also discuss the potential as well as shortcomings for therapeutic intervention in the face of our current understanding of viral PP2A targeting.
Collapse
Affiliation(s)
| | | | - Goedele Noella Maertens
- Department of Infectious Disease, Section of Molecular Virology, St Mary’s Hospital, Imperial College London, London, United Kingdom
| |
Collapse
|
38
|
Protein Phosphatase 2A (PP2A) mutations in brain function, development, and neurologic disease. Biochem Soc Trans 2021; 49:1567-1588. [PMID: 34241636 DOI: 10.1042/bst20201313] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022]
Abstract
By removing Ser/Thr-specific phosphorylations in a multitude of protein substrates in diverse tissues, Protein Phosphatase type 2A (PP2A) enzymes play essential regulatory roles in cellular signalling and physiology, including in brain function and development. Here, we review current knowledge on PP2A gene mutations causally involved in neurodevelopmental disorders and intellectual disability, focusing on PPP2CA, PPP2R1A and PPP2R5D. We provide insights into the impact of these mutations on PP2A structure, substrate specificity and potential function in neurobiology and brain development.
Collapse
|
39
|
Sandal P, Jong CJ, Merrill RA, Song J, Strack S. Protein phosphatase 2A - structure, function and role in neurodevelopmental disorders. J Cell Sci 2021; 134:270819. [PMID: 34228795 DOI: 10.1242/jcs.248187] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neurodevelopmental disorders (NDDs), including intellectual disability (ID), autism and schizophrenia, have high socioeconomic impact, yet poorly understood etiologies. A recent surge of large-scale genome or exome sequencing studies has identified a multitude of mostly de novo mutations in subunits of the protein phosphatase 2A (PP2A) holoenzyme that are strongly associated with NDDs. PP2A is responsible for at least 50% of total Ser/Thr dephosphorylation in most cell types and is predominantly found as trimeric holoenzymes composed of catalytic (C), scaffolding (A) and variable regulatory (B) subunits. PP2A can exist in nearly 100 different subunit combinations in mammalian cells, dictating distinct localizations, substrates and regulatory mechanisms. PP2A is well established as a regulator of cell division, growth, and differentiation, and the roles of PP2A in cancer and various neurodegenerative disorders, such as Alzheimer's disease, have been reviewed in detail. This Review summarizes and discusses recent reports on NDDs associated with mutations of PP2A subunits and PP2A-associated proteins. We also discuss the potential impact of these mutations on the structure and function of the PP2A holoenzymes and the etiology of NDDs.
Collapse
Affiliation(s)
- Priyanka Sandal
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Chian Ju Jong
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Ronald A Merrill
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Jianing Song
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
40
|
Myöhänen TT, Mertens F, Norrbacka S, Cui H. Deletion or inhibition of prolyl oligopeptidase blocks lithium-induced phosphorylation of GSK3b and Akt by activation of protein phosphatase 2A. Basic Clin Pharmacol Toxicol 2021; 129:287-296. [PMID: 34196102 DOI: 10.1111/bcpt.13632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/15/2021] [Accepted: 06/27/2021] [Indexed: 11/27/2022]
Abstract
Alterations in prolyl oligopeptidase (PREP) activity have been connected, for example, with bipolar and major depressive disorder, and several studies have reported that lack or inhibition of PREP blocks the effects of lithium on inositol 1,4,5-triphosphate (IP3 ) levels. However, the impact of PREP modulation on other intracellular targets of lithium, such as glycogen synthase kinase 3 beta (GSK3b) or protein kinase B (Akt), has not been studied. We recently found that PREP regulates protein phosphatase 2A (PP2A), and because GSK3b and Akt are PP2A substrates, we studied if PREP-related lithium insensitivity is dependent on PP2A. To assess this, HEK-293 and SH-SY5Y cells with PREP deletion or PREP inhibition (KYP-2047) were exposed to lithium, and thereafter, the phosphorylation levels of GSK3b and Akt were measured by Western blot. As expected, PREP deletion and inhibition blocked the lithium-induced phosphorylation on GSK3b and Akt in both cell lines. When lithium exposure was combined with okadaic acid, a PP2A inhibitor, KYP-2047 did not have effect on lithium-induced GSK3b and Akt phosphorylation. Therefore, we conclude that PREP deletion or inhibition blocks the intracellular effects of lithium on GSK3b and Akt via PP2A activation.
Collapse
Affiliation(s)
- Timo T Myöhänen
- Division of Pharmacology and Pharmacotherapy/Drug Research Program, University of Helsinki, Helsinki, Finland.,Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, University of Turku, Turku, Finland.,School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Freke Mertens
- Division of Pharmacology and Pharmacotherapy/Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Susanna Norrbacka
- Division of Pharmacology and Pharmacotherapy/Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Hengjing Cui
- Division of Pharmacology and Pharmacotherapy/Drug Research Program, University of Helsinki, Helsinki, Finland.,Department of Pharmacy, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Yan L, Shen R, Cao Z, Han C, Zhang Y, Liu Y, Yang X, Xie M, Li H. A Novel Missense Variant in the Gene PPP2R5D Causes a Rare Neurodevelopmental Disorder with Increased Phenotype. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6661860. [PMID: 33628804 PMCID: PMC7895568 DOI: 10.1155/2021/6661860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 02/01/2021] [Indexed: 12/27/2022]
Abstract
PPP2R5D-related neurodevelopmental disorder, which is mainly caused by de novo missense variants in the PPP2R5D gene, is a rare autosomal dominant genetic disorder with about 100 patients and a total of thirteen pathogenic variants known to exist globally so far. Here, we present a 24-month-old Chinese boy with developmental delay and other common clinical characteristics of PPP2R5D-related neurodevelopmental disorder including hypotonia, macrocephaly, intellectual disability, speech impairment, and behavioral abnormality. Trio-whole exome sequencing (WES) and Sanger sequencing were performed to identify the causal gene variant. The pathogenicity of the variant was evaluated using bioinformatics tools. We identified a novel pathogenic variant in the PPP2R5D gene (c.620G>T, p.Trp207Leu). The variant is located in the variant hotspot region of this gene and is predicted to cause PPP2R5D protein dysfunction due to an increase in local hydrophobicity and unstable three-dimensional structure. We report a novel pathogenic variant of PPP2R5D associated with PPP2R5D-related neurodevelopmental disorder from a Chinese family. Our findings expanded the phenotypic and mutational spectrum of PPP2R5D-related neurodevelopmental disorder.
Collapse
Affiliation(s)
- Lulu Yan
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Ru Shen
- Department of Laboratory Medicine of Kunming Children's Hospital, Key Laboratory of Child Critical Disease Research of Yunnan Province, Kunming, Yunnan 650034, China
| | - Zongfu Cao
- National Centre for Human Genetic Resource, National Research Institute for Family Planning, Beijing 100081, China
| | - Chunxiao Han
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Yuxin Zhang
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Yingwen Liu
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Xiangchun Yang
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Min Xie
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, China
| |
Collapse
|
42
|
Papke CM, Smolen KA, Swingle MR, Cressey L, Heng RA, Toporsian M, Deng L, Hagen J, Shen Y, Chung WK, Kettenbach AN, Honkanen RE. A disorder-related variant (E420K) of a PP2A-regulatory subunit (PPP2R5D) causes constitutively active AKT-mTOR signaling and uncoordinated cell growth. J Biol Chem 2021; 296:100313. [PMID: 33482199 PMCID: PMC7952134 DOI: 10.1016/j.jbc.2021.100313] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/08/2023] Open
Abstract
Functional genomic approaches have facilitated the discovery of rare genetic disorders and improved efforts to decipher their underlying etiology. PPP2R5D-related disorder is an early childhood onset condition characterized by intellectual disability, hypotonia, autism-spectrum disorder, macrocephaly, and dysmorphic features. The disorder is caused by de novo single nucleotide changes in PPP2R5D, which generate heterozygous dominant missense variants. PPP2R5D is known to encode a B'-type (B'56δ) regulatory subunit of a PP2A-serine/threonine phosphatase. To help elucidate the molecular mechanisms altered in PPP2R5D-related disorder, we used a CRISPR-single-base editor to generate HEK-293 cells in which a single transition (c.1258G>A) was introduced into one allele, precisely recapitulating a clinically relevant E420K variant. Unbiased quantitative proteomic and phosphoproteomic analyses of endogenously expressed proteins revealed heterozygous-dominant changes in kinase/phosphatase signaling. These data combined with orthogonal validation studies revealed a previously unrecognized interaction of PPP2R5D with AKT in human cells, leading to constitutively active AKT-mTOR signaling, increased cell size, and uncoordinated cellular growth in E420K-variant cells. Rapamycin reduced cell size and dose-dependently reduced RPS6 phosphorylation in E420K-variant cells, suggesting that inhibition of mTOR1 can suppress both the observed RPS6 hyperphosphorylation and increased cell size. Together, our findings provide a deeper understanding of PPP2R5D and insight into how the E420K-variant alters signaling networks influenced by PPP2R5D. Our comprehensive approach, which combines precise genome editing, isobaric tandem mass tag labeling of peptides generated from endogenously expressed proteins, and concurrent liquid chromatography-mass spectrometry (LC-MS3), also provides a roadmap that can be used to rapidly explore the etiologies of additional genetic disorders.
Collapse
Affiliation(s)
- Cinta M Papke
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Kali A Smolen
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Mark R Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Lauren Cressey
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Richard A Heng
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Mourad Toporsian
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Liyong Deng
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Jacob Hagen
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA; Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA.
| | - Richard E Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
43
|
Ziats CA, Patterson WG, Friez M. Syndromic Autism Revisited: Review of the Literature and Lessons Learned. Pediatr Neurol 2021; 114:21-25. [PMID: 33189026 DOI: 10.1016/j.pediatrneurol.2020.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/01/2020] [Accepted: 06/19/2020] [Indexed: 11/29/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by deficits in communication, stereotyped behaviors, restricted interests, and impaired social skills. The severity of the neurobehavioral phenotype is variable and historically has been distinguished based on the presence or absence of additional symptoms, termed syndromic and nonsyndromic or idiopathic autism, respectively. However, although the advancement in genetic molecular technologies has brought an increased understanding of the pathophysiology of autism, most of this success has been in the diagnosis of syndromic disease, whereas the etiology of nonsyndromic autism remains less understood. Here we review the common and rare genetic syndromes that feature autism, specifically highlighting deletion and duplication syndromes, chromosomal anomalies, and monogenic disorders. We show that the study of syndromic autism provides insight into the phenotypic and molecular heterogeneity of neurodevelopmental disease and suggests how study of these disorders can be helpful in understanding disease mechanisms implicated in nonsyndromic autism.
Collapse
Affiliation(s)
- Catherine A Ziats
- Greenwood Genetic Center, J.C. Self Research Institute, Greenwood, South Carolina.
| | - Wesley G Patterson
- Greenwood Genetic Center, J.C. Self Research Institute, Greenwood, South Carolina
| | - Michael Friez
- Greenwood Genetic Center, J.C. Self Research Institute, Greenwood, South Carolina
| |
Collapse
|
44
|
Hetzelt KLML, Kerling F, Kraus C, Rauch C, Thiel CT, Winterholler M, Reis A, Zweier C. Early-onset parkinsonism in PPP2R5D-related neurodevelopmental disorder. Eur J Med Genet 2020; 64:104123. [PMID: 33338668 DOI: 10.1016/j.ejmg.2020.104123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/16/2020] [Accepted: 12/11/2020] [Indexed: 11/29/2022]
Abstract
PPP2R5D-related neurodevelopmental disorder (NDD) is a rare autosomal-dominant disease with developmental delay and mild to severe intellectual disability. So far, fewer than 30 affected individuals with mostly recurrent, de novo missense variants in PPP2R5D were reported. Recently, parkinsonism with an onset between 20 and 40 years was reported in four adult individuals with the same p.(Glu200Lys) variant in PPP2R5D. By trio exome sequencing we now identified the variant p.(Glu198Lys) in a 29 year old woman presenting with typical clinical manifestations of PPP2R5D-related neurodevelopmental disorder and additionally with motor decline and levodopa responsive, early-onset parkinsonism from her mid-twenties on. Accordingly, a clear reduction of dopamine transporter in the striatum on both sides was revealed by brain scintigraphy. Our findings further expand the molecular and clinical spectrum of PPP2R5D-related NDD and confirm the association with parkinsonism in early adulthood. This has marked implications for prognosis of PPP2R5D-related NDDs and for the therapeutic management of motor decline and parkinson-like symptoms in affected individuals.
Collapse
Affiliation(s)
- Katalin L M L Hetzelt
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, Erlangen, Germany
| | - Frank Kerling
- Department of Neurology, Epilepsy and Movement Disorders Center, Sana-Krankenhaus Rummelsberg, Schwarzenbruck/Nuremberg, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, Erlangen, Germany
| | - Christophe Rauch
- Department of Neurology, Epilepsy and Movement Disorders Center, Sana-Krankenhaus Rummelsberg, Schwarzenbruck/Nuremberg, Germany
| | - Christian T Thiel
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, Erlangen, Germany
| | - Martin Winterholler
- Department of Neurology, Epilepsy and Movement Disorders Center, Sana-Krankenhaus Rummelsberg, Schwarzenbruck/Nuremberg, Germany
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, Erlangen, Germany
| | - Christiane Zweier
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, Erlangen, Germany.
| |
Collapse
|
45
|
Lenaerts L, Reynhout S, Verbinnen I, Laumonnier F, Toutain A, Bonnet-Brilhault F, Hoorne Y, Joss S, Chassevent AK, Smith-Hicks C, Loeys B, Joset P, Steindl K, Rauch A, Mehta SG, Chung WK, Devriendt K, Holder SE, Jewett T, Baldwin LM, Wilson WG, Towner S, Srivastava S, Johnson HF, Daumer-Haas C, Baethmann M, Ruiz A, Gabau E, Jain V, Varghese V, Al-Beshri A, Fulton S, Wechsberg O, Orenstein N, Prescott K, Childs AM, Faivre L, Moutton S, Sullivan JA, Shashi V, Koudijs SM, Heijligers M, Kivuva E, McTague A, Male A, van Ierland Y, Plecko B, Maystadt I, Hamid R, Hannig VL, Houge G, Janssens V. The broad phenotypic spectrum of PPP2R1A-related neurodevelopmental disorders correlates with the degree of biochemical dysfunction. Genet Med 2020; 23:352-362. [PMID: 33106617 PMCID: PMC7862067 DOI: 10.1038/s41436-020-00981-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Neurodevelopmental disorders (NDD) caused by protein phosphatase 2A (PP2A) dysfunction have mainly been associated with de novo variants in PPP2R5D and PPP2CA, and more rarely in PPP2R1A. Here, we aimed to better understand the latter by characterizing 30 individuals with de novo and often recurrent variants in this PP2A scaffolding Aα subunit. Methods Most cases were identified through routine clinical diagnostics. Variants were biochemically characterized for phosphatase activity and interaction with other PP2A subunits. Results We describe 30 individuals with 16 different variants in PPP2R1A, 21 of whom had variants not previously reported. The severity of developmental delay ranged from mild learning problems to severe intellectual disability (ID) with or without epilepsy. Common features were language delay, hypotonia, and hypermobile joints. Macrocephaly was only seen in individuals without B55α subunit-binding deficit, and these patients had less severe ID and no seizures. Biochemically more disruptive variants with impaired B55α but increased striatin binding were associated with profound ID, epilepsy, corpus callosum hypoplasia, and sometimes microcephaly. Conclusion We significantly expand the phenotypic spectrum of PPP2R1A-related NDD, revealing a broader clinical presentation of the patients and that the functional consequences of the variants are more diverse than previously reported.
Collapse
Affiliation(s)
- Lisa Lenaerts
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Sara Reynhout
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Frédéric Laumonnier
- UMR1253, iBrain, University of Tours, INSERM, Tours, France.,Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France
| | - Annick Toutain
- UMR1253, iBrain, University of Tours, INSERM, Tours, France.,Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR1253, iBrain, University of Tours, INSERM, Tours, France.,Excellence Center in Autism and Neurodevelopmental Disorders, Centre Hospitalier Régional Universitaire, Tours, France
| | - Yana Hoorne
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Shelagh Joss
- West of Scotland Centre for Genomic Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | | | | | - Bart Loeys
- Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Schlieren, Zurich, Switzerland
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Schlieren, Zurich, Switzerland
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren, Zurich, Switzerland
| | - Sarju G Mehta
- East Anglian Regional Medical Genetics Service, Addenbrookes Hospital, Cambridge, UK
| | - Wendy K Chung
- Columbia University Medical Center, New York, NY, USA
| | - Koenraad Devriendt
- Department of Human Genetics, University of Leuven (KU Leuven), Leuven, Belgium
| | - Susan E Holder
- North West Thames Regional Genetics Service, Harrow, London, UK
| | - Tamison Jewett
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Lauren M Baldwin
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - William G Wilson
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Shelley Towner
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | | | - Hannah F Johnson
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | | | - Martina Baethmann
- Pediatric Neurology, Sozialpädiatrisches Zentrum, Klinikum Dritter Orden München, Munich, Germany
| | - Anna Ruiz
- Genetics Laboratory, UDIAT-Centre Diagnòstic, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Elisabeth Gabau
- Paediatric Unit, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Vani Jain
- All Wales Medical Genomics Service, University Hospital of Wales, Cardiff, UK
| | - Vinod Varghese
- All Wales Medical Genomics Service, University Hospital of Wales, Cardiff, UK
| | - Ali Al-Beshri
- Internal Medicine & Medical Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Oded Wechsberg
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Maccabi Healthcare Services, Tel Aviv, Israel
| | - Naama Orenstein
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Katrina Prescott
- Yorkshire Regional Genetics Department, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Anne-Marie Childs
- Department of Neurology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Laurence Faivre
- Centre de référence Anomalies du Développement et Syndromes malformatifs, FHU TRANSLAD, UMR1231 GAD, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Sébastien Moutton
- CPDPN, Pôle mère enfant, Maison de Santé Bordeaux Bagatelle, Talence, France
| | - Jennifer A Sullivan
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, USA
| | - Vandana Shashi
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, USA
| | | | - Malou Heijligers
- Department of Clinical Genetics, Maastricht UMC+, Maastricht, The Netherlands
| | - Emma Kivuva
- Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
| | - Amy McTague
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Alison Male
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital, London, UK
| | | | - Barbara Plecko
- Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Isabelle Maystadt
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Rizwan Hamid
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium. .,KU Leuven Brain Institute (LBI), Leuven, Belgium.
| |
Collapse
|
46
|
Christensen KR, Nairn AC. cAMP-regulated phosphoproteins DARPP-32, ARPP16/19, and RCS modulate striatal signal transduction through protein kinases and phosphatases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:39-65. [PMID: 33706938 DOI: 10.1016/bs.apha.2020.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Decades of research led by Paul Greengard identified protein phosphorylation as a ubiquitous and vital post-translational modification involved in many neuronal signaling pathways. In particular, his discovery that second messenger-regulated protein phosphorylation plays a central role in the propagation and transduction of signals in the nervous system has been essential in understanding the molecular mechanisms of neuronal communication. The establishment of dopamine (DA) as an essential neurotransmitter in the central nervous system, combined with observations that DA activates G-protein-coupled receptors to control the production of cyclic adenosine monophosphate (cAMP) in postsynaptic neurons, has provided fundamental insight into the regulation of neurotransmission. Notably, DA signaling in the striatum is involved in many neurological functions such as control of locomotion, reward, addiction, and learning, among others. This review focuses on the history, characterization, and function of cAMP-mediated regulation of serine/threonine protein phosphatases and their role in DA-mediated signaling in striatal neurons. Several small, heat- and acid-stable proteins, including DARPP-32, RCS, and ARPP-16/19, were discovered by the Greengard laboratory to be regulated by DA- and cAMP signaling, and found to undergo a complex but coordinated sequence of phosphorylation and dephosphorylation events. These studies have contributed significantly to the establishment of protein phosphorylation as a ubiquitous and vital process in signal propagation in neurons, paradigm shifting discoveries at the time. Understanding DA-mediated signaling in the context of signal propagation has led to numerous insights into human conditions and the development of treatments and therapies.
Collapse
Affiliation(s)
- Kyle R Christensen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States.
| |
Collapse
|
47
|
Kim CY, Wirth T, Hubsch C, Németh AH, Okur V, Anheim M, Drouot N, Tranchant C, Rudolf G, Chelly J, Tatton-Brown K, Blauwendraat C, Vonsattel JPG, Cortes E, Alcalay RN, Chung WK. Early-Onset Parkinsonism Is a Manifestation of the PPP2R5D p.E200K Mutation. Ann Neurol 2020; 88:1028-1033. [PMID: 32743835 DOI: 10.1002/ana.25863] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
PPP2R5D-related neurodevelopmental disorder is characterized by a range of neurodevelopmental and behavioral manifestations. We report the association of early-onset parkinsonism with the PPP2R5D p.E200K mutation. Clinical characterization and exome sequencing were performed on three patients, with postmortem neuropathologic examination for one patient. All patients had mild developmental delay and developed levodopa-responsive parkinsonism between the ages of 25 and 40 years. The PPP2R5D c.598G>A (p.E200K) mutation was identified in all patients. Neuropathologic examination demonstrated uneven, focally severe neuronal loss and gliosis in the substantia nigra pars compacta, without Lewy bodies. Our findings suggest the PPP2R5D p.E200K mutation to be a possible new cause of early-onset parkinsonism. ANN NEUROL 2020;88:1028-1033.
Collapse
Affiliation(s)
- Christine Y Kim
- Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA.,Center for Neuroepidemiology and Clinical Neurological Research, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Thomas Wirth
- Unit of Functional Neurosurgery, National Hospital for Neurology and Neurosurgery, University College London Hospital, London, UK.,Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Cécile Hubsch
- Fondation Ophtalmologique A. de Rothschild, Paris, France
| | - Andrea H Németh
- Oxford University Hospitals NHS Trust and University of Oxford, Oxford, UK
| | - Volkan Okur
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Mathieu Anheim
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Nathalie Drouot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Gabrielle Rudolf
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Jamel Chelly
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Laboratoire de diagnostic génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jean Paul G Vonsattel
- Department of Pathology and Cell Biology, Columbia University Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Etty Cortes
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roy N Alcalay
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
48
|
Genç Ö, An JY, Fetter RD, Kulik Y, Zunino G, Sanders SJ, Davis GW. Homeostatic plasticity fails at the intersection of autism-gene mutations and a novel class of common genetic modifiers. eLife 2020; 9:55775. [PMID: 32609087 PMCID: PMC7394548 DOI: 10.7554/elife.55775] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/07/2020] [Indexed: 01/08/2023] Open
Abstract
We identify a set of common phenotypic modifiers that interact with five independent autism gene orthologs (RIMS1, CHD8, CHD2, WDFY3, ASH1L) causing a common failure of presynaptic homeostatic plasticity (PHP) in Drosophila. Heterozygous null mutations in each autism gene are demonstrated to have normal baseline neurotransmission and PHP. However, PHP is sensitized and rendered prone to failure. A subsequent electrophysiology-based genetic screen identifies the first known heterozygous mutations that commonly genetically interact with multiple ASD gene orthologs, causing PHP to fail. Two phenotypic modifiers identified in the screen, PDPK1 and PPP2R5D, are characterized. Finally, transcriptomic, ultrastructural and electrophysiological analyses define one mechanism by which PHP fails; an unexpected, maladaptive up-regulation of CREG, a conserved, neuronally expressed, stress response gene and a novel repressor of PHP. Thus, we define a novel genetic landscape by which diverse, unrelated autism risk genes may converge to commonly affect the robustness of synaptic transmission.
Collapse
Affiliation(s)
- Özgür Genç
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Joon-Yong An
- Department of Psychiatry UCSF Weill Institute for Neurosciences University of California, San Francisco, San Francisco, United States.,School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Richard D Fetter
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Yelena Kulik
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Giulia Zunino
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Stephan J Sanders
- Department of Psychiatry UCSF Weill Institute for Neurosciences University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| |
Collapse
|
49
|
Jong CJ, Merrill RA, Wilkerson EM, Herring LE, Graves LM, Strack S. Reduction of protein phosphatase 2A (PP2A) complexity reveals cellular functions and dephosphorylation motifs of the PP2A/B'δ holoenzyme. J Biol Chem 2020; 295:5654-5668. [PMID: 32156701 PMCID: PMC7186168 DOI: 10.1074/jbc.ra119.011270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a large enzyme family responsible for most cellular Ser/Thr dephosphorylation events. PP2A substrate specificity, localization, and regulation by second messengers rely on more than a dozen regulatory subunits (including B/R2, B'/R5, and B″/R3), which form the PP2A heterotrimeric holoenzyme by associating with a dimer comprising scaffolding (A) and catalytic (C) subunits. Because of partial redundancy and high endogenous expression of PP2A holoenzymes, traditional approaches of overexpressing, knocking down, or knocking out PP2A regulatory subunits have yielded only limited insights into their biological roles and substrates. To this end, here we sought to reduce the complexity of cellular PP2A holoenzymes. We used tetracycline-inducible expression of pairs of scaffolding and regulatory subunits with complementary charge-reversal substitutions in their interaction interfaces. For each of the three regulatory subunit families, we engineered A/B charge-swap variants that could bind to one another, but not to endogenous A and B subunits. Because endogenous Aα was targeted by a co-induced shRNA, endogenous B subunits were rapidly degraded, resulting in expression of predominantly a single PP2A heterotrimer composed of the A/B charge-swap pair and the endogenous catalytic subunit. Using B'δ/PPP2R5D, we show that PP2A complexity reduction, but not PP2A overexpression, reveals a role of this holoenzyme in suppression of extracellular signal-regulated kinase signaling and protein kinase A substrate dephosphorylation. When combined with global phosphoproteomics, the PP2A/B'δ reduction approach identified consensus dephosphorylation motifs in its substrates and suggested that residues surrounding the phosphorylation site play roles in PP2A substrate specificity.
Collapse
Affiliation(s)
- Chian Ju Jong
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| | - Ronald A Merrill
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| | - Emily M Wilkerson
- Michael Hooker Proteomics Facility, University of North Carolina, Chapel Hill, North Carolina 27516
| | - Laura E Herring
- Michael Hooker Proteomics Facility, University of North Carolina, Chapel Hill, North Carolina 27516
| | - Lee M Graves
- Michael Hooker Proteomics Facility, University of North Carolina, Chapel Hill, North Carolina 27516
| | - Stefan Strack
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242.
| |
Collapse
|
50
|
Huang C, Liu T, Wang Q, Hou W, Zhou C, Song Z, Shi YS, Gao X, Chen G, Yin Z, Hu Y. Loss of PP2A Disrupts the Retention of Radial Glial Progenitors in the Telencephalic Niche to Impair the Generation for Late-Born Neurons During Cortical Development†. Cereb Cortex 2020; 30:4183-4196. [PMID: 32186707 DOI: 10.1093/cercor/bhaa042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Telencephalic radial glial progenitors (RGPs) are retained in the ventricular zone (VZ), the niche for neural stem cells during cortical development. However, the underlying mechanism is not well understood. To study whether protein phosphatase 2A (PP2A) may regulate the above process, we generate Ppp2cα conditional knockout (cKO) mice, in which PP2A catalytic subunit α (PP2Acα) is inactivated in neural progenitor cells in the dorsal telencephalon. We show that RGPs are ectopically distributed in cortical areas outside of the VZ in Ppp2cα cKO embryos. Whereas deletion of PP2Acα does not affect the proliferation of RGPs, it significantly impairs the generation of late-born neurons. We find complete loss of apical adherens junctions (AJs) in the ventricular membrane in Ppp2cα cKO cortices. We observe abundant colocalization for N-cadherin and PP2Acα in control AJs. Moreover, in vitro analysis reveals direct interactions of N-cadherin to PP2Acα and to β-catenin. Overall, this study not only uncovers a novel function of PP2Acα in retaining RGPs into the VZ but also demonstrates the impact of PP2A-dependent retention of RGPs on the generation for late-born neurons.
Collapse
Affiliation(s)
- Chaoli Huang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Tingting Liu
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Qihui Wang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Weikang Hou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Cuihua Zhou
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Zeyuan Song
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Zhenyu Yin
- Department of Geriatric, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Yimin Hu
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|