1
|
Nygren D, Moll U, Braun O, Karlsson U, Jönsson G. Cantú Syndrome With Acromegaloid Features, Multiple Endocrinopathies, and Infection Susceptibility. JCEM CASE REPORTS 2025; 3:luaf068. [PMID: 40236613 PMCID: PMC11997666 DOI: 10.1210/jcemcr/luaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Indexed: 04/17/2025]
Abstract
Cantú syndrome involves fetal polyhydramniosis, congenital hypertrichosis, and macrosomia. Distinctive features include acromegaloid features with broad nasal bridge and macroglossia as well as cardiac abnormalities, including patent ductus arteriosus. We present a case in a male patient, who presented with cardiac abnormalities in childhood, but was diagnosed with the syndrome in adulthood after many years of atypical symptoms such as multiple endocrinopathies and infection susceptibility. He had surgery for a patent ductus arteriosus in early childhood. During adulthood, he developed idiopathic pericarditis. Extensive rheumatological investigations were made, and in parallel, several endocrinopathies were found. These included thyroiditis with subsequent hypothyroidism, idiopathic partial hypocortisolism, and GH insufficiency. In addition, he had mild neutropenia and required hospitalization twice because of Streptococcus pyogenes infections. Immunodeficiency screening has not revealed a specific primary immunodeficiency, yet transient neutropenia, low count of CD8+ effector memory T cells, as well as lymphocyte responses, was seen during bacteremia. The diagnose was made after a trio-whole genome sequencing identified a pathogenic missense variant of the gene ABCC9 (c.3460C > T;p. (Arg1154Trp)) causing Cantú syndrome.
Collapse
Affiliation(s)
- David Nygren
- Division of Infection Medicine, Lund University, S-221 85 Lund, Sweden
- Department of Infectious Diseases, Skåne University Hospital, S221 85 Lund, Sweden
| | - Ulrika Moll
- Department of Endocrinology, Skåne University Hospital, S-221 85 Lund, Sweden
- Institution of Clinical Sciences, Lund University, S-223 62 Lund, Sweden
| | - Oscar Braun
- Institution of Clinical Sciences, Lund University, S-223 62 Lund, Sweden
- Department of Cardiology, Skåne University Hospital, S-221 85 Lund, Sweden
| | - Ulf Karlsson
- Department of Infectious Diseases, Skåne University Hospital, S221 85 Lund, Sweden
| | - Göran Jönsson
- Department of Infectious Diseases, Skåne University Hospital, S221 85 Lund, Sweden
| |
Collapse
|
2
|
Bowen RM, York NW, Padawer-Curry J, Bauer AQ, Lee JM, Nichols CG. Control of neurovascular coupling by ATP-sensitive potassium channels. J Cereb Blood Flow Metab 2025:271678X251313906. [PMID: 39819176 PMCID: PMC11748405 DOI: 10.1177/0271678x251313906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025]
Abstract
Regional blood flow within the brain is tightly coupled to regional neuronal activity, a process known as neurovascular coupling (NVC). In this study, we demonstrate the striking role of SUR2- and Kir6.1-dependent ATP-sensitive potassium (KATP) channels in control of NVC in the sensory cortex of conscious mice, in response to mechanical stimuli. We demonstrate that either globally increased (pinacidil-activated) or decreased (glibenclamide-inhibited) KATP activity markedly disrupts NVC; pinacidil-activation is capable of completely abolishing stimulus-evoked cortical hemodynamic responses, while glibenclamide slows and reduces the response. The response is similarly slowed and reduced in SUR2 KO animals, while animals expressing gain-of-function (GOF) mutations in Kir6.1, which underlie Cantú syndrome, exhibit baseline reduction of NVC as well as increased sensitivity to pinacidil. In revealing the dramatic effects of either increasing or decreasing SUR2/Kir6.1-dependent KATP activity on NVC, whether pharmacologically or genetically induced, the study has important implications both for monogenic KATP channel diseases and for more common brain pathologies.
Collapse
Affiliation(s)
- Ryan M Bowen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Nathaniel W York
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO, USA
- Center for the Investigation of Membrane Excitability and Diseases, Washington University in St. Louis, St. Louis, MO, USA
| | - Jonah Padawer-Curry
- Imaging Sciences PhD Program, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Adam Q Bauer
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO, USA
- Center for the Investigation of Membrane Excitability and Diseases, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
3
|
Efthymiou S, Scala M, Nagaraj V, Ochenkowska K, Komdeur FL, Liang RA, Abdel-Hamid MS, Sultan T, Barøy T, Van Ghelue M, Vona B, Maroofian R, Zafar F, Alkuraya FS, Zaki MS, Severino M, Duru KC, Tryon RC, Brauteset LV, Ansari M, Hamilton M, van Haelst MM, van Haaften G, Zara F, Houlden H, Samarut É, Nichols CG, Smeland MF, McClenaghan C. Novel loss-of-function variants expand ABCC9-related intellectual disability and myopathy syndrome. Brain 2024; 147:1822-1836. [PMID: 38217872 PMCID: PMC11068106 DOI: 10.1093/brain/awae010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/22/2023] [Accepted: 12/30/2023] [Indexed: 01/15/2024] Open
Abstract
Loss-of-function mutation of ABCC9, the gene encoding the SUR2 subunit of ATP sensitive-potassium (KATP) channels, was recently associated with autosomal recessive ABCC9-related intellectual disability and myopathy syndrome (AIMS). Here we identify nine additional subjects, from seven unrelated families, harbouring different homozygous loss-of-function variants in ABCC9 and presenting with a conserved range of clinical features. All variants are predicted to result in severe truncations or in-frame deletions within SUR2, leading to the generation of non-functional SUR2-dependent KATP channels. Affected individuals show psychomotor delay and intellectual disability of variable severity, microcephaly, corpus callosum and white matter abnormalities, seizures, spasticity, short stature, muscle fatigability and weakness. Heterozygous parents do not show any conserved clinical pathology but report multiple incidences of intra-uterine fetal death, which were also observed in an eighth family included in this study. In vivo studies of abcc9 loss-of-function in zebrafish revealed an exacerbated motor response to pentylenetetrazole, a pro-convulsive drug, consistent with impaired neurodevelopment associated with an increased seizure susceptibility. Our findings define an ABCC9 loss-of-function-related phenotype, expanding the genotypic and phenotypic spectrum of AIMS and reveal novel human pathologies arising from KATP channel dysfunction.
Collapse
Affiliation(s)
- Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marcello Scala
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16147 Genoa, Italy
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Vini Nagaraj
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| | - Katarzyna Ochenkowska
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), and Department of Neuroscience, Université de Montréal, Montreal H2X 0A9, Quebec, Canada
| | - Fenne L Komdeur
- Section Clinical Genetics, Department of Human Genetics and Amsterdam Reproduction and Development, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, The Netherlands
| | - Robin A Liang
- Department of Medical Genetics, Division of Child and Adolescent Health, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Tipu Sultan
- Department of Pediatric Neurology, Children Hospital, University of Child Health Sciences, Lahore, Punjab 54000, Pakistan
| | - Tuva Barøy
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Marijke Van Ghelue
- Department of Medical Genetics, Division of Child and Adolescent Health, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Barbara Vona
- Institute of Human Genetics and Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Faisal Zafar
- Department of Paediatric Neurology, Children’s Hospital and Institute of Child Health, Multan, Punjab 60000, Pakistan
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | | | - Kingsley C Duru
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| | - Robert C Tryon
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO 63110, USA
| | - Lin Vigdis Brauteset
- Division of Habilitation for Children, Innlandet Hospital Sanderud, Hamar 2312, Norway
| | - Morad Ansari
- South East Scotland Genetic Service, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Mark Hamilton
- West of Scotland Clinical Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Mieke M van Haelst
- Section Clinical Genetics, Department of Human Genetics and Amsterdam Reproduction and Development, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, The Netherlands
| | - Gijs van Haaften
- Department of Genetics, University Medical Center, Utrecht, 3584 CX, The Netherlands
| | - Federico Zara
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Éric Samarut
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), and Department of Neuroscience, Université de Montréal, Montreal H2X 0A9, Quebec, Canada
| | - Colin G Nichols
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO 63110, USA
| | - Marie F Smeland
- Department of Pediatric Rehabilitation, University Hospital of North Norway, 9019 Tromsø, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
| | - Conor McClenaghan
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| |
Collapse
|
4
|
Yang Y, Chen L. Functional dissection of KATP channel structures reveals the importance of a conserved interface. Structure 2024; 32:168-176.e2. [PMID: 38101402 DOI: 10.1016/j.str.2023.11.008] [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: 08/03/2023] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
ATP-sensitive potassium channels (KATP) are inhibited by ATP but activated by Mg-ADP, coupling the intracellular ATP/ADP ratio to the potassium conductance of the plasma membrane. Although there has been progress in determining the structure of KATP, the functional significance of the domain-domain interface in the gating properties of KATP channels remains incompletely understood. In this study, we define the structure of KATP as two modules: KATPcore and SURABC. Based on this model, we identified two functionally important interfaces between these two modules, namely interface I and interface II. Further structure-guided mutagenesis experiments indicate that destabilizing interface II by deleting ECL3 on the SUR1 subunit impairs KNtp-independent Mg-ADP activation, demonstrating the essential role of intramolecular interactions between KATPcore and SURABC in Mg-ADP activation. Additionally, interface II is functionally conserved between SUR1 and SUR2, and the hydrophobic residue F351 on ECL3 of SUR1 is crucial for maintaining the stability of this interface.
Collapse
Affiliation(s)
- Yaxiong Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; National Biomedical Imaging Center, Peking University, Beijing 100871, China.
| |
Collapse
|
5
|
Ding D, Hou T, Wei M, Wu JX, Chen L. The inhibition mechanism of the SUR2A-containing K ATP channel by a regulatory helix. Nat Commun 2023; 14:3608. [PMID: 37330603 PMCID: PMC10276813 DOI: 10.1038/s41467-023-39379-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 06/10/2023] [Indexed: 06/19/2023] Open
Abstract
KATP channels are metabolic sensors for intracellular ATP/ADP ratios, play essential roles in many physiological processes, and are implicated in a spectrum of pathological conditions. SUR2A-containing KATP channels differ from other subtypes in their sensitivity to Mg-ADP activation. However, the underlying structural mechanism remains poorly understood. Here we present a series of cryo-EM structures of SUR2A in the presence of different combinations of Mg-nucleotides and the allosteric inhibitor repaglinide. These structures uncover regulatory helix (R helix) on the NBD1-TMD2 linker, which wedges between NBD1 and NBD2. R helix stabilizes SUR2A in the NBD-separated conformation to inhibit channel activation. The competitive binding of Mg-ADP with Mg-ATP to NBD2 mobilizes the R helix to relieve such inhibition, allowing channel activation. The structures of SUR2B in similar conditions suggest that the C-terminal 42 residues of SUR2B enhance the structural dynamics of NBD2 and facilitate the dissociation of the R helix and the binding of Mg-ADP to NBD2, promoting NBD dimerization and subsequent channel activation.
Collapse
Affiliation(s)
- Dian Ding
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Tianyi Hou
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Miao Wei
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Jing-Xiang Wu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China.
| |
Collapse
|
6
|
Gisladottir RS, Helgason A, Halldorsson BV, Helgason H, Borsky M, Chien YR, Gudnason J, Gudjonsson SA, Moisik S, Dediu D, Thorleifsson G, Tragante V, Bustamante M, Jonsdottir GA, Stefansdottir L, Rutsdottir G, Magnusson SH, Hardarson M, Ferkingstad E, Halldorsson GH, Rognvaldsson S, Skuladottir A, Ivarsdottir EV, Norddahl G, Thorgeirsson G, Jonsdottir I, Ulfarsson MO, Holm H, Stefansson H, Thorsteinsdottir U, Gudbjartsson DF, Sulem P, Stefansson K. Sequence variants affecting voice pitch in humans. SCIENCE ADVANCES 2023; 9:eabq2969. [PMID: 37294764 PMCID: PMC10256171 DOI: 10.1126/sciadv.abq2969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/04/2023] [Indexed: 06/11/2023]
Abstract
The genetic basis of the human vocal system is largely unknown, as are the sequence variants that give rise to individual differences in voice and speech. Here, we couple data on diversity in the sequence of the genome with voice and vowel acoustics in speech recordings from 12,901 Icelanders. We show how voice pitch and vowel acoustics vary across the life span and correlate with anthropometric, physiological, and cognitive traits. We found that voice pitch and vowel acoustics have a heritable component and discovered correlated common variants in ABCC9 that associate with voice pitch. The ABCC9 variants also associate with adrenal gene expression and cardiovascular traits. By showing that voice and vowel acoustics are influenced by genetics, we have taken important steps toward understanding the genetics and evolution of the human vocal system.
Collapse
Affiliation(s)
- Rosa S. Gisladottir
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Department of Icelandic and Comparative Cultural Studies, University of Iceland, Saemundargata 2, 102 Reykjavik, Iceland
| | - Agnar Helgason
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Saemundargata 10, 102 Reykjavik, Iceland
| | - Bjarni V. Halldorsson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Department of Engineering, Reykjavik University, Menntavegur 1, 101 Reykjavik, Iceland
| | - Hannes Helgason
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Michal Borsky
- Department of Engineering, Reykjavik University, Menntavegur 1, 101 Reykjavik, Iceland
| | - Yu-Ren Chien
- Department of Engineering, Reykjavik University, Menntavegur 1, 101 Reykjavik, Iceland
| | - Jon Gudnason
- Department of Engineering, Reykjavik University, Menntavegur 1, 101 Reykjavik, Iceland
| | | | - Scott Moisik
- Division of Linguistics and Multilingual Studies, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Dan Dediu
- Department of Catalan Philology and General Linguistics, University of Barcelona, Gran Via 585, Barcelona 08007, Spain
- University of Barcelona Institute for Complex Systems (UBICS), Martí Franquès 1, Barcelona 08028, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain
| | | | | | | | | | | | | | | | | | - Egil Ferkingstad
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Gisli H. Halldorsson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Dunhagi 5, 107 Reykjavik, Iceland
| | | | | | | | | | - Gudmundur Thorgeirsson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Ingileif Jonsdottir
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Magnus O. Ulfarsson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Dunhagi 5, 107 Reykjavik, Iceland
| | - Hilma Holm
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Daniel F. Gudbjartsson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Dunhagi 5, 107 Reykjavik, Iceland
| | - Patrick Sulem
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics/Amgen Inc., Sturlugata 8, 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| |
Collapse
|
7
|
McClenaghan C, Mukadam MA, Roeglin J, Tryon RC, Grabner M, Dayal A, Meyer GA, Nichols CG. Skeletal muscle delimited myopathy and verapamil toxicity in SUR2 mutant mouse models of AIMS. EMBO Mol Med 2023; 15:e16883. [PMID: 37154692 PMCID: PMC10245035 DOI: 10.15252/emmm.202216883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
ABCC9-related intellectual disability and myopathy syndrome (AIMS) arises from loss-of-function (LoF) mutations in the ABCC9 gene, which encodes the SUR2 subunit of ATP-sensitive potassium (KATP ) channels. KATP channels are found throughout the cardiovascular system and skeletal muscle and couple cellular metabolism to excitability. AIMS individuals show fatigability, muscle spasms, and cardiac dysfunction. We found reduced exercise performance in mouse models of AIMS harboring premature stop codons in ABCC9. Given the roles of KATP channels in all muscles, we sought to determine how myopathy arises using tissue-selective suppression of KATP and found that LoF in skeletal muscle, specifically, underlies myopathy. In isolated muscle, SUR2 LoF results in abnormal generation of unstimulated forces, potentially explaining painful spasms in AIMS. We sought to determine whether excessive Ca2+ influx through CaV 1.1 channels was responsible for myopathology but found that the Ca2+ channel blocker verapamil unexpectedly resulted in premature death of AIMS mice and that rendering CaV 1.1 channels nonpermeable by mutation failed to reverse pathology; results which caution against the use of calcium channel blockers in AIMS.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Maya A Mukadam
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Jacob Roeglin
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Robert C Tryon
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Manfred Grabner
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Anamika Dayal
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Gretchen A Meyer
- Program in Physical Therapy, Departments of Orthopaedic Surgery, Neurology and Biomedical EngineeringWashington University School of MedicineSt. LouisMOUSA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
8
|
El-Meanawy SK, Dooge H, Wexler AC, Kosmach AC, Serban L, Santos EA, Alvarado FJ, Hacker TA, Ramratnam M. Overexpression of a Short Sulfonylurea Splice Variant Increases Cardiac Glucose Uptake and Uncouples Mitochondria by Regulating ROMK Activity. Life (Basel) 2023; 13:1015. [PMID: 37109544 PMCID: PMC10146620 DOI: 10.3390/life13041015] [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: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The mitochondrial splice variant of the sulfonylurea receptor (SUR2A-55) is associated with protection from myocardial ischemia-reperfusion (IR) injury, increased mitochondrial ATP sensitive K+ channel activity (mitoKATP) and altered glucose metabolism. While mitoKATP channels composed of CCDC51 and ABCB8 exist, the mitochondrial K+ pore regulated by SUR2A-55 is unknown. We explored whether SUR2A-55 regulates ROMK to form an alternate mitoKATP. We assessed glucose uptake in mice overexpressing SUR2A-55 (TGSUR2A-55) compared with WT mice during IR injury. We then examined the expression level of ROMK and the effect of ROMK modulation on mitochondrial membrane potential (Δψm) in WT and TGSUR2A-55 mice. TGSUR2A-55 had increased glucose uptake compared to WT mice during IR injury. The expression of ROMK was similar in WT compared to TGSUR2A-55 mice. ROMK inhibition hyperpolarized resting cardiomyocyte Δψm from TGSUR2A-55 mice but not from WT mice. In addition, TGSUR2A-55 and ROMK inhibitor treated WT isolated cardiomyocytes had enhanced mitochondrial uncoupling. ROMK inhibition blocked diazoxide induced Δψm depolarization and prevented preservation of Δψm from FCCP perfusion in WT and to a lesser degree TGSUR2A-55 mice. In conclusion, cardio-protection from SUR2A-55 is associated with ROMK regulation, enhanced mitochondrial uncoupling and increased glucose uptake.
Collapse
Affiliation(s)
- Sarah K. El-Meanawy
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Holly Dooge
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Allison C. Wexler
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Anna C. Kosmach
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Lara Serban
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Elizabeth A. Santos
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Francisco J. Alvarado
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Timothy A. Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| |
Collapse
|
9
|
Abstract
Ubiquitously expressed throughout the body, ATP-sensitive potassium (KATP) channels couple cellular metabolism to electrical activity in multiple tissues; their unique assembly as four Kir6 pore-forming subunits and four sulfonylurea receptor (SUR) subunits has resulted in a large armory of selective channel opener and inhibitor drugs. The spectrum of monogenic pathologies that result from gain- or loss-of-function mutations in these channels, and the potential for therapeutic correction of these pathologies, is now clear. However, while available drugs can be effective treatments for specific pathologies, cross-reactivity with the other Kir6 or SUR subfamily members can result in drug-induced versions of each pathology and may limit therapeutic usefulness. This review discusses the background to KATP channel physiology, pathology, and pharmacology and considers the potential for more specific or effective therapeutic agents.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
10
|
The interaction between polyphyllin I and SQLE protein induces hepatotoxicity through SREBP-2/HMGCR/SQLE/LSS pathway. J Pharm Anal 2023; 13:39-54. [PMID: 36820075 PMCID: PMC9937801 DOI: 10.1016/j.jpha.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/21/2022] Open
Abstract
Polyphyllin I (PPI) and polyphyllin II (PII) are the main active substances in the Paris polyphylla. However, liver toxicity of these compounds has impeded their clinical application and the potential hepatotoxicity mechanisms remain to be elucidated. In this work, we found that PPI and PII exposure could induce significant hepatotoxicity in human liver cell line L-02 and zebrafish in a dose-dependent manner. The results of the proteomic analysis in L-02 cells and transcriptome in zebrafish indicated that the hepatotoxicity of PPI and PII was associated with the cholesterol biosynthetic pathway disorders, which were alleviated by the cholesterol biosynthesis inhibitor lovastatin. Additionally, 3-hydroxy-3-methy-lglutaryl CoA reductase (HMGCR) and squalene epoxidase (SQLE), the two rate-limiting enzymes in the cholesterol synthesis, selected as the potential targets, were confirmed by the molecular docking, the overexpression, and knockdown of HMGCR or SQLE with siRNA. Finally, the pull-down and surface plasmon resonance technology revealed that PPI could directly bind with SQLE but not with HMGCR. Collectively, these data demonstrated that PPI-induced hepatotoxicity resulted from the direct binding with SQLE protein and impaired the sterol-regulatory element binding protein 2/HMGCR/SQLE/lanosterol synthase pathways, thus disturbing the cholesterol biosynthesis pathway. The findings of this research can contribute to a better understanding of the key role of SQLE as a potential target in drug-induced hepatotoxicity and provide a therapeutic strategy for the prevention of drug toxic effects with similar structures in the future.
Collapse
|
11
|
McClenaghan C, Nichols CG. Kir6.1 and SUR2B in Cantú syndrome. Am J Physiol Cell Physiol 2022; 323:C920-C935. [PMID: 35876283 PMCID: PMC9467476 DOI: 10.1152/ajpcell.00154.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
Collapse
Affiliation(s)
- Conor McClenaghan
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| |
Collapse
|
12
|
Wang Z, Bian W, Yan Y, Zhang DM. Functional Regulation of K ATP Channels and Mutant Insight Into Clinical Therapeutic Strategies in Cardiovascular Diseases. Front Pharmacol 2022; 13:868401. [PMID: 35837280 PMCID: PMC9274113 DOI: 10.3389/fphar.2022.868401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
ATP-sensitive potassium channels (KATP channels) play pivotal roles in excitable cells and link cellular metabolism with membrane excitability. The action potential converts electricity into dynamics by ion channel-mediated ion exchange to generate systole, involved in every heartbeat. Activation of the KATP channel repolarizes the membrane potential and decreases early afterdepolarization (EAD)-mediated arrhythmias. KATP channels in cardiomyocytes have less function under physiological conditions but they open during severe and prolonged anoxia due to a reduced ATP/ADP ratio, lessening cellular excitability and thus preventing action potential generation and cell contraction. Small active molecules activate and enhance the opening of the KATP channel, which induces the repolarization of the membrane and decreases the occurrence of malignant arrhythmia. Accumulated evidence indicates that mutation of KATP channels deteriorates the regulatory roles in mutation-related diseases. However, patients with mutations in KATP channels still have no efficient treatment. Hence, in this study, we describe the role of KATP channels and subunits in angiocardiopathy, summarize the mutations of the KATP channels and the functional regulation of small active molecules in KATP channels, elucidate the potential mechanisms of mutant KATP channels and provide insight into clinical therapeutic strategies.
Collapse
Affiliation(s)
- Zhicheng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weikang Bian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yufeng Yan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Structural insights into the mechanism of pancreatic K ATP channel regulation by nucleotides. Nat Commun 2022; 13:2770. [PMID: 35589716 PMCID: PMC9120461 DOI: 10.1038/s41467-022-30430-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
ATP-sensitive potassium channels (KATP) are metabolic sensors that convert the intracellular ATP/ADP ratio to the excitability of cells. They are involved in many physiological processes and implicated in several human diseases. Here we present the cryo-EM structures of the pancreatic KATP channel in both the closed state and the pre-open state, resolved in the same sample. We observe the binding of nucleotides at the inhibitory sites of the Kir6.2 channel in the closed but not in the pre-open state. Structural comparisons reveal the mechanism for ATP inhibition and Mg-ADP activation, two fundamental properties of KATP channels. Moreover, the structures also uncover the activation mechanism of diazoxide-type KATP openers. KATP channels are energy sensors. Here, authors report the Cryo-EM structures of pancreatic KATP in both the closed state and the pre-open state. These structures illuminate the mechanism of KATP channel regulation by the intracellular nucleotides.
Collapse
|
14
|
Ding D, Wu JX, Duan X, Ma S, Lai L, Chen L. Structural identification of vasodilator binding sites on the SUR2 subunit. Nat Commun 2022; 13:2675. [PMID: 35562524 PMCID: PMC9106677 DOI: 10.1038/s41467-022-30428-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/28/2022] [Indexed: 11/09/2022] Open
Abstract
ATP-sensitive potassium channels (KATP), composed of Kir6 and SUR subunits, convert the metabolic status of the cell into electrical signals. Pharmacological activation of SUR2- containing KATP channels by class of small molecule drugs known as KATP openers leads to hyperpolarization of excitable cells and to vasodilation. Thus, KATP openers could be used to treat cardiovascular diseases. However, where these vasodilators bind to KATP and how they activate the channel remains elusive. Here, we present cryo-EM structures of SUR2A and SUR2B subunits in complex with Mg-nucleotides and P1075 or levcromakalim, two chemically distinct KATP openers that are specific to SUR2. Both P1075 and levcromakalim bind to a common site in the transmembrane domain (TMD) of the SUR2 subunit, which is between TMD1 and TMD2 and is embraced by TM10, TM11, TM12, TM14, and TM17. These KATP openers synergize with Mg-nucleotides to stabilize SUR2 in the NBD-dimerized occluded state to activate the channel.
Collapse
Affiliation(s)
- Dian Ding
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China.,National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Jing-Xiang Wu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China.,National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Xinli Duan
- Beijing Jingtai Technology Co., Ltd., Beijing, China
| | - Songling Ma
- Beijing Jingtai Technology Co., Ltd., Beijing, China
| | - Lipeng Lai
- Beijing Jingtai Technology Co., Ltd., Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China. .,Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China. .,National Biomedical Imaging Center, Peking University, 100871, Beijing, China.
| |
Collapse
|
15
|
Ando K, Tong L, Peng D, Vázquez-Liébanas E, Chiyoda H, He L, Liu J, Kawakami K, Mochizuki N, Fukuhara S, Grutzendler J, Betsholtz C. KCNJ8/ABCC9-containing K-ATP channel modulates brain vascular smooth muscle development and neurovascular coupling. Dev Cell 2022; 57:1383-1399.e7. [PMID: 35588738 DOI: 10.1016/j.devcel.2022.04.019] [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: 02/24/2021] [Revised: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan.
| | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Hirohisa Chiyoda
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Jianping Liu
- Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden.
| |
Collapse
|
16
|
Zaytseva A, Tulintseva T, Fomicheva Y, Mikhailova V, Treshkur T, Kostareva A. Case Report: Loss-of-Function ABCC9 Genetic Variant Associated With Ventricular Fibrillation. Front Genet 2022; 13:718853. [PMID: 35495129 PMCID: PMC9044080 DOI: 10.3389/fgene.2022.718853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic variants in the ABCC9 gene, encoding the SUR2 auxiliary subunit from KATP channels, were previously linked with various inherited diseases. This wide range of congenital disorders includes multisystem and cardiovascular pathologies. The gain-of-function mutations result in Cantu syndrome, acromegaloid facial appearance, hypertrichosis, and acromegaloid facial features. The loss-of-function mutations in the ABCC9 gene were associated with the Brugada syndrome, early repolarization syndrome, and dilated cardiomyopathy. Here, we reported a patient with a loss-of-function variant in the ABCC9 gene, identified by target high-throughput sequencing. The female proband presented with several episodes of ventricular fibrillation and hypokalemia upon emotional stress. This case sheds light on the consequences of KATP channel dysfunction in the cardiovascular system and underlines the complexity of the clinical presentation of ABCC9-related diseases.
Collapse
Affiliation(s)
- Anastasia Zaytseva
- Almazov National Medical Research Centre, St Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St Petersburg, Russia
- *Correspondence: Anastasia Zaytseva,
| | | | - Yulya Fomicheva
- Almazov National Medical Research Centre, St Petersburg, Russia
| | | | | | - Anna Kostareva
- Almazov National Medical Research Centre, St Petersburg, Russia
- Department of Woman and Child Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
17
|
Gauthreaux KM, Teylan MA, Katsumata Y, Mock C, Culhane JE, Chen YC, Chan KCG, Fardo DW, Dugan AJ, Cykowski MD, Jicha GA, Kukull WA, Nelson PT. Limbic-Predominant Age-Related TDP-43 Encephalopathy: Medical and Pathologic Factors Associated With Comorbid Hippocampal Sclerosis. Neurology 2022; 98:e1422-e1433. [PMID: 35121671 PMCID: PMC8992604 DOI: 10.1212/wnl.0000000000200001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 01/03/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Limbic-predominant age-related Tar DNA binding protein 43 (TDP-43) encephalopathy neuropathologic change (LATE-NC) is present in ≈25% of older persons' brains and is strongly associated with cognitive impairment. Hippocampal sclerosis (HS) pathology is often comorbid with LATE-NC, but the clinical and pathologic correlates of HS in LATE-NC are not well understood. METHODS This retrospective autopsy cohort study used data derived from the National Alzheimer's Coordinating Center Neuropathology Data Set, which included neurologic status, medical histories, and neuropathologic results. All autopsies were performed in 2014 or later. Among participants with LATE-NC, those who also had HS pathology were compared with those without HS with regard to candidate risk factors or common underlying diseases. Statistical significance was set at nominal p < 0.05 in this exploratory study. RESULTS A total of 408 participants were included (n = 221 were LATE-NC+/HS-, n = 145 were LATE-NC+/HS+, and n = 42 were LATE-NC-/HS+). Most of the included LATE-NC+ participants were severely impaired cognitively (83.3% with dementia). Compared to HS- participants, LATE-NC+ participants with HS trended toward having worse cognitive status and scored lower on the Personal Care and Orientation domains (both p = 0.03). Among LATE-NC+ participants with Braak neurofibrillary tangle (NFT) stages 0 to IV (n = 88), HS+ participants were more impaired in the Memory and Orientation domains (both p = 0.02). There were no differences (HS+ compared with HS-) in the proportion with clinical histories of seizures, stroke, cardiac bypass procedures, diabetes, or hypertension. The HS+ group lacking TDP-43 proteinopathy (n = 42) was relatively likely to have had strokes (p = 0.03). When LATE-NC+ participants with or without HS were compared, there were no differences in Alzheimer disease neuropathologies (Thal β-amyloid phases or Braak NFT stages) or Lewy body pathologies. However, the HS+ group was less likely to have amygdala-restricted TDP-43 proteinopathy (LATE-NC stage 1) and more likely to have neocortical TDP-43 proteinopathy (LATE-NC stage 3) (p < 0.001). LATE-NC+ brains with HS also tended to have more severe circle of Willis atherosclerosis and arteriolosclerosis pathologies. DISCUSSION In this cohort skewed toward participants with severe dementia, LATE-NC+ HS pathology was not associated with seizures or with Alzheimer-type pathologies. Rather, the presence of comorbid HS pathology was associated with more widespread TDP-43 proteinopathy and with more severe non-β-amyloid vessel wall pathologies.
Collapse
Affiliation(s)
- Kathryn M Gauthreaux
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Merilee A Teylan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Yuriko Katsumata
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Charles Mock
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Jessica E Culhane
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Yen-Chi Chen
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Kwun C G Chan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - David W Fardo
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Adam J Dugan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Matthew D Cykowski
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Gregory A Jicha
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Walter A Kukull
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Peter T Nelson
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington.
| |
Collapse
|
18
|
Transcriptome-Guided Identification of Drugs for Repurposing to Treat Age-Related Hearing Loss. Biomolecules 2022; 12:biom12040498. [PMID: 35454087 PMCID: PMC9028743 DOI: 10.3390/biom12040498] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Age-related hearing loss (ARHL) or presbycusis is a prevalent condition associated with social isolation, cognitive impairment, and dementia. Age-related changes in the cochlea, the auditory portion of the inner ear, are the primary cause of ARHL. Unfortunately, there are currently no pharmaceutical approaches to treat ARHL. To examine the biological processes underlying age-related changes in the cochlea and identify candidate drugs for rapid repurposing to treat ARHL, we utilized bulk RNA sequencing to obtain transcriptomes from the functional substructures of the cochlea—the sensorineural structures, including the organ of Corti and spiral ganglion neurons (OC/SGN) and the stria vascularis and spiral ligament (SV/SL)—in young (6-week-old) and old (2-year-old) C57BL/6 mice. Transcriptomic analyses revealed both overlapping and unique patterns of gene expression and gene enrichment between substructures and with ageing. Based on these age-related transcriptional changes, we queried the protein products of genes differentially expressed with ageing in DrugBank and identified 27 FDA/EMA-approved drugs that are suitable to be repurposed to treat ARHL. These drugs target the protein products of genes that are differentially expressed with ageing uniquely in either the OC/SGN or SV/SL and that interrelate diverse biological processes. Further transcriptomic analyses revealed that most genes differentially expressed with ageing in both substructures encode protein products that are promising drug target candidates but are, nevertheless, not yet linked to approved drugs. Thus, with this study, we apply a novel approach to characterize the druggable genetic landscape for ARHL and propose a list of drugs to test in pre-clinical studies as potential treatment options for ARHL.
Collapse
|
19
|
|
20
|
Bauer B, Mally A, Liedtke D. Zebrafish Embryos and Larvae as Alternative Animal Models for Toxicity Testing. Int J Mol Sci 2021; 22:13417. [PMID: 34948215 PMCID: PMC8707050 DOI: 10.3390/ijms222413417] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Prerequisite to any biological laboratory assay employing living animals is consideration about its necessity, feasibility, ethics and the potential harm caused during an experiment. The imperative of these thoughts has led to the formulation of the 3R-principle, which today is a pivotal scientific standard of animal experimentation worldwide. The rising amount of laboratory investigations utilizing living animals throughout the last decades, either for regulatory concerns or for basic science, demands the development of alternative methods in accordance with 3R to help reduce experiments in mammals. This demand has resulted in investigation of additional vertebrate species displaying favourable biological properties. One prominent species among these is the zebrafish (Danio rerio), as these small laboratory ray-finned fish are well established in science today and feature outstanding biological characteristics. In this review, we highlight the advantages and general prerequisites of zebrafish embryos and larvae before free-feeding stages for toxicological testing, with a particular focus on cardio-, neuro, hepato- and nephrotoxicity. Furthermore, we discuss toxicokinetics, current advances in utilizing zebrafish for organ toxicity testing and highlight how advanced laboratory methods (such as automation, advanced imaging and genetic techniques) can refine future toxicological studies in this species.
Collapse
Affiliation(s)
- Benedikt Bauer
- Institute of Pharmacology and Toxicology, Julius-Maximilians-University, 97078 Würzburg, Germany; (B.B.); (A.M.)
| | - Angela Mally
- Institute of Pharmacology and Toxicology, Julius-Maximilians-University, 97078 Würzburg, Germany; (B.B.); (A.M.)
| | - Daniel Liedtke
- Institute of Human Genetics, Julius-Maximilians-University, 97074 Würzburg, Germany
| |
Collapse
|
21
|
Singareddy SS, Roessler HI, McClenaghan C, Ikle JM, Tryon RC, van Haaften G, Nichols CG. ATP-sensitive potassium channels in zebrafish cardiac and vascular smooth muscle. J Physiol 2021; 600:299-312. [PMID: 34820842 DOI: 10.1113/jp282157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
ATP-sensitive potassium channels (KATP channels) are hetero-octameric nucleotide-gated ion channels that couple cellular metabolism to excitability in various tissues. In the heart, KATP channels are activated during ischaemia and potentially during adrenergic stimulation. In the vasculature, they are normally active at a low level, reducing vascular tone, but the ubiquitous nature of these channels leads to complex and poorly understood channelopathies as a result of gain- or loss-of-function mutations. Zebrafish (ZF) models of these channelopathies may provide insights to the link between molecular dysfunction and complex pathophysiology, but this requires understanding the tissue dependence of channel activity and subunit specificity. Thus far, direct analysis of ZF KATP expression and functional properties has only been performed in pancreatic β-cells. Using a comprehensive combination of genetically modified fish, electrophysiology and gene expression analysis, we demonstrate that ZF cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. However, in contrast to mammalian cardiovascular KATP channels, ZF channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. The results provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome. KEY POINTS: Zebrafish cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. In contrast to mammalian cardiovascular KATP channels, zebrafish channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. We provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome.
Collapse
Affiliation(s)
- Soma S Singareddy
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Helen I Roessler
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Conor McClenaghan
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Jennifer M Ikle
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Robert C Tryon
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Gijs van Haaften
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Colin G Nichols
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
22
|
Dugan AJ, Nelson PT, Katsumata Y, Shade LMP, Boehme KL, Teylan MA, Cykowski MD, Mukherjee S, Kauwe JSK, Hohman TJ, Schneider JA, Fardo DW. Analysis of genes (TMEM106B, GRN, ABCC9, KCNMB2, and APOE) implicated in risk for LATE-NC and hippocampal sclerosis provides pathogenetic insights: a retrospective genetic association study. Acta Neuropathol Commun 2021; 9:152. [PMID: 34526147 PMCID: PMC8442328 DOI: 10.1186/s40478-021-01250-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is the most prevalent subtype of TDP-43 proteinopathy, affecting up to 1/3rd of aged persons. LATE-NC often co-occurs with hippocampal sclerosis (HS) pathology. It is currently unknown why some individuals with LATE-NC develop HS while others do not, but genetics may play a role. Previous studies found associations between LATE-NC phenotypes and specific genes: TMEM106B, GRN, ABCC9, KCNMB2, and APOE. Data from research participants with genomic and autopsy measures from the National Alzheimer’s Coordinating Center (NACC; n = 631 subjects included) and the Religious Orders Study and Memory and the Rush Aging Project (ROSMAP; n = 780 included) were analyzed in the current study. Our goals were to reevaluate disease-associated genetic variants using newly collected data and to query whether the specific genotype/phenotype associations could provide new insights into disease-driving pathways. Research subjects included in prior LATE/HS genome-wide association studies (GWAS) were excluded. Single nucleotide variants (SNVs) within 10 kb of TMEM106B, GRN, ABCC9, KCNMB2, and APOE were tested for association with HS and LATE-NC, and separately for Alzheimer’s pathologies, i.e. amyloid plaques and neurofibrillary tangles. Significantly associated SNVs were identified. When results were meta-analyzed, TMEM106B, GRN, and APOE had significant gene-based associations with both LATE and HS, whereas ABCC9 had significant associations with HS only. In a sensitivity analysis limited to LATE-NC + cases, ABCC9 variants were again associated with HS. By contrast, the associations of TMEM106B, GRN, and APOE with HS were attenuated when adjusting for TDP-43 proteinopathy, indicating that these genes may be associated primarily with TDP-43 proteinopathy. None of these genes except APOE appeared to be associated with Alzheimer’s-type pathology. In summary, using data not included in prior studies of LATE or HS genomics, we replicated several previously reported gene-based associations and found novel evidence that specific risk alleles can differentially affect LATE-NC and HS.
Collapse
|
23
|
Maggi L, Bonanno S, Altamura C, Desaphy JF. Ion Channel Gene Mutations Causing Skeletal Muscle Disorders: Pathomechanisms and Opportunities for Therapy. Cells 2021; 10:cells10061521. [PMID: 34208776 PMCID: PMC8234207 DOI: 10.3390/cells10061521] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle ion channelopathies (SMICs) are a large heterogeneous group of rare genetic disorders caused by mutations in genes encoding ion channel subunits in the skeletal muscle mainly characterized by myotonia or periodic paralysis, potentially resulting in long-term disabilities. However, with the development of new molecular technologies, new genes and new phenotypes, including progressive myopathies, have been recently discovered, markedly increasing the complexity in the field. In this regard, new advances in SMICs show a less conventional role of ion channels in muscle cell division, proliferation, differentiation, and survival. Hence, SMICs represent an expanding and exciting field. Here, we review current knowledge of SMICs, with a description of their clinical phenotypes, cellular and molecular pathomechanisms, and available treatments.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
- Correspondence:
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (C.A.); (J.-F.D.)
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (C.A.); (J.-F.D.)
| |
Collapse
|
24
|
Roessler HI, van der Heuvel LM, Shields K, Guilliams KP, Knoers NVAM, van Haaften G, Grange DK, van Haelst MM. Behavioral and cognitive functioning in individuals with Cantú syndrome. Am J Med Genet A 2021; 185:2434-2444. [PMID: 34056838 DOI: 10.1002/ajmg.a.62348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 11/07/2022]
Abstract
Cantú syndrome (CS) is caused by pathogenic variants in ABCC9 and KCNJ8 encoding the regulatory and pore-forming subunits of ATP-sensitive potassium (KATP ) channels. CS is characterized by congenital hypertrichosis, distinctive facial features, peripheral edema, and cardiac and neurodevelopmental abnormalities. Behavioral and cognitive issues have been self-reported by some CS individuals, but results of formal standardized investigations have not been published. To assess the cognitive profile, social functioning, and psychiatric symptoms in a large group of CS subjects systematically in a cross-sectional manner, we invited 35 individuals (1-69 years) with confirmed ABCC9 variants and their relatives to complete various commonly applied standardized age-related questionnaires, including the Kaufman brief intelligence test 2, the social responsiveness scale-2, and the Achenbach system of empirically based assessment. The majority of CS individuals demonstrated average verbal and nonverbal intelligence compared to the general population. Fifteen percent of cases showed social functioning strongly associated with a clinical diagnosis of autism spectrum disorder. Both externalizing and internalizing problems were also present in this cohort. In particular, anxiety, anxiety or attention deficit hyperactivity disorder, and autism spectrum behaviors were predominantly observed in the younger subjects in the cohort (≥25%), but this percentage decreased markedly in adults.
Collapse
Affiliation(s)
- Helen I Roessler
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lieke M van der Heuvel
- Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kathleen Shields
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kristin P Guilliams
- Division of Pediatric Neurology, Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nine V A M Knoers
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Gijs van Haaften
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Dorothy K Grange
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for the Investigation of Membrane Excitability Diseases (CIMED), St. Louis, Missouri, USA
| | - Mieke M van Haelst
- Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Clinical Genetics, VU Medical Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Zhang H, Hanson A, de Almeida TS, Emfinger C, McClenaghan C, Harter T, Yan Z, Cooper PE, Brown GS, Arakel EC, Mecham RP, Kovacs A, Halabi CM, Schwappach B, Remedi MS, Nichols CG. Complex consequences of Cantu syndrome SUR2 variant R1154Q in genetically modified mice. JCI Insight 2021; 6:145934. [PMID: 33529173 PMCID: PMC8021106 DOI: 10.1172/jci.insight.145934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/27/2021] [Indexed: 01/10/2023] Open
Abstract
Cantu syndrome (CS) is caused by gain-of-function (GOF) mutations in pore-forming (Kir6.1, KCNJ8) and accessory (SUR2, ABCC9) ATP-sensitive potassium (KATP) channel subunits, the most common mutations being SUR2[R1154Q] and SUR2[R1154W], carried by approximately 30% of patients. We used CRISPR/Cas9 genome engineering to introduce the equivalent of the human SUR2[R1154Q] mutation into the mouse ABCC9 gene. Along with minimal CS disease features, R1154Q cardiomyocytes and vascular smooth muscle showed much lower KATP current density and pinacidil activation than WT cells. Almost complete loss of SUR2-dependent protein and KATP in homozygous R1154Q ventricles revealed underlying diazoxide-sensitive SUR1-dependent KATP channel activity. Surprisingly, sequencing of SUR2 cDNA revealed 2 distinct transcripts, one encoding full-length SUR2 protein; and the other with an in-frame deletion of 93 bases (corresponding to 31 amino acids encoded by exon 28) that was present in approximately 40% and approximately 90% of transcripts from hetero- and homozygous R1154Q tissues, respectively. Recombinant expression of SUR2A protein lacking exon 28 resulted in nonfunctional channels. CS tissue from SUR2[R1154Q] mice and human induced pluripotent stem cell-derived (hiPSC-derived) cardiomyocytes showed only full-length SUR2 transcripts, although further studies will be required in order to fully test whether SUR2[R1154Q] or other CS mutations might result in aberrant splicing and variable expressivity of disease features in human CS.
Collapse
Affiliation(s)
- Haixia Zhang
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Alex Hanson
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Tobias Scherf de Almeida
- Department of Molecular Biology, Center for Biochemistry and Molecular Cell Biology, Heart Research Center Göttingen, University Medicine Göttingen, Göttingen, Germany
| | - Christopher Emfinger
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Theresa Harter
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Zihan Yan
- Center for the Investigation of Membrane Excitability Diseases and.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research
| | - Paige E Cooper
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - G Schuyler Brown
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Eric C Arakel
- Department of Molecular Biology, Center for Biochemistry and Molecular Cell Biology, Heart Research Center Göttingen, University Medicine Göttingen, Göttingen, Germany
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | | | - Carmen M Halabi
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Blanche Schwappach
- Department of Molecular Biology, Center for Biochemistry and Molecular Cell Biology, Heart Research Center Göttingen, University Medicine Göttingen, Göttingen, Germany
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases and.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and.,Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Maqoud F, Scala R, Hoxha M, Zappacosta B, Tricarico D. ATP-sensitive potassium channel subunits in the neuroinflammation: novel drug targets in neurodegenerative disorders. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:130-149. [PMID: 33463481 DOI: 10.2174/1871527320666210119095626] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/07/2020] [Accepted: 08/28/2020] [Indexed: 11/22/2022]
Abstract
Arachidonic acids and its metabolites modulate plenty of ligand-gated, voltage-dependent ion channels, and metabolically regulated potassium channels including ATP-sensitive potassium channels (KATP). KATP channels are hetero-multimeric complexes of sulfonylureas receptors (SUR1, SUR2A or SUR2B) and the pore-forming subunits (Kir6.1 and Kir6.2) likewise expressed in the pre-post synapsis of neurons and inflammatory cells, thereby affecting their proliferation and activity. KATP channels are involved in amyloid-β (Aβ)-induced pathology, therefore emerging as therapeutic targets against Alzheimer's and related diseases. The modulation of these channels can represent an innovative strategy for the treatment of neurodegenerative disorders; nevertheless, the currently available drugs are not selective for brain KATP channels and show contrasting effects. This phenomenon can be a consequence of the multiple physiological roles of the different varieties of KATP channels. Openings of cardiac and muscular KATP channel subunits, is protective against caspase-dependent atrophy in these tissues and some neurodegenerative disorders, whereas in some neuroinflammatory diseases benefits can be obtained through the inhibition of neuronal KATP channel subunits. For example, glibenclamide exerts an anti-inflammatory effect in respiratory, digestive, urological, and central nervous system (CNS) diseases, as well as in ischemia-reperfusion injury associated with abnormal SUR1-Trpm4/TNF-α or SUR1-Trpm4/ Nos2/ROS signaling. Despite this strategy is promising, glibenclamide may have limited clinical efficacy due to its unselective blocking action of SUR2A/B subunits also expressed in cardiovascular apparatus with pro-arrhythmic effects and SUR1 expressed in pancreatic beta cells with hypoglycemic risk. Alternatively, neuronal selective dual modulators showing agonist/antagonist actions on KATP channels can be an option.
Collapse
Affiliation(s)
- Fatima Maqoud
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| | - Rosa Scala
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| | - Malvina Hoxha
- Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, "Catholic University Our Lady of Good Counsel", Tirana. Albania
| | - Bruno Zappacosta
- Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, "Catholic University Our Lady of Good Counsel", Tirana. Albania
| | - Domenico Tricarico
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| |
Collapse
|
27
|
Sieliwonczyk E, Matchkov VV, Vandendriessche B, Alaerts M, Bakkers J, Loeys B, Schepers D. Inherited Ventricular Arrhythmia in Zebrafish: Genetic Models and Phenotyping Tools. Rev Physiol Biochem Pharmacol 2021; 184:33-68. [PMID: 34533615 DOI: 10.1007/112_2021_65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In the last years, the field of inheritable ventricular arrhythmia disease modelling has changed significantly with a push towards the use of novel cellular cardiomyocyte based models. However, there is a growing need for new in vivo models to study the disease pathology at the tissue and organ level. Zebrafish provide an excellent opportunity for in vivo modelling of inheritable ventricular arrhythmia syndromes due to the remarkable similarity between their cardiac electrophysiology and that of humans. Additionally, many state-of-the-art methods in gene editing and electrophysiological phenotyping are available for zebrafish research. In this review, we give a comprehensive overview of the published zebrafish genetic models for primary electrical disorders and arrhythmogenic cardiomyopathy. We summarise and discuss the strengths and weaknesses of the different technical approaches for the generation of genetically modified zebrafish disease models, as well as the electrophysiological approaches in zebrafish phenotyping. By providing this detailed overview, we aim to draw attention to the potential of the zebrafish model for studying arrhythmia syndromes at the organ level and as a platform for personalised medicine and drug testing.
Collapse
Affiliation(s)
- Ewa Sieliwonczyk
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| | - Vladimir V Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Bert Vandendriessche
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Maaike Alaerts
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Jeroen Bakkers
- Hubrecht Institute for Developmental and Stem Cell Biology, Utrecht, The Netherlands
| | - Bart Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Dorien Schepers
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.,Laboratory for Molecular, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
28
|
Scala R, Maqoud F, Zizzo N, Mele A, Camerino GM, Zito FA, Ranieri G, McClenaghan C, Harter TM, Nichols CG, Tricarico D. Pathophysiological Consequences of KATP Channel Overactivity and Pharmacological Response to Glibenclamide in Skeletal Muscle of a Murine Model of Cantù Syndrome. Front Pharmacol 2020; 11:604885. [PMID: 33329006 PMCID: PMC7734337 DOI: 10.3389/fphar.2020.604885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cantù syndrome (CS) arises from mutations in ABCC9 and KCNJ8 genes that lead to gain of function (GOF) of ATP-sensitive potassium (KATP) channels containing SUR2A and Kir6.1 subunits, respectively, of KATP channels. Pathological consequences of CS have been reported for cardiac and smooth muscle cells but consequences in skeletal muscle are unknown. Children with CS show muscle hypotonia and adult manifest fatigability. We analyzed muscle properties of Kir6.1[V65M] CS mice, by measurements of forelimb strength and ultrasonography of hind-limb muscles, as well as assessing KATP channel properties in native Flexor digitorum brevis (FDB) and Soleus (SOL) fibers by the patch-clamp technique in parallel with histopathological, immunohistochemical and Polymerase Chain Reaction (PCR) analysis. Forelimb strength was lower in Kir6.1wt/VM mice than in WT mice. Also, a significant enhancement of echodensity was observed in hind-limb muscles of Kir6.1wt/VM mice relative to WT, suggesting the presence of fibrous tissue. There was a higher KATP channel current amplitude in Kir6.1wt/VM FDB fibers relative to WT and a reduced response to glibenclamide. The IC50 of glibenclamide to block KATP channels in FDB fibers was 1.3 ± 0.2 × 10−7 M in WT and 1.2 ± 0.1 × 10−6 M in Kir6.1wt/VM mice, respectively; and it was 1.2 ± 0.4 × 10−7 M in SOL WT fibers but not measurable in Kir6.1wt/VM fibers. The sensitivity of the KATP channel to MgATP was not modified in Kir6.1wt/VM fibers. Histopathological/immunohistochemical analysis of SOL revealed degeneration plus regressive-necrotic lesions with regeneration, and up-regulation of Atrogin-1, MuRF1, and BNIP3 mRNA/proteins in Kir6.1wt/VM mice. Kir6.1wt/VM mutation in skeletal muscle leads to changes of the KATP channel response to glibenclamide in FDB and SOL fibers, and it is associated with histopathological and gene expression changes in slow-twitch muscle, suggesting marked atrophy and autophagy.
Collapse
Affiliation(s)
- Rosa Scala
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Fatima Maqoud
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Nicola Zizzo
- Section of Veterinary Pathology and Comparative Oncology, Department of Veterinary Medicine, University of Bari "Aldo Moro", Valenzano, Italy
| | - Antonietta Mele
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Giulia Maria Camerino
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Alfredo Zito
- Interventional and Medical Oncology Unit, Department of Pathology National Cancer Research Centre, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Girolamo Ranieri
- Interventional and Medical Oncology Unit, Department of Pathology National Cancer Research Centre, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Conor McClenaghan
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Theresa M Harter
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Colin G Nichols
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Domenico Tricarico
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
29
|
Kortüm F, Niceta M, Magliozzi M, Dumic Kubat K, Robertson SP, Moresco A, Dentici ML, Baban A, Leoni C, Onesimo R, Obregon MG, Digilio MC, Zampino G, Novelli A, Tartaglia M, Kutsche K. Cantú syndrome versus Zimmermann-Laband syndrome: Report of nine individuals with ABCC9 variants. Eur J Med Genet 2020; 63:103996. [PMID: 32622958 DOI: 10.1016/j.ejmg.2020.103996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/23/2020] [Accepted: 06/28/2020] [Indexed: 11/17/2022]
Abstract
Cantú syndrome (CS) is a rare developmental disorder characterized by a coarse facial appearance, macrocephaly, hypertrichosis, skeletal and cardiovascular anomalies and caused by heterozygous gain-of-function variants in ABCC9 and KCNJ8, encoding subunits of heterooctameric ATP-sensitive potassium (KATP) channels. CS shows considerable clinical overlap with Zimmermann-Laband syndrome (ZLS), a rare condition with coarse facial features, hypertrichosis, gingival overgrowth, intellectual disability of variable degree, and hypoplasia or aplasia of terminal phalanges and/or nails. ZLS is caused by heterozygous gain-of-function variants in KCNH1 or KCNN3, and gain-of-function KCNK4 variants underlie the clinically similar FHEIG (facial dysmorphism, hypertrichosis, epilepsy, intellectual disability/developmental delay, and gingival overgrowth) syndrome; KCNH1, KCNN3 and KCNK4 encode potassium channels. Within our research project on ZLS, we performed targeted Sanger sequencing of ABCC9 in 15 individuals tested negative for a mutation in the ZLS-associated genes and found two individuals harboring a heterozygous pathogenic ABCC9 missense variant. Through a collaborative effort, we identified a total of nine individuals carrying a monoallelic ABCC9 variant: five sporadic patients and four members of two unrelated families. Among the six detected ABCC9 missense variants, four [p.(Pro252Leu), p.(Thr259Lys), p.(Ala1064Pro), and p.(Arg1197His)] were novel. Systematic assessment of the clinical features in the nine cases with an ABCC9 variant highlights the significant clinical overlap between ZLS and CS that includes early developmental delay, hypertrichosis, gingival overgrowth, joint laxity, and hypoplasia of terminal phalanges and nails. Gain of K+ channel activity possibly accounts for significant clinical similarities of CS, ZLS and FHEIG syndrome and defines a new subgroup of potassium channelopathies.
Collapse
Affiliation(s)
- Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcello Niceta
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Monia Magliozzi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Angelica Moresco
- Paediatric Hospital Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Maria Lisa Dentici
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Anwar Baban
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart - ERN GUARD-Heart, Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children Hospital and Research Institute, Rome, Italy
| | - Chiara Leoni
- Center of Rare Diseases and Congenital Defects, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Roberta Onesimo
- Center of Rare Diseases and Congenital Defects, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | | | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Center of Rare Diseases and Congenital Defects, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Antonio Novelli
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
30
|
McClenaghan C, Huang Y, Matkovich SJ, Kovacs A, Weinheimer CJ, Perez R, Broekelmann TJ, Harter TM, Lee JM, Remedi MS, Nichols CG. The Mechanism of High-Output Cardiac Hypertrophy Arising From Potassium Channel Gain-of-Function in Cantú Syndrome. FUNCTION 2020; 1:zqaa004. [PMID: 32865539 PMCID: PMC7446247 DOI: 10.1093/function/zqaa004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 01/06/2023] Open
Abstract
Dramatic cardiomegaly arising from gain-of-function (GoF) mutations in the ATP-sensitive potassium (KATP) channels genes, ABCC9 and KCNJ8, is a characteristic feature of Cantú syndrome (CS). How potassium channel over-activity results in cardiac hypertrophy, as well as the long-term consequences of cardiovascular remodeling in CS, is unknown. Using genome-edited mouse models of CS, we therefore sought to dissect the pathophysiological mechanisms linking KATP channel GoF to cardiac remodeling. We demonstrate that chronic reduction of systemic vascular resistance in CS is accompanied by elevated renin-angiotensin signaling, which drives cardiac enlargement and blood volume expansion. Cardiac enlargement in CS results in elevation of basal cardiac output, which is preserved in aging. However, the cardiac remodeling includes altered gene expression patterns that are associated with pathological hypertrophy and are accompanied by decreased exercise tolerance, suggestive of reduced cardiac reserve. Our results identify a high-output cardiac hypertrophy phenotype in CS which is etiologically and mechanistically distinct from other myocardial hypertrophies, and which exhibits key features of high-output heart failure (HOHF). We propose that CS is a genetically-defined HOHF disorder and that decreased vascular smooth muscle excitability is a novel mechanism for HOHF pathogenesis.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yan Huang
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Scot J Matkovich
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla J Weinheimer
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ron Perez
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas J Broekelmann
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theresa M Harter
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin-Moo Lee
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
31
|
Davis MJ, Kim HJ, Zawieja SD, Castorena-Gonzalez JA, Gui P, Li M, Saunders BT, Zinselmeyer BH, Randolph GJ, Remedi MS, Nichols CG. Kir6.1-dependent K ATP channels in lymphatic smooth muscle and vessel dysfunction in mice with Kir6.1 gain-of-function. J Physiol 2020; 598:3107-3127. [PMID: 32372450 DOI: 10.1113/jp279612] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Spontaneous contractions are essential for normal lymph transport and these contractions are exquisitely sensitive to the KATP channel activator pinacidil. KATP channel Kir6.1 and SUR2B subunits are expressed in mouse lymphatic smooth muscle (LSM) and form functional KATP channels as verified by electrophysiological techniques. Global deletion of Kir6.1 or SUR2 subunits results in severely impaired lymphatic contractile responses to pinacidil. Smooth muscle-specific expression of Kir6.1 gain-of-function mutant (GoF) subunits results in profound lymphatic contractile dysfunction and LSM hyperpolarization that is partially rescued by the KATP inhibitor glibenclamide. In contrast, lymphatic endothelial-specific expression of Kir6.1 GoF has essentially no effect on lymphatic contractile function. The high sensitivity of LSM to KATP channel GoF offers an explanation for the lymphoedema observed in patients with Cantú syndrome, a disorder caused by gain-of-function mutations in genes encoding Kir6.1 or SUR2, and suggests that glibenclamide may be an appropriate therapeutic agent. ABSTRACT This study aimed to understand the functional expression of KATP channel subunits in distinct lymphatic cell types, and assess the consequences of altered KATP channel activity on lymphatic pump function. KATP channel subunits Kir6.1 and SUR2B were expressed in mouse lymphatic muscle by PCR, but only Kir6.1 was expressed in lymphatic endothelium. Spontaneous contractions of popliteal lymphatics from wild-type (WT) (C57BL/6J) mice, assessed by pressure myography, were very sensitive to inhibition by the SUR2-specific KATP channel activator pinacidil, which hyperpolarized both mouse and human lymphatic smooth muscle (LSM). In vessels from mice with deletion of Kir6.1 (Kir6.1-/- ) or SUR2 (SUR2[STOP]) subunits, contractile parameters were not significantly different from those of WT vessels, suggesting that basal KATP channel activity in LSM is not an essential component of the lymphatic pacemaker, and does not exert a strong influence over contractile strength. However, these vessels were >100-fold less sensitive than WT vessels to pinacidil. Smooth muscle-specific expression of a Kir6.1 gain-of-function (GoF) subunit resulted in severely impaired lymphatic contractions and hyperpolarized LSM. Membrane potential and contractile activity was partially restored by the KATP channel inhibitor glibenclamide. In contrast, lymphatic endothelium-specific expression of Kir6.1 GoF subunits had negligible effects on lymphatic contraction frequency or amplitude. Our results demonstrate a high sensitivity of lymphatic contractility to KATP channel activators through activation of Kir6.1/SUR2-dependent channels in LSM. In addition, they offer an explanation for the lymphoedema observed in patients with Cantú syndrome, a disorder caused by gain-of-function mutations in genes encoding Kir6.1/SUR2.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Hae Jin Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Scott D Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Jorge A Castorena-Gonzalez
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Peichun Gui
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Min Li
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Brian T Saunders
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Bernd H Zinselmeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Maria S Remedi
- Department of Medicine, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| |
Collapse
|
32
|
Martin GM, Sung MW, Shyng SL. Pharmacological chaperones of ATP-sensitive potassium channels: Mechanistic insight from cryoEM structures. Mol Cell Endocrinol 2020; 502:110667. [PMID: 31821855 PMCID: PMC6994177 DOI: 10.1016/j.mce.2019.110667] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
ATP-sensitive potassium (KATP) channels are uniquely evolved protein complexes that couple cell energy levels to cell excitability. They govern a wide range of physiological processes including hormone secretion, neuronal transmission, vascular dilation, and cardiac and neuronal preconditioning against ischemic injuries. In pancreatic β-cells, KATP channels composed of Kir6.2 and SUR1, encoded by KCNJ11 and ABCC8, respectively, play a key role in coupling blood glucose concentration to insulin secretion. Mutations in ABCC8 or KCNJ11 that diminish channel function result in congenital hyperinsulinism. Many of these mutations principally hamper channel biogenesis and hence trafficking to the cell surface. Several small molecules have been shown to correct channel biogenesis and trafficking defects. Here, we review studies aimed at understanding how mutations impair channel biogenesis and trafficking and how pharmacological ligands overcome channel trafficking defects, particularly highlighting recent cryo-EM structural studies which have shed light on the mechanisms of channel assembly and pharmacological chaperones.
Collapse
Affiliation(s)
- Gregory M Martin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Min Woo Sung
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|