1
|
Torres M, Pederson B, Wang H, Lin LL, Wang HH, Bugarin-Lapuz A, Zhao Z, Qi L. Purkinje cell-specific deficiency in SEL1L-hrd1 endoplasmic reticulum-associated degradation causes progressive cerebellar ataxia in mice. JCI Insight 2024; 9:e174725. [PMID: 39352758 DOI: 10.1172/jci.insight.174725] [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: 08/10/2023] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Recent studies have identified multiple genetic variants of SEL1L-HRD1 endoplasmic reticulum-associated degradation (ERAD) in humans with neurodevelopmental disorders and locomotor dysfunctions, including ataxia. However, the relevance and importance of SEL1L-HRD1 ERAD in the pathogenesis of ataxia remain unexplored. Here, we showed that SEL1L deficiency in Purkinje cells leads to early-onset progressive cerebellar ataxia with progressive loss of Purkinje cells with age. Mice with Purkinje cell-specific deletion of SEL1L (Sel1LPcp2Cre) exhibited motor dysfunction beginning around 9 weeks of age. Transmission electron microscopy analysis revealed dilated ER and fragmented nuclei in Purkinje cells of adult Sel1LPcp2Cre mice, indicative of altered ER homeostasis and cell death. Finally, loss of Purkinje cells was associated with a secondary neurodegeneration of granular cells, as well as robust activation of astrocytes and proliferation of microglia, in the cerebellums of Sel1LPcp2Cre mice. These data demonstrate the pathophysiological importance of SEL1L-HRD1 ERAD in Purkinje cells in the pathogenesis of cerebellar ataxia.
Collapse
Affiliation(s)
- Mauricio Torres
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Brent Pederson
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hui Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Huilun Helen Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Amara Bugarin-Lapuz
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Dematteis G, Tapella L, Casali C, Talmon M, Tonelli E, Reano S, Ariotti A, Pessolano E, Malecka J, Chrostek G, Kulkovienė G, Umbrasas D, Distasi C, Grilli M, Ladds G, Filigheddu N, Fresu LG, Mikoshiba K, Matute C, Ramos-Gonzalez P, Jekabsone A, Calì T, Brini M, Biggiogera M, Cavaliere F, Miggiano R, Genazzani AA, Lim D. ER-mitochondria distance is a critical parameter for efficient mitochondrial Ca 2+ uptake and oxidative metabolism. Commun Biol 2024; 7:1294. [PMID: 39390051 PMCID: PMC11467464 DOI: 10.1038/s42003-024-06933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
IP3 receptor (IP3R)-mediated Ca2+ transfer at the mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) drives mitochondrial Ca2+ uptake and oxidative metabolism and is linked to different pathologies, including Parkinson's disease (PD). The dependence of Ca2+ transfer efficiency on the ER-mitochondria distance remains unexplored. Employing molecular rulers that stabilize ER-mitochondrial distances at 5 nm resolution, and using genetically encoded Ca2+ indicators targeting the ER lumen and the sub-mitochondrial compartments, we now show that a distance of ~20 nm is optimal for Ca2+ transfer and mitochondrial oxidative metabolism due to enrichment of IP3R at MERCS. In human iPSC-derived astrocytes from PD patients, 20 nm MERCS were specifically reduced, which correlated with a reduction of mitochondrial Ca2+ uptake. Stabilization of the ER-mitochondrial interaction at 20 nm, but not at 10 nm, fully rescued mitochondrial Ca2+ uptake in PD astrocytes. Our work determines with precision the optimal distance for Ca2+ flux between ER and mitochondria and suggests a new paradigm for fine control over mitochondrial function.
Collapse
Affiliation(s)
- Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Claudio Casali
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani"; University of Pavia, Pavia, Italy
| | - Maria Talmon
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Elisa Tonelli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Simone Reano
- Interdipartimental Center for Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale, Novara, Italy
| | - Adele Ariotti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Emanuela Pessolano
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Justyna Malecka
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gabriela Chrostek
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gabrielė Kulkovienė
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Danielius Umbrasas
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Carla Distasi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Nicoletta Filigheddu
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luigia Grazia Fresu
- Department of Health Sciences, School of Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Katsuhiko Mikoshiba
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech Univeristy, Shanghai, China
| | - Carlos Matute
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Madrid, Spain
| | - Paula Ramos-Gonzalez
- CIBERNED, Madrid, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU); Achucarro Basque Center for Neuroscience, The Basque Biomodels Platform for Human Research (BBioH) at Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Aiste Jekabsone
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tito Calì
- Study Center for Neurodegeneration (CESNE), Department of Biomedical Sciences (DSB), Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Pharmaceutical and Pharmacological Sciences (DSF), Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani"; University of Pavia, Pavia, Italy
| | - Fabio Cavaliere
- CIBERNED, Madrid, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU); Achucarro Basque Center for Neuroscience; The Basque Biomodels Platform for Human Research (BBioH) at Achucarro Basque Center for Neuroscience, Fundación Biofisica Bizkaia, Leioa, Spain
| | - Riccardo Miggiano
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
3
|
Chesneau B, Calvas P, Cassagne M, Varenne F, Rozet JM, Bonneville F, Chassaing N, Fournié P, Fares-Taie L, Plaisancié J. ITPR1: The missing gene in miosis-ataxia syndrome? Am J Med Genet A 2024; 194:e63655. [PMID: 38711238 DOI: 10.1002/ajmg.a.63655] [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: 01/24/2024] [Revised: 03/23/2024] [Accepted: 04/26/2024] [Indexed: 05/08/2024]
Abstract
The association of early-onset non-progressive ataxia and miosis is an extremely rare phenotypic entity occasionally reported in the literature. To date, only one family (two siblings and their mother) has benefited from a genetic diagnosis by the identification of a missense heterozygous variant (p.Arg36Cys) in the ITPR1 gene. This gene encodes the inositol 1,4,5-trisphosphate receptor type 1, an intracellular channel that mediates calcium release from the endoplasmic reticulum. Deleterious variants in this gene are known to be associated with two types of spinocerebellar ataxia, SCA15 and SCA29, and with Gillespie syndrome that is associated with ataxia, partial iris hypoplasia, and intellectual disability. In this work, we describe a novel individual carrying a heterozygous missense variant (p.Arg36Pro) at the same position in the N-terminal suppressor domain of ITPR1 as the family previously reported, with the same phenotype associating early-onset non-progressive ataxia and miosis. This second report confirms the implication of ITPR1 in the miosis-ataxia syndrome and therefore broadens the clinical spectrum of the gene. Moreover, the high specificity of the phenotype makes it a recognizable syndrome of genetic origin.
Collapse
Affiliation(s)
- Bertrand Chesneau
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, site constitutif, CHU Toulouse, Toulouse, France
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Patrick Calvas
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, site constitutif, CHU Toulouse, Toulouse, France
| | | | - Fanny Varenne
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, site constitutif, CHU Toulouse, Toulouse, France
- Service d'Ophtalmologie, Hôpital Purpan, Toulouse, France
| | - Jean-Michel Rozet
- Laboratoire de Génétique Ophtalmologique, INSERM U1163, Institut Imagine, Paris, France
| | - Fabrice Bonneville
- Département de Neuroradiologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Nicolas Chassaing
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, site constitutif, CHU Toulouse, Toulouse, France
| | - Pierre Fournié
- Service d'Ophtalmologie, Hôpital Purpan, Toulouse, France
| | - Lucas Fares-Taie
- Laboratoire de Génétique Ophtalmologique, INSERM U1163, Institut Imagine, Paris, France
| | - Julie Plaisancié
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, site constitutif, CHU Toulouse, Toulouse, France
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
4
|
In Lee J, Choi JY, Yang S. Discovery of a de novo ITPR1 missense mutation in a patient with early-onset cerebellar ataxia: A rare case report of spinocerebellar ataxia 29. Mol Genet Genomic Med 2024; 12:e2466. [PMID: 38860480 PMCID: PMC11165338 DOI: 10.1002/mgg3.2466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Spinocerebellar ataxia 29 (SCA29) is a rare genetic disorder characterized by early-onset ataxia, gross motor delay, and infantile hypotonia, and is primarily associated with variants in the ITPR1 gene. Cases of SCA29 in Asia are rarely reported, limiting our understanding of this disease. METHODS A female Korean infant, demonstrating clinical features of SCA29, underwent evaluation and rehabilitation at our outpatient clinic from the age of 3 months to the current age of 4 years. Trio-based genome sequencing tests were performed on the patient and her biological parents. RESULTS The infant initially presented with macrocephaly, hypotonia, and nystagmus, with nonspecific findings on initial neuroimaging. Subsequent follow-up revealed gross motor delay, early onset ataxia, strabismus, and cognitive impairment. Further neuroimaging revealed atrophy of the cerebellum and vermis, and genetic analysis revealed a de novo pathogenic heterozygous c.800C>T, p.Thr267Met missense mutation in the ITPR1 gene (NM_001378452.1). CONCLUSION This is the first reported case of SCA29 in a Korean patient, expanding the genetic and phenotypic spectrum of ITPR1-related ataxias. Our case highlights the importance of recognizing early-onset ataxic symptoms, central hypotonia, and gross motor delays with poor ocular fixation, cognitive deficits, and isolated cerebellar atrophy as crucial clinical indicators of SCA29.
Collapse
Affiliation(s)
- Jae In Lee
- Department of Rehabilitation Medicine, College of MedicineChungnam National UniversityDaejeonRepublic of Korea
- Daejeon Chungcheong Regional Medical Rehabilitation CenterChungnam National University HospitalDaejeonRepublic of Korea
| | - Ja Young Choi
- Department of Rehabilitation Medicine, College of MedicineChungnam National UniversityDaejeonRepublic of Korea
- Daejeon Chungcheong Regional Medical Rehabilitation CenterChungnam National University HospitalDaejeonRepublic of Korea
| | - Shin‐Seung Yang
- Department of Rehabilitation Medicine, College of MedicineChungnam National UniversityDaejeonRepublic of Korea
- Daejeon Chungcheong Regional Medical Rehabilitation CenterChungnam National University HospitalDaejeonRepublic of Korea
| |
Collapse
|
5
|
Kochkina EN, Kopylova EE, Rogachevskaja OA, Kovalenko NP, Kabanova NV, Kotova PD, Bystrova MF, Kolesnikov SS. Agonist-Induced Ca 2+ Signaling in HEK-293-Derived Cells Expressing a Single IP 3 Receptor Isoform. Cells 2024; 13:562. [PMID: 38607001 PMCID: PMC11011116 DOI: 10.3390/cells13070562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
In mammals, three genes encode IP3 receptors (IP3Rs), which are involved in agonist-induced Ca2+ signaling in cells of apparently all types. Using the CRISPR/Cas9 approach for disruption of two out of three IP3R genes in HEK-293 cells, we generated three monoclonal cell lines, IP3R1-HEK, IP3R2-HEK, and IP3R3-HEK, with the single functional isoform, IP3R1, IP3R2, and IP3R3, respectively. All engineered cells responded to ACh with Ca2+ transients in an "all-or-nothing" manner, suggesting that each IP3R isotype was capable of mediating CICR. The sensitivity of cells to ACh strongly correlated with the affinity of IP3 binding to an IP3R isoform they expressed. Based on a mathematical model of intracellular Ca2+ signals induced by thapsigargin, a SERCA inhibitor, we developed an approach for estimating relative Ca2+ permeability of Ca2+ store and showed that all three IP3R isoforms contributed to Ca2+ leakage from ER. The relative Ca2+ permeabilities of Ca2+ stores in IP3R1-HEK, IP3R2-HEK, and IP3R3-HEK cells were evaluated as 1:1.75:0.45. Using the genetically encoded sensor R-CEPIA1er for monitoring Ca2+ signals in ER, engineered cells were ranged by resting levels of stored Ca2+ as IP3R3-HEK ≥ IP3R1-HEK > IP3R2-HEK. The developed cell lines could be helpful for further assaying activity, regulation, and pharmacology of individual IP3R isoforms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stanislav S. Kolesnikov
- Institute of Cell Biophysics, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 3 Institutskaya Street, 142290 Pushchino, Russia
| |
Collapse
|
6
|
Perry JL, Scribano FJ, Gebert JT, Engevik KA, Ellis JM, Hyser JM. Host IP 3R channels are dispensable for rotavirus Ca 2+ signaling but critical for intercellular Ca 2+ waves that prime uninfected cells for rapid virus spread. mBio 2024; 15:e0214523. [PMID: 38112482 PMCID: PMC10790754 DOI: 10.1128/mbio.02145-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023] Open
Abstract
IMPORTANCE Many viruses exploit host Ca2+ signaling to facilitate their replication; however, little is known about how Ca2+ signals from different host and viral channels contribute to the overall dysregulation of Ca2+ signaling or promote virus replication. Using cells lacking IP3R, a host ER Ca2+ channel, we delineated intracellular Ca2+ signals within virus-infected cells and intercellular Ca2+ waves (ICWs), which increased Ca2+ signaling in neighboring, uninfected cells. In infected cells, IP3R was dispensable for rotavirus-induced Ca2+ signaling and replication, suggesting the rotavirus NSP4 viroporin supplies these signals. However, IP3R-mediated ICWs increase rotavirus replication kinetics and spread, indicating that the Ca2+ signals from the ICWs may prime nearby uninfected cells to better support virus replication upon eventual infection. This "pre-emptive priming" of uninfected cells by exploiting host intercellular pathways in the vicinity of virus-infected cells represents a novel mechanism for viral reprogramming of the host to gain a replication advantage.
Collapse
Affiliation(s)
- Jacob L. Perry
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Francesca J. Scribano
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - John T. Gebert
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen A. Engevik
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jenna M. Ellis
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph M. Hyser
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
Tolonen JP, Parolin Schnekenberg R, McGowan S, Sims D, McEntagart M, Elmslie F, Shears D, Stewart H, Tofaris GK, Dabir T, Morrison PJ, Johnson D, Hadjivassiliou M, Ellard S, Shaw‐Smith C, Znaczko A, Dixit A, Suri M, Sarkar A, Harrison RE, Jones G, Houlden H, Ceravolo G, Jarvis J, Williams J, Shanks ME, Clouston P, Rankin J, Blumkin L, Lerman‐Sagie T, Ponger P, Raskin S, Granath K, Uusimaa J, Conti H, McCann E, Joss S, Blakes AJ, Metcalfe K, Kingston H, Bertoli M, Kneen R, Lynch SA, Martínez Albaladejo I, Moore AP, Jones WD, Becker EB, Németh AH. Detailed Analysis of ITPR1 Missense Variants Guides Diagnostics and Therapeutic Design. Mov Disord 2024; 39:141-151. [PMID: 37964426 PMCID: PMC10952845 DOI: 10.1002/mds.29651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND The ITPR1 gene encodes the inositol 1,4,5-trisphosphate (IP3 ) receptor type 1 (IP3 R1), a critical player in cerebellar intracellular calcium signaling. Pathogenic missense variants in ITPR1 cause congenital spinocerebellar ataxia type 29 (SCA29), Gillespie syndrome (GLSP), and severe pontine/cerebellar hypoplasia. The pathophysiological basis of the different phenotypes is poorly understood. OBJECTIVES We aimed to identify novel SCA29 and GLSP cases to define core phenotypes, describe the spectrum of missense variation across ITPR1, standardize the ITPR1 variant nomenclature, and investigate disease progression in relation to cerebellar atrophy. METHODS Cases were identified using next-generation sequencing through the Deciphering Developmental Disorders study, the 100,000 Genomes project, and clinical collaborations. ITPR1 alternative splicing in the human cerebellum was investigated by quantitative polymerase chain reaction. RESULTS We report the largest, multinational case series of 46 patients with 28 unique ITPR1 missense variants. Variants clustered in functional domains of the protein, especially in the N-terminal IP3 -binding domain, the carbonic anhydrase 8 (CA8)-binding region, and the C-terminal transmembrane channel domain. Variants outside these domains were of questionable clinical significance. Standardized transcript annotation, based on our ITPR1 transcript expression data, greatly facilitated analysis. Genotype-phenotype associations were highly variable. Importantly, while cerebellar atrophy was common, cerebellar volume loss did not correlate with symptom progression. CONCLUSIONS This dataset represents the largest cohort of patients with ITPR1 missense variants, expanding the clinical spectrum of SCA29 and GLSP. Standardized transcript annotation is essential for future reporting. Our findings will aid in diagnostic interpretation in the clinic and guide selection of variants for preclinical studies. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jussi Pekka Tolonen
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute of Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Ricardo Parolin Schnekenberg
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Oxford Center for Genomic MedicineOxford University Hospitals National Health Service Foundation Trust, University of OxfordOxfordUK
| | - Simon McGowan
- Centre for Computational Biology, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - David Sims
- Centre for Computational Biology, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Meriel McEntagart
- South West Regional Genetics ServiceSt. George's University HospitalsLondonUK
| | - Frances Elmslie
- South West Regional Genetics ServiceSt. George's University HospitalsLondonUK
| | - Debbie Shears
- Oxford Center for Genomic MedicineOxford University Hospitals National Health Service Foundation Trust, University of OxfordOxfordUK
| | - Helen Stewart
- Oxford Center for Genomic MedicineOxford University Hospitals National Health Service Foundation Trust, University of OxfordOxfordUK
| | - George K. Tofaris
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute of Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Tabib Dabir
- Northern Ireland Regional Genetics ServiceBelfast City HospitalBelfastUK
| | - Patrick J. Morrison
- Patrick G. Johnston Centre for Cancer Research and Cell BiologyQueen's University BelfastBelfastUK
| | - Diana Johnson
- Sheffield Clinical Genetics ServiceSheffield Children's NHS Foundation TrustSheffieldUK
| | - Marios Hadjivassiliou
- Department of NeurologyRoyal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation TrustSheffieldUK
| | - Sian Ellard
- Exeter Genomics LaboratoryRoyal Devon University Healthcare NHS Foundation TrustUK
| | - Charles Shaw‐Smith
- Peninsula Clinical Genetics Service, Royal Devon University HospitalRoyal Devon University Healthcare NHS Foundation TrustExeterUK
| | - Anna Znaczko
- Peninsula Clinical Genetics Service, Royal Devon University HospitalRoyal Devon University Healthcare NHS Foundation TrustExeterUK
| | - Abhijit Dixit
- Department of Clinical GeneticsNottingham University Hospitals NHS TrustNottinghamUK
| | - Mohnish Suri
- Department of Clinical GeneticsNottingham University Hospitals NHS TrustNottinghamUK
| | - Ajoy Sarkar
- Department of Clinical GeneticsNottingham University Hospitals NHS TrustNottinghamUK
| | - Rachel E. Harrison
- Department of Clinical GeneticsNottingham University Hospitals NHS TrustNottinghamUK
| | - Gabriela Jones
- Department of Clinical GeneticsNottingham University Hospitals NHS TrustNottinghamUK
| | - Henry Houlden
- Department of Neuromuscular DisordersUCL Queen Square Institute of Neurology, University College LondonLondonUK
| | - Giorgia Ceravolo
- Department of Neuromuscular DisordersUCL Queen Square Institute of Neurology, University College LondonLondonUK
- Unit of Pediatric Emergency, Department of Adult and Childhood Human PathologyUniversity Hospital of MessinaMessinaItaly
| | - Joanna Jarvis
- Birmingham Women's and Children's NHS Foundation TrustBirminghamUK
| | - Jonathan Williams
- Oxford Regional Genetics Laboratory, Churchill HospitalOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Morag E. Shanks
- Oxford Regional Genetics Laboratory, Churchill HospitalOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Penny Clouston
- Oxford Regional Genetics Laboratory, Churchill HospitalOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Julia Rankin
- Department of Clinical GeneticsRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - Lubov Blumkin
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Pediatric Movement Disorders Service, Pediatric Neurology UnitEdith Wolfson Medical CenterHolonIsrael
| | - Tally Lerman‐Sagie
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Magen Center for Rare Diseases‐Metabolic, NeurogeneticWolfson Medical CenterHolonIsrael
| | - Penina Ponger
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Movement Disorders Unit, Department of NeurologyTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Salmo Raskin
- Genetika Centro de Aconselhamento e LaboratórioCuritibaBrazil
| | - Katariina Granath
- Research Unit of Clinical MedicineMedical Research Center, Oulu University Hospital and University of OuluOuluFinland
| | - Johanna Uusimaa
- Research Unit of Clinical MedicineMedical Research Center, Oulu University Hospital and University of OuluOuluFinland
| | - Hector Conti
- All Wales Medical Genomics ServiceWrexham Maelor HospitalWrexhamUK
| | - Emma McCann
- Liverpool Women's Hospital Foundation TrustLiverpoolUK
| | - Shelagh Joss
- West of Scotland Centre for Genomic MedicineQueen Elizabeth University HospitalGlasgowUK
| | - Alexander J.M. Blakes
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
- Manchester Centre for Genomic MedicineUniversity of Manchester, St. Mary's Hospital, Manchester Academic Health Science CentreManchesterUK
| | - Kay Metcalfe
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
- Manchester Centre for Genomic MedicineUniversity of Manchester, St. Mary's Hospital, Manchester Academic Health Science CentreManchesterUK
| | - Helen Kingston
- Manchester Centre for Genomic MedicineUniversity of Manchester, St. Mary's Hospital, Manchester Academic Health Science CentreManchesterUK
| | - Marta Bertoli
- Northern Genetics ServiceInternational Centre for LifeNewcastle upon TyneUK
| | - Rachel Kneen
- Department of NeurologyAlder Hey Children's NHS Foundation TrustLiverpoolUK
| | - Sally Ann Lynch
- Department of Clinical GeneticsChildren's Health Ireland (CHI) at CrumlinDublinIreland
| | | | | | - Wendy D. Jones
- North East Thames Regional Genetics ServiceGreat Ormond Street Hospital for Children, Great Ormond Street NHS Foundation TrustLondonUK
| | | | - Esther B.E. Becker
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute of Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Andrea H. Németh
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Oxford Center for Genomic MedicineOxford University Hospitals National Health Service Foundation Trust, University of OxfordOxfordUK
| |
Collapse
|
8
|
Deng F, Fu M, Zhao C, Lei J, Xu T, Ji B, Ding H, Zhang Y, Chen J, Qiu J, Gao Q. Calcium signals and potential therapy targets in ovarian cancer (Review). Int J Oncol 2023; 63:125. [PMID: 37711071 PMCID: PMC10552713 DOI: 10.3892/ijo.2023.5573] [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: 03/23/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023] Open
Abstract
Ovarian cancer (OC) is a deadly disease. The poor prognosis and high lethality of OC are attributed to its high degrees of aggressiveness, resistance to chemotherapy and recurrence rates. Calcium ion (Ca2+) signaling has received attention in recent years, as it appears to form an essential part of various aspects of cancer pathophysiology and is a potential therapeutic target for OC treatment. Disruption of normal Ca2+ signaling pathways can induce changes in cell cycle progression, apoptosis, proliferation and migration and invasion, leading to the development of the malignant phenotype of tumors. In the present review, the main roles of ion channel/receptor/pump‑triggered Ca2+ signaling pathways located at the plasma membrane and organelle Ca2+ transport in OC are summarized. In addition, the potential of Ca2+ signaling as a novel target for the development of effective treatment strategies for OC was discussed. Furthering the understanding into the role of Ca2+ signaling in OC is expected to facilitated the identification of novel therapeutic targets and improved clinical outcomes for patients.
Collapse
Affiliation(s)
- Fengying Deng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Mengyu Fu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Chenxuan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiahui Lei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Bingyu Ji
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yueming Zhang
- Department of Gynecology and Obstetrics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215100, P.R. China
| | - Jie Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215153, P.R. China
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
9
|
Lemos FO, de Ridder I, Bootman MD, Bultynck G, Parys JB. The Complex Effects of PKM2 and PKM2:IP 3R Disruption on Intracellular Ca 2+ Handling and Cellular Functions. Cells 2023; 12:2527. [PMID: 37947604 PMCID: PMC10647343 DOI: 10.3390/cells12212527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Pyruvate kinase M (PKM) 2 was described to interact with the inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and suppress its activity. To further investigate the physiological importance of the PKM2:IP3R interaction, we developed and characterized HeLa PKM2 knockout (KO) cells. In the HeLa PKM2 KO cells, the release of Ca2+ to the cytosol appears to be more sensitive to low agonist concentrations than in HeLa wild-type (WT) cells. However, upon an identical IP3-induced Ca2+ release, Ca2+ uptake in the mitochondria is decreased in HeLa PKM2 KO cells, which may be explained by the smaller number of contact sites between the ER and the mitochondria. Furthermore, in HeLa PKM2 KO cells, mitochondria are more numerous, though they are smaller and less branched and have a hyperpolarized membrane potential. TAT-D5SD, a cell-permeable peptide representing a sequence derived from IP3R1 that can disrupt the PKM2:IP3R interaction, induces Ca2+ release into the cytosol and Ca2+ uptake into mitochondria in both HeLa WT and PKM2 KO cells. Moreover, TAT-D5SD induced apoptosis in HeLa WT and PKM2 KO cells but not in HeLa cells completely devoid of IP3Rs. These results indicate that PKM2 separately regulates cytosolic and mitochondrial Ca2+ handling and that the cytotoxic effect of TAT-D5SD depends on IP3R activity but not on PKM2. However, the tyrosine kinase Lck, which also interacts with the D5SD sequence, is expressed neither in HeLa WT nor PKM2 KO cells, and we can also exclude a role for PKM1, which is upregulated in HeLa PKM2 KO cells, indicating that the TAT-D5SD peptide has a more complex mode of action than anticipated.
Collapse
Affiliation(s)
- Fernanda O. Lemos
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI—B802, 3000 Leuven, Belgium; (I.d.R.); (G.B.)
| | - Ian de Ridder
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI—B802, 3000 Leuven, Belgium; (I.d.R.); (G.B.)
| | - Martin D. Bootman
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK;
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI—B802, 3000 Leuven, Belgium; (I.d.R.); (G.B.)
| | - Jan B. Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI—B802, 3000 Leuven, Belgium; (I.d.R.); (G.B.)
| |
Collapse
|
10
|
Becchetti A. Interplay of Ca 2+ and K + signals in cell physiology and cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:15-46. [PMID: 38007266 DOI: 10.1016/bs.ctm.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
The cytoplasmic Ca2+ concentration and the activity of K+ channels on the plasma membrane regulate cellular processes ranging from mitosis to oriented migration. The interplay between Ca2+ and K+ signals is intricate, and different cell types rely on peculiar cellular mechanisms. Derangement of these mechanisms accompanies the neoplastic progression. The calcium signals modulated by voltage-gated (KV) and calcium-dependent (KCa) K+ channel activity regulate progression of the cell division cycle, the release of growth factors, apoptosis, cell motility and migration. Moreover, KV channels regulate the cell response to the local microenvironment by assembling with cell adhesion and growth factor receptors. This chapter summarizes the pathophysiological roles of Ca2+ and K+ fluxes in normal and cancer cells, by concentrating on several biological systems in which these functions have been studied in depth, such as early embryos, mammalian cell lines, T lymphocytes, gliomas and colorectal cancer cells. A full understanding of the underlying mechanisms will offer a comprehensive view of the ion channel implication in cancer biology and suggest potential pharmacological targets for novel therapeutic approaches in oncology.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
11
|
Rönkkö J, Rodriguez Y, Rasila T, Torregrosa-Muñumer R, Pennonen J, Kvist J, Kuuluvainen E, Bosch LVD, Hietakangas V, Bultynck G, Tyynismaa H, Ylikallio E. Human IP 3 receptor triple knockout stem cells remain pluripotent despite altered mitochondrial metabolism. Cell Calcium 2023; 114:102782. [PMID: 37481871 DOI: 10.1016/j.ceca.2023.102782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are ER Ca2+-release channels that control a broad set of cellular processes. Animal models lacking IP3Rs in different combinations display severe developmental phenotypes. Given the importance of IP3Rs in human diseases, we investigated their role in human induced pluripotent stem cells (hiPSC) by developing single IP3R and triple IP3R knockouts (TKO). Genome edited TKO-hiPSC lacking all three IP3R isoforms, IP3R1, IP3R2, IP3R3, failed to generate Ca2+ signals in response to agonists activating GPCRs, but retained stemness and pluripotency. Steady state metabolite profiling and flux analysis of TKO-hiPSC indicated distinct alterations in tricarboxylic acid cycle metabolites consistent with a deficiency in their pyruvate utilization via pyruvate dehydrogenase, shifting towards pyruvate carboxylase pathway. These results demonstrate that IP3Rs are not essential for hiPSC identity and pluripotency but regulate mitochondrial metabolism. This set of knockout hiPSC is a valuable resource for investigating IP3Rs in human cell types of interest.
Collapse
Affiliation(s)
- Julius Rönkkö
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Yago Rodriguez
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Tiina Rasila
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Rubén Torregrosa-Muñumer
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Jana Pennonen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Emilia Kuuluvainen
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, 00790, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, 00790, Finland
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven - University of Leuven, 3000, Leuven, Belgium; VIB Center for Brain & Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
| | - Ville Hietakangas
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, 00790, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, 00790, Finland
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Leuven, 3000, Belgium
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Emil Ylikallio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland; Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland.
| |
Collapse
|
12
|
Baker MR, Fan G, Arige V, Yule DI, Serysheva II. Understanding IP 3R channels: From structural underpinnings to ligand-dependent conformational landscape. Cell Calcium 2023; 114:102770. [PMID: 37393815 PMCID: PMC10529787 DOI: 10.1016/j.ceca.2023.102770] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are ubiquitously expressed large-conductance Ca2+-permeable channels predominantly localized to the endoplasmic reticulum (ER) membranes of virtually all eukaryotic cell types. IP3Rs work as Ca2+ signaling hubs through which diverse extracellular stimuli and intracellular inputs are processed and then integrated to result in delivery of Ca2+ from the ER lumen to generate cytosolic Ca2+ signals with precise temporal and spatial properties. IP3R-mediated Ca2+ signals control a vast repertoire of cellular functions ranging from gene transcription and secretion to the more enigmatic brain activities such as learning and memory. IP3Rs open and release Ca2+ when they bind both IP3 and Ca2+, the primary channel agonists. Despite overwhelming evidence supporting functional interplay between IP3 and Ca2+ in activation and inhibition of IP3Rs, the mechanistic understanding of how IP3R channels convey their gating through the interplay of two primary agonists remains one of the major puzzles in the field. The last decade has seen much progress in the use of cryogenic electron microscopy to elucidate the molecular mechanisms of ligand binding, ion permeation, ion selectivity and gating of the IP3R channels. The results of these studies, summarized in this review, provide a prospective view of what the future holds in structural and functional research of IP3Rs.
Collapse
Affiliation(s)
- Mariah R Baker
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School at The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School at The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | - Vikas Arige
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - David I Yule
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA.
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School at The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Perry JL, Scribano FJ, Gebert JT, Engevik KA, Ellis JM, Hyser JM. The Inositol Trisphosphate Receptor (IP 3 R) is Dispensable for Rotavirus-induced Ca 2+ Signaling and Replication but Critical for Paracrine Ca 2+ Signals that Prime Uninfected Cells for Rapid Virus Spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552719. [PMID: 37609335 PMCID: PMC10441394 DOI: 10.1101/2023.08.09.552719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Rotavirus is a leading cause of viral gastroenteritis. A hallmark of rotavirus infection is an increase in cytosolic Ca 2+ caused by the nonstructural protein 4 (NSP4). NSP4 is a viral ion channel that releases Ca 2+ from the endoplasmic reticulum (ER) and the increase in Ca 2+ signaling is critical for rotavirus replication. In addition to NSP4 itself, host inositol 1,4,5- trisphosphate receptor (IP 3 R) ER Ca 2+ channels may contribute to rotavirus-induced Ca 2+ signaling and by extension, virus replication. Thus, we set out to determine the role of IP 3 R Ca 2+ signaling during rotavirus infection using IP 3 R-knockout MA104-GCaMP6s cells (MA104- GCaMP6s-IP 3 R-KO), generated by CRISPR/Cas9 genome editing. Live Ca 2+ imaging showed that IP 3 R-KO did not reduce Ca 2+ signaling in infected cells but eliminated rotavirus-induced intercellular Ca 2+ waves (ICWs) and therefore the increased Ca 2+ signaling in surrounding, uninfected cells. Further, MA104-GCaMP6s-IP 3 R-TKO cells showed similar rotavirus susceptibility, single-cycle replication, and viral protein expression as parental MA104- GCaMP6s cells. However, MA104-GCaMP6s-IP 3 R-TKO cells exhibited significantly smaller rotavirus plaques, decreased multi-round replication kinetics, and delayed virus spread, suggesting that rotavirus-induced ICW Ca 2+ signaling stimulates virus replication and spread. Inhibition of ICWs by blocking the P2Y1 receptor also resulted in decreased rotavirus plaque size. Conversely, exogenous expression of P2Y1 in LLC-MK2-GCaMP6s cells, which natively lack P2Y1 and rotavirus ICWs, rescued the generation of rotavirus-induced ICWs and enabled plaque formation. In conclusion, this study shows that NSP4 Ca 2+ signals fully support rotavirus replication in individual cells; however, IP 3 R is critical for rotavirus-induced ICWs and virus spread by priming Ca 2+ -dependent pathways in surrounding cells. Importance Many viruses exploit host Ca 2+ signaling to facilitate their replication; however, little is known about how distinct types of Ca 2+ signals contribute to the overall dysregulation of Ca 2+ signaling or promote virus replication. Using cells lacking IP 3 R, a host ER Ca 2+ channel, we could differentiate between intracellular Ca 2+ signals within virus-infected cells and intercellular Ca 2+ waves (ICWs), which increase Ca 2+ signaling in neighboring, uninfected cells. In infected cells, IP 3 R was dispensable for rotavirus-induced Ca 2+ signaling and replication, suggesting the rotavirus NSP4 viroporin supplies these signals. However, IP 3 R-mediated ICWs increase rotavirus replication kinetics and spread, indicating that the Ca 2+ signals from the ICWs may prime nearby uninfected cells to better support virus replication upon eventual infection. This "pre-emptive priming" of uninfected cells by exploiting host intercellular pathways in the vicinity of virus-infected cells represents a novel mechanism for viral reprogramming of the host to gain a replication advantage.
Collapse
|
14
|
Ling Z, Noda K, Frey BL, Hu M, Fok SW, Smith LM, Sanchez PG, Ren X. Newly synthesized glycoprotein profiling to identify molecular signatures of warm ischemic injury in donor lungs. Am J Physiol Lung Cell Mol Physiol 2023; 325:L30-L44. [PMID: 37130807 PMCID: PMC10292982 DOI: 10.1152/ajplung.00412.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/24/2023] [Accepted: 04/29/2023] [Indexed: 05/04/2023] Open
Abstract
Despite recent technological advances such as ex vivo lung perfusion (EVLP), the outcome of lung transplantation remains unsatisfactory with ischemic injury being a common cause for primary graft dysfunction. New therapeutic developments are hampered by limited understanding of pathogenic mediators of ischemic injury to donor lung grafts. Here, to identify novel proteomic effectors underlying the development of lung graft dysfunction, using bioorthogonal protein engineering, we selectively captured and identified newly synthesized glycoproteins (NewS-glycoproteins) produced during EVLP with unprecedented temporal resolution of 4 h. Comparing the NewS-glycoproteomes in lungs with and without warm ischemic injury, we discovered highly specific proteomic signatures with altered synthesis in ischemic lungs, which exhibited close association to hypoxia response pathways. Inspired by the discovered protein signatures, pharmacological modulation of the calcineurin pathway during EVLP of ischemic lungs offered graft protection and improved posttransplantation outcome. In summary, the described EVLP-NewS-glycoproteomics strategy delivers an effective new means to reveal molecular mediators of donor lung pathophysiology and offers the potential to guide future therapeutic development.NEW & NOTEWORTHY This study developed and implemented a bioorthogonal strategy to chemoselectively label, enrich, and characterize newly synthesized (NewS-)glycoproteins during 4-h ex vivo lung perfusion (EVLP). Through this approach, the investigators uncovered specific proteomic signatures associated with warm ischemic injury in donor lung grafts. These signatures exhibit high biological relevance to ischemia-reperfusion injury, validating the robustness of the presented approach.
Collapse
Affiliation(s)
- Zihan Ling
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Brian L Frey
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States
| | - Michael Hu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Shierly W Fok
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States
| | - Pablo G Sanchez
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
15
|
Gouriou Y, Gonnot F, Wehbi M, Brun C, Gomez L, Bidaux G. High-sensitivity calcium biosensor on the mitochondrial surface reveals that IP3R channels participate in the reticular Ca2+ leak towards mitochondria. PLoS One 2023; 18:e0285670. [PMID: 37294746 PMCID: PMC10256219 DOI: 10.1371/journal.pone.0285670] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/27/2023] [Indexed: 06/11/2023] Open
Abstract
Genetically encoded biosensors based on fluorescent proteins (FPs) are widely used to monitor dynamics and sub-cellular spatial distribution of calcium ion (Ca2+) fluxes and their role in intracellular signaling pathways. The development of different mutations in the Ca2+-sensitive elements of the cameleon probes has allowed sensitive range of Ca2+ measurements in almost all cellular compartments. Region of the endoplasmic reticulum (ER) tethered to mitochondria, named as the mitochondrial-associated membranes (MAMs), has received an extended attention since the last 5 years. Indeed, as MAMs are essential for calcium homeostasis and mitochondrial function, molecular tools have been developed to assess quantitatively Ca2+ levels in the MAMs. However, sensitivity of the first generation Ca2+ biosensors on the surface of the outer-mitochondrial membrane (OMM) do not allow to measure μM or sub-μM changes in Ca2+ concentration which prevents to measure the native activity (unstimulated exogenously) of endogenous channels. In this study, we assembled a new ratiometric highly sensitive Ca2+ biosensor expressed on the surface of the outer-mitochondrial membrane (OMM). It allows the detection of smaller differences than the previous biosensor in or at proximity of the MAMs. Noteworthy, we demonstrated that IP3-receptors have an endogenous activity which participate to the Ca2+ leak channel on the surface of the OMM during hypoxia or when SERCA activity is blocked.
Collapse
Affiliation(s)
- Yves Gouriou
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| | - Fabrice Gonnot
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| | - Mariam Wehbi
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| | - Camille Brun
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| | - Ludovic Gomez
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| | - Gabriel Bidaux
- Univ-Lyon CarMeN Laboratory, Inserm U1060, Université Claude Bernard Lyon 1, INRAE, Bron, France
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, Bron, France
| |
Collapse
|
16
|
Terry LE, Arige V, Neumann J, Wahl AM, Knebel TR, Chaffer JW, Malik S, Liston A, Humblet-Baron S, Bultynck G, Yule DI. Missense mutations in inositol 1,4,5-trisphosphate receptor type 3 result in leaky Ca 2+ channels and activation of store-operated Ca 2+ entry. iScience 2022; 25:105523. [PMID: 36444295 PMCID: PMC9700043 DOI: 10.1016/j.isci.2022.105523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Mutations in all subtypes of the inositol 1,4,5-trisphosphate receptor Ca2+ release channel are associated with human diseases. In this report, we investigated the functionality of three neuropathy-associated missense mutations in IP3R3 (V615M, T1424M, and R2524C). The mutants only exhibited function when highly over-expressed compared to endogenous hIP3R3. All variants resulted in elevated basal cytosolic Ca2+ levels, decreased endoplasmic reticulum Ca2+ store content, and constitutive store-operated Ca2+ entry in the absence of any stimuli, consistent with a leaky IP3R channel pore. These variants differed in channel function; when stably over-expressed the R2524C mutant was essentially dead, V615M was poorly functional, and T1424M exhibited activity greater than that of the corresponding wild-type following threshold stimulation. These results demonstrate that a common feature of these mutations is decreased IP3R3 function. In addition, these mutations exhibit a novel phenotype manifested as a constitutively open channel, which inappropriately gates SOCE in the absence of stimulation.
Collapse
Affiliation(s)
- Lara E. Terry
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Julika Neumann
- KU Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Amanda M. Wahl
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Taylor R. Knebel
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - James W. Chaffer
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Adrian Liston
- KU Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | | | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
17
|
Bonzerato CG, Keller KR, Schulman JJ, Gao X, Szczesniak LM, Wojcikiewicz RJH. Endogenous Bok is stable at the endoplasmic reticulum membrane and does not mediate proteasome inhibitor-induced apoptosis. Front Cell Dev Biol 2022; 10:1094302. [PMID: 36601536 PMCID: PMC9806350 DOI: 10.3389/fcell.2022.1094302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Controversy surrounds the cellular role of the Bcl-2 family protein Bok. On one hand, it has been shown that all endogenous Bok is bound to inositol 1,4,5-trisphosphate receptors (IP3Rs), while other data suggest that Bok can act as a pro-apoptotic mitochondrial outer membrane permeabilization mediator, apparently kept at very low and non-apoptotic levels by efficient proteasome-mediated degradation. Here we show that 1) endogenous Bok is expressed at readily-detectable levels in key cultured cells (e.g., mouse embryonic fibroblasts and HCT116 cells) and is not constitutively degraded by the proteasome, 2) proteasome inhibitor-induced apoptosis is not mediated by Bok, 3) endogenous Bok expression level is critically dependent on the presence of IP3Rs, 4) endogenous Bok is rapidly degraded by the ubiquitin-proteasome pathway in the absence of IP3Rs at the endoplasmic reticulum membrane, and 5) charged residues in the transmembrane region of Bok affect its stability, ability to interact with Mcl-1, and pro-apoptotic activity when over-expressed. Overall, these data indicate that endogenous Bok levels are not governed by proteasomal activity (except when IP3Rs are deleted) and that while endogenous Bok plays little or no role in apoptotic signaling, exogenous Bok can mediate apoptosis in a manner dependent on its transmembrane domain.
Collapse
|
18
|
Genovese M, Guidone D, Buccirossi M, Borrelli A, Rodriguez-Gimeno A, Bertozzi F, Bandiera T, Galietta LJV. Pharmacological potentiators of the calcium signaling cascade identified by high-throughput screening. PNAS NEXUS 2022; 2:pgac288. [PMID: 36712939 PMCID: PMC9830948 DOI: 10.1093/pnasnexus/pgac288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pharmacological modulators of the Ca2+ signaling cascade are important research tools and may translate into novel therapeutic strategies for a series of human diseases. We carried out a screening of a maximally diverse chemical library using the Ca2+-sensitive Cl- channel TMEM16A as a functional readout. We found compounds that were able to potentiate UTP-dependent TMEM16A activation. Mechanism of action of these compounds was investigated by a panel of assays that looked at intracellular Ca2+ mobilization triggered by extracellular agonists or by caged-IP3 photolysis, PIP2 breakdown by phospholipase C, and ion channel activity on nuclear membrane. One compound appears as a selective potentiator of inositol triphosphate receptor type 1 (ITPR1) with a possible application for some forms of spinocerebellar ataxia. A second compound is instead a potentiator of the P2RY2 purinergic receptor, an activity that could promote fluid secretion in dry eye and chronic obstructive respiratory diseases.
Collapse
Affiliation(s)
- Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, 80078 Naples, Italy
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, 80078 Naples, Italy
| | - Martina Buccirossi
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, 80078 Naples, Italy
| | - Anna Borrelli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, 80078 Naples, Italy
| | | | - Fabio Bertozzi
- D3 PharmaChemistry, Italian Institute of Technology (IIT), Via Morego, 3016163, Genoa, Italy
| | - Tiziano Bandiera
- D3 PharmaChemistry, Italian Institute of Technology (IIT), Via Morego, 3016163, Genoa, Italy
| | - Luis J V Galietta
- To whom correspondence should be addressed. Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.
| |
Collapse
|
19
|
Arige V, Terry LE, Wagner LE, Malik S, Baker MR, Fan G, Joseph SK, Serysheva II, Yule DI. Functional determination of calcium-binding sites required for the activation of inositol 1,4,5-trisphosphate receptors. Proc Natl Acad Sci U S A 2022; 119:e2209267119. [PMID: 36122240 PMCID: PMC9522344 DOI: 10.1073/pnas.2209267119] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/23/2022] [Indexed: 01/25/2023] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) initiate a diverse array of physiological responses by carefully orchestrating intracellular calcium (Ca2+) signals in response to various external cues. Notably, IP3R channel activity is determined by several obligatory factors, including IP3, Ca2+, and ATP. The critical basic amino acid residues in the N-terminal IP3-binding core (IBC) region that facilitate IP3 binding are well characterized. In contrast, the residues conferring regulation by Ca2+ have yet to be ascertained. Using comparative structural analysis of Ca2+-binding sites identified in two main families of intracellular Ca2+-release channels, ryanodine receptors (RyRs) and IP3Rs, we identified putative acidic residues coordinating Ca2+ in the cytosolic calcium sensor region in IP3Rs. We determined the consequences of substituting putative Ca2+ binding, acidic residues in IP3R family members. We show that the agonist-induced Ca2+ release, single-channel open probability (P0), and Ca2+ sensitivities are markedly altered when the negative charge on the conserved acidic side chain residues is neutralized. Remarkably, neutralizing the negatively charged side chain on two of the residues individually in the putative Ca2+-binding pocket shifted the Ca2+ required to activate IP3R to higher concentrations, indicating that these residues likely are a component of the Ca2+ activation site in IP3R. Taken together, our findings indicate that Ca2+ binding to a well-conserved activation site is a common underlying mechanism resulting in increased channel activity shared by IP3Rs and RyRs.
Collapse
Affiliation(s)
- Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - Lara E. Terry
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - Larry E. Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - Mariah R. Baker
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Suresh K. Joseph
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Irina I. Serysheva
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| |
Collapse
|
20
|
Arige V, Yule DI. Spatial and temporal crosstalk between the cAMP and Ca 2+ signaling systems. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119293. [PMID: 35588944 DOI: 10.1016/j.bbamcr.2022.119293] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/31/2022]
Abstract
The ubiquitous secondary messengers, Ca2+ and cAMP, play a vital role in shaping a diverse array of physiological processes. More significantly, accumulating evidence over the past several decades underpin extensive crosstalk between these two canonical messengers in discrete sub-cellular nanodomains across various cell types. Within such specialized nanodomains, each messenger fine-tunes signaling to maintain homeostasis by manipulating the activities of cellular machinery accountable for the metabolism or activity of the complementary pathway. Interaction between these messengers is ensured by scaffolding proteins which tether components of the signaling machinery in close proximity. Disruption of dynamic communications between Ca2+ and cAMP at these loci consequently is linked to several pathological conditions. This review summarizes recent novel mechanisms underlying effective crosstalk between Ca2+ and cAMP in such nanodomains.
Collapse
Affiliation(s)
- Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA..
| |
Collapse
|
21
|
Quarato G, Llambi F, Guy CS, Min J, Actis M, Sun H, Narina S, Pruett-Miller SM, Peng J, Rankovic Z, Green DR. Ca 2+-mediated mitochondrial inner membrane permeabilization induces cell death independently of Bax and Bak. Cell Death Differ 2022; 29:1318-1334. [PMID: 35726022 DOI: 10.1038/s41418-022-01025-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
The ability of mitochondria to buffer a rapid rise in cytosolic Ca2+ is a hallmark of proper cell homeostasis. Here, we employed m-3M3FBS, a putative phospholipase C (PLC) agonist, to explore the relationships between intracellular Ca2+ imbalance, mitochondrial physiology, and cell death. m-3M3FBS induced a potent dose-dependent Ca2+ release from the endoplasmic reticulum (ER), followed by a rise in intra-mitochondrial Ca2+. When the latter exceeded the organelle buffering capacity, an abrupt mitochondrial inner membrane permeabilization (MIMP) occurred, releasing matrix contents into the cytosol. MIMP was followed by cell death that was independent of Bcl-2 family members and inhibitable by the intracellular Ca2+ chelator BAPTA-AM. Cyclosporin A (CsA), capable of blocking the mitochondrial permeability transition (MPT), completely prevented cell death induced by m-3M3FBS. However, CsA acted upstream of mitochondria by preventing Ca2+ release from ER stores. Therefore, loss of Ca2+ intracellular balance and mitochondrial Ca2+ overload followed by MIMP induced a cell death process that is distinct from Bcl-2 family-regulated mitochondrial outer membrane permeabilization (MOMP). Further, the inhibition of cell death by CsA or its analogues can be independent of effects on the MPT.
Collapse
Affiliation(s)
- Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Fabien Llambi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Relay Therapeutics, Cambridge, MA, 02139, USA
| | - Cliff S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jaeki Min
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Marisa Actis
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Huan Sun
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
22
|
Nugues C, Helassa N, Haynes LP. Mitosis, Focus on Calcium. Front Physiol 2022; 13:951979. [PMID: 35784871 PMCID: PMC9247304 DOI: 10.3389/fphys.2022.951979] [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: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
The transformation of a single fertilised egg into an adult human consisting of tens of trillions of highly diverse cell types is a marvel of biology. The expansion is largely achieved by cell duplication through the process of mitosis. Mitosis is essential for normal growth, development, and tissue repair and is one of the most tightly regulated biological processes studied. This regulation is designed to ensure accurate segregation of chromosomes into each new daughter cell since errors in this process can lead to genetic imbalances, aneuploidy, that can lead to diseases including cancer. Understanding how mitosis operates and the molecular mechanisms that ensure its fidelity are therefore not only of significant intellectual value but provide unique insights into disease pathology. The purpose of this review is to revisit historical evidence that mitosis can be influenced by the ubiquitous second messenger calcium and to discuss this in the context of new findings revealing exciting new information about its role in cell division.
Collapse
Affiliation(s)
- Charlotte Nugues
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
23
|
Zhao J, Zhang H, Fan X, Yu X, Huai J. Lipid Dyshomeostasis and Inherited Cerebellar Ataxia. Mol Neurobiol 2022; 59:3800-3828. [PMID: 35420383 PMCID: PMC9148275 DOI: 10.1007/s12035-022-02826-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/01/2022] [Indexed: 12/04/2022]
Abstract
Cerebellar ataxia is a form of ataxia that originates from dysfunction of the cerebellum, but may involve additional neurological tissues. Its clinical symptoms are mainly characterized by the absence of voluntary muscle coordination and loss of control of movement with varying manifestations due to differences in severity, in the site of cerebellar damage and in the involvement of extracerebellar tissues. Cerebellar ataxia may be sporadic, acquired, and hereditary. Hereditary ataxia accounts for the majority of cases. Hereditary ataxia has been tentatively divided into several subtypes by scientists in the field, and nearly all of them remain incurable. This is mainly because the detailed mechanisms of these cerebellar disorders are incompletely understood. To precisely diagnose and treat these diseases, studies on their molecular mechanisms have been conducted extensively in the past. Accumulating evidence has demonstrated that some common pathogenic mechanisms exist within each subtype of inherited ataxia. However, no reports have indicated whether there is a common mechanism among the different subtypes of inherited cerebellar ataxia. In this review, we summarize the available references and databases on neurological disorders characterized by cerebellar ataxia and show that a subset of genes involved in lipid homeostasis form a new group that may cause ataxic disorders through a common mechanism. This common signaling pathway can provide a valuable reference for future diagnosis and treatment of ataxic disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Huan Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xue Yu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
24
|
Schmitz EA, Takahashi H, Karakas E. Structural basis for activation and gating of IP 3 receptors. Nat Commun 2022; 13:1408. [PMID: 35301323 PMCID: PMC8930994 DOI: 10.1038/s41467-022-29073-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/24/2022] [Indexed: 12/29/2022] Open
Abstract
A pivotal component of the calcium (Ca2+) signaling toolbox in cells is the inositol 1,4,5-triphosphate (IP3) receptor (IP3R), which mediates Ca2+ release from the endoplasmic reticulum (ER), controlling cytoplasmic and organellar Ca2+ concentrations. IP3Rs are co-activated by IP3 and Ca2+, inhibited by Ca2+ at high concentrations, and potentiated by ATP. However, the underlying molecular mechanisms are unclear. Here we report cryo-electron microscopy (cryo-EM) structures of human type-3 IP3R obtained from a single dataset in multiple gating conformations: IP3-ATP bound pre-active states with closed channels, IP3-ATP-Ca2+ bound active state with an open channel, and IP3-ATP-Ca2+ bound inactive state with a closed channel. The structures demonstrate how IP3-induced conformational changes prime the receptor for activation by Ca2+, how Ca2+ binding leads to channel opening, and how ATP modulates the activity, providing insights into the long-sought questions regarding the molecular mechanism underpinning receptor activation and gating. IP3 receptors are intracellular calcium channels involved in numerous signaling pathways. Here, the authors present the cryo-EM structures of type-3 IP3 receptors in multiple gating conformations, including the active state revealing the molecular mechanism of the receptor activation.
Collapse
Affiliation(s)
- Emily A Schmitz
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA.,Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hirohide Takahashi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA.,Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Erkan Karakas
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA. .,Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
25
|
Tariq K, Luikart BW. Striking a balance: PIP 2 and PIP 3 signaling in neuronal health and disease. EXPLORATION OF NEUROPROTECTIVE THERAPY 2022; 1:86-100. [PMID: 35098253 PMCID: PMC8797975 DOI: 10.37349/ent.2021.00008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phosphoinositides are membrane phospholipids involved in a variety of cellular processes like growth, development, metabolism, and transport. This review focuses on the maintenance of cellular homeostasis of phosphatidylinositol 4,5-bisphosphate (PIP2), and phosphatidylinositol 3,4,5-trisphosphate (PIP3). The critical balance of these PIPs is crucial for regulation of neuronal form and function. The activity of PIP2 and PIP3 can be regulated through kinases, phosphatases, phospholipases and cholesterol microdomains. PIP2 and PIP3 carry out their functions either indirectly through their effectors activating integral signaling pathways, or through direct regulation of membrane channels, transporters, and cytoskeletal proteins. Any perturbations to the balance between PIP2 and PIP3 signaling result in neurodevelopmental and neurodegenerative disorders. This review will discuss the upstream modulators and downstream effectors of the PIP2 and PIP3 signaling, in the context of neuronal health and disease.
Collapse
Affiliation(s)
- Kamran Tariq
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
26
|
A non-canonical role for pyruvate kinase M2 as a functional modulator of Ca 2+ signalling through IP 3 receptors. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119206. [PMID: 35026348 DOI: 10.1016/j.bbamcr.2021.119206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022]
Abstract
Pyruvate kinase isoform M2 (PKM2) is a rate-limiting glycolytic enzyme that is widely expressed in embryonic tissues. The expression of PKM2 declines in some tissues following embryogenesis, while other pyruvate kinase isozymes are upregulated. However, PKM2 is highly expressed in cancer cells and is believed to play a role in supporting anabolic processes during tumour formation. In this study, PKM2 was identified as an inositol 1,4,5-trisphosphate receptor (IP3R)-interacting protein by mass spectrometry. The PKM2:IP3R interaction was further characterized by pull-down and co-immunoprecipitation assays, which showed that PKM2 interacted with all three IP3R isoforms. Moreover, fluorescence microscopy indicated that both IP3R and PKM2 localized at the endoplasmic reticulum. PKM2 binds to IP3R at a highly conserved 21-amino acid site (corresponding to amino acids 2078-2098 in mouse type 1 IP3R isoform). Synthetic peptides (denoted 'TAT-D5SD' and 'D5SD'), based on the amino acid sequence at this site, disrupted the PKM2:IP3R interaction and potentiated IP3R-mediated Ca2+ release both in intact cells (TAT-D5SD peptide) and in a unidirectional 45Ca2+ flux assay on permeabilized cells (D5SD peptide). The TAT-D5SD peptide did not affect the enzymatic activity of PKM2. Reducing PKM2 protein expression using siRNA increased IP3R-mediated Ca2+ signalling in intact cells without altering the ER Ca2+ content. These data identify PKM2 as an IP3R-interacting protein that inhibits intracellular Ca2+ signalling. The elevated expression of PKM2 in cancer cells is therefore not solely connected to its canonical role in glycolytic metabolism, rather PKM2 also has a novel non-canonical role in regulating intracellular signalling.
Collapse
|
27
|
Dhanya SK, Hasan G. Deficits Associated With Loss of STIM1 in Purkinje Neurons Including Motor Coordination Can Be Rescued by Loss of Septin 7. Front Cell Dev Biol 2021; 9:794807. [PMID: 34993201 PMCID: PMC8724567 DOI: 10.3389/fcell.2021.794807] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/26/2022] Open
Abstract
Septins are cytoskeletal proteins that can assemble to form heteromeric filamentous complexes and regulate a range of membrane-associated cellular functions. SEPT7, a member of the septin family, functions as a negative regulator of the plasma membrane–localized store-operated Ca2+ entry (SOCE) channel, Orai in Drosophila neurons, and in human neural progenitor cells. Knockdown of STIM, a Ca2+ sensor in the endoplasmic reticulum (ER) and an integral component of SOCE, leads to flight deficits in Drosophila that can be rescued by partial loss of SEPT7 in neurons. Here, we tested the effect of reducing and removing SEPT7 in mouse Purkinje neurons (PNs) with the loss of STIM1. Mice with the complete knockout of STIM1 in PNs exhibit several age-dependent changes. These include altered gene expression in PNs, which correlates with increased synapses between climbing fiber (CF) axons and Purkinje neuron (PN) dendrites and a reduced ability to learn a motor coordination task. Removal of either one or two copies of the SEPT7 gene in STIM1KO PNs restored the expression of a subset of genes, including several in the category of neuron projection development. Importantly, the rescue of gene expression in these animals is accompanied by normal CF-PN innervation and an improved ability to learn a motor coordination task in aging mice. Thus, the loss of SEPT7 in PNs further modulates cerebellar circuit function in STIM1KO animals. Our findings are relevant in the context of identifying SEPT7 as a putative therapeutic target for various neurodegenerative diseases caused by reduced intracellular Ca2+ signaling.
Collapse
Affiliation(s)
- Sreeja Kumari Dhanya
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- SASTRA University, Thanjavur, India
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- *Correspondence: Gaiti Hasan,
| |
Collapse
|
28
|
Young MP, Schug ZT, Booth DM, Yule DI, Mikoshiba K, Hajnόczky G, Joseph SK. Metabolic adaptation to the chronic loss of Ca 2+ signaling induced by KO of IP 3 receptors or the mitochondrial Ca 2+ uniporter. J Biol Chem 2021; 298:101436. [PMID: 34801549 PMCID: PMC8672050 DOI: 10.1016/j.jbc.2021.101436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023] Open
Abstract
Calcium signaling is essential for regulating many biological processes. Endoplasmic reticulum inositol trisphosphate receptors (IP3Rs) and the mitochondrial Ca2+ uniporter (MCU) are key proteins that regulate intracellular Ca2+ concentration. Mitochondrial Ca2+ accumulation activates Ca2+-sensitive dehydrogenases of the tricarboxylic acid (TCA) cycle that maintain the biosynthetic and bioenergetic needs of both normal and cancer cells. However, the interplay between calcium signaling and metabolism is not well understood. In this study, we used human cancer cell lines (HEK293 and HeLa) with stable KOs of all three IP3R isoforms (triple KO [TKO]) or MCU to examine metabolic and bioenergetic responses to the chronic loss of cytosolic and/or mitochondrial Ca2+ signaling. Our results show that TKO cells (exhibiting total loss of Ca2+ signaling) are viable, displaying a lower proliferation and oxygen consumption rate, with no significant changes in ATP levels, even when made to rely solely on the TCA cycle for energy production. MCU KO cells also maintained normal ATP levels but showed increased proliferation, oxygen consumption, and metabolism of both glucose and glutamine. However, MCU KO cells were unable to maintain ATP levels and died when relying solely on the TCA cycle for energy. We conclude that constitutive Ca2+ signaling is dispensable for the bioenergetic needs of both IP3R TKO and MCU KO human cancer cells, likely because of adequate basal glycolytic and TCA cycle flux. However, in MCU KO cells, the higher energy expenditure associated with increased proliferation and oxygen consumption makes these cells more prone to bioenergetic failure under conditions of metabolic stress.
Collapse
Affiliation(s)
- Michael P Young
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Zachary T Schug
- Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - David M Booth
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - David I Yule
- Department of Pharmacology & Physiology, University of Rochester, Rochester, New York, USA
| | - Katsuhiko Mikoshiba
- Shanghai Institute of Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China; Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Japan
| | - Gyӧrgy Hajnόczky
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Suresh K Joseph
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
29
|
Rosa N, Ivanova H, Wagner LE, Kale J, La Rovere R, Welkenhuyzen K, Louros N, Karamanou S, Shabardina V, Lemmens I, Vandermarliere E, Hamada K, Ando H, Rousseau F, Schymkowitz J, Tavernier J, Mikoshiba K, Economou A, Andrews DW, Parys JB, Yule DI, Bultynck G. Bcl-xL acts as an inhibitor of IP 3R channels, thereby antagonizing Ca 2+-driven apoptosis. Cell Death Differ 2021; 29:788-805. [PMID: 34750538 PMCID: PMC8990011 DOI: 10.1038/s41418-021-00894-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/09/2022] Open
Abstract
Anti-apoptotic Bcl-2-family members not only act at mitochondria but also at the endoplasmic reticulum, where they impact Ca2+ dynamics by controlling IP3 receptor (IP3R) function. Current models propose distinct roles for Bcl-2 vs. Bcl-xL, with Bcl-2 inhibiting IP3Rs and preventing pro-apoptotic Ca2+ release and Bcl-xL sensitizing IP3Rs to low [IP3] and promoting pro-survival Ca2+ oscillations. We here demonstrate that Bcl-xL too inhibits IP3R-mediated Ca2+ release by interacting with the same IP3R regions as Bcl-2. Via in silico superposition, we previously found that the residue K87 of Bcl-xL spatially resembled K17 of Bcl-2, a residue critical for Bcl-2's IP3R-inhibitory properties. Mutagenesis of K87 in Bcl-xL impaired its binding to IP3R and abrogated Bcl-xL's inhibitory effect on IP3Rs. Single-channel recordings demonstrate that purified Bcl-xL, but not Bcl-xLK87D, suppressed IP3R single-channel openings stimulated by sub-maximal and threshold [IP3]. Moreover, we demonstrate that Bcl-xL-mediated inhibition of IP3Rs contributes to its anti-apoptotic properties against Ca2+-driven apoptosis. Staurosporine (STS) elicits long-lasting Ca2+ elevations in wild-type but not in IP3R-knockout HeLa cells, sensitizing the former to STS treatment. Overexpression of Bcl-xL in wild-type HeLa cells suppressed STS-induced Ca2+ signals and cell death, while Bcl-xLK87D was much less effective in doing so. In the absence of IP3Rs, Bcl-xL and Bcl-xLK87D were equally effective in suppressing STS-induced cell death. Finally, we demonstrate that endogenous Bcl-xL also suppress IP3R activity in MDA-MB-231 breast cancer cells, whereby Bcl-xL knockdown augmented IP3R-mediated Ca2+ release and increased the sensitivity towards STS, without altering the ER Ca2+ content. Hence, this study challenges the current paradigm of divergent functions for Bcl-2 and Bcl-xL in Ca2+-signaling modulation and reveals that, similarly to Bcl-2, Bcl-xL inhibits IP3R-mediated Ca2+ release and IP3R-driven cell death. Our work further underpins that IP3R inhibition is an integral part of Bcl-xL's anti-apoptotic function.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Hristina Ivanova
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Larry E Wagner
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Justin Kale
- Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Rita La Rovere
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Kirsten Welkenhuyzen
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Nikolaos Louros
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Spyridoula Karamanou
- KU Leuven, Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Campus Gasthuisberg P.O, Box 1037, Herestraat 49, 3000, Leuven, Belgium
| | - Victoria Shabardina
- Institut of Evolutionary Biology, CSIC-Universitat Pompeu Fabra, Passeig Marítim de la Barceloneta 37-49, 08003, Barcelona, Spain
| | - Irma Lemmens
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biomolecular Medicine, Ghent University, and Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | | | - Kozo Hamada
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Hideaki Ando
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, 351-0198, Saitama, Japan
| | - Frederic Rousseau
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Joost Schymkowitz
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biomolecular Medicine, Ghent University, and Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Katsuhiko Mikoshiba
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China.,Faculty of Science, Toho University, Miyama 2-2-1, Funabashi, 274-8510, Chiba, Japan
| | - Anastassios Economou
- KU Leuven, Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Campus Gasthuisberg P.O, Box 1037, Herestraat 49, 3000, Leuven, Belgium
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
30
|
Arige V, Terry LE, Malik S, Knebel TR, Wagner II LE, Yule DI. CREB regulates the expression of type 1 inositol 1,4,5-trisphosphate receptors. J Cell Sci 2021; 134:jcs258875. [PMID: 34533188 PMCID: PMC8601716 DOI: 10.1242/jcs.258875] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022] Open
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) play a central role in regulating intracellular Ca2+ signals in response to a variety of internal and external cues. Dysregulation of IP3R signaling is the underlying cause for numerous pathological conditions. It is well established that the activities of IP3Rs are governed by several post-translational modifications, including phosphorylation by protein kinase A (PKA). However, the long-term effects of PKA activation on expression of IP3R subtypes remains largely unexplored. In this report, we investigate the effects of chronic stimulation and tonic activity of PKA on the expression of IP3R subtypes. We demonstrate that expression of the type 1 IP3R (IP3R1) is augmented upon prolonged activation of PKA or upon ectopic overexpression of cyclic AMP-response element-binding protein (CREB) without altering IP3R2 and IP3R3 abundance. By contrast, inhibition of PKA or blocking CREB diminished IP3R1 expression. We also demonstrate that agonist-induced Ca2+-release mediated by IP3R1 is significantly attenuated upon blocking of CREB. Moreover, CREB - by regulating the expression of KRAS-induced actin-interacting protein (KRAP) - ensures correct localization and licensing of IP3R1. Overall, we report a crucial role for CREB in governing both the expression and correct localization of IP3R1. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | | | | | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
31
|
Muñoz Cardona ML, López Mahecha JM. Gillespie’s Syndrome Phenotype in A Patient with a Homozygous Variant of Uncertain Significance in the ITPR1 Gene. Neuroophthalmology 2021; 46:186-189. [DOI: 10.1080/01658107.2021.1982991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
32
|
Woll KA, Van Petegem F. Calcium Release Channels: Structure and Function of IP3 Receptors and Ryanodine Receptors. Physiol Rev 2021; 102:209-268. [PMID: 34280054 DOI: 10.1152/physrev.00033.2020] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ca2+-release channels are giant membrane proteins that control the release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. The two members, ryanodine receptors (RyRs) and inositol-1,4,5-trisphosphate Receptors (IP3Rs), are evolutionarily related and are both activated by cytosolic Ca2+. They share a common architecture, but RyRs have evolved additional modules in the cytosolic region. Their massive size allows for the regulation by tens of proteins and small molecules, which can affect the opening and closing of the channels. In addition to Ca2+, other major triggers include IP3 for the IP3Rs, and depolarization of the plasma membrane for a particular RyR subtype. Their size has made them popular targets for study via electron microscopic methods, with current structures culminating near 3Å. The available structures have provided many new mechanistic insights int the binding of auxiliary proteins and small molecules, how these can regulate channel opening, and the mechanisms of disease-associated mutations. They also help scrutinize previously proposed binding sites, as some of these are now incompatible with the structures. Many questions remain around the structural effects of post-translational modifications, additional binding partners, and the higher-order complexes these channels can make in situ. This review summarizes our current knowledge about the structures of Ca2+-release channels and how this informs on their function.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
Stochastic reaction-diffusion modeling of calcium dynamics in 3D dendritic spines of Purkinje cells. Biophys J 2021; 120:2112-2123. [PMID: 33887224 PMCID: PMC8390834 DOI: 10.1016/j.bpj.2021.03.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/22/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) is a second messenger assumed to control changes in synaptic strength in the form of both long-term depression and long-term potentiation at Purkinje cell dendritic spine synapses via inositol trisphosphate (IP3)-induced Ca2+ release. These Ca2+ transients happen in response to stimuli from parallel fibers (PFs) from granule cells and climbing fibers (CFs) from the inferior olivary nucleus. These events occur at low numbers of free Ca2+, requiring stochastic single-particle methods when modeling them. We use the stochastic particle simulation program MCell to simulate Ca2+ transients within a three-dimensional Purkinje cell dendritic spine. The model spine includes the endoplasmic reticulum, several Ca2+ transporters, and endogenous buffer molecules. Our simulations successfully reproduce properties of Ca2+ transients in different dynamical situations. We test two different models of the IP3 receptor (IP3R). The model with nonlinear concentration response of binding of activating Ca2+ reproduces experimental results better than the model with linear response because of the filtering of noise. Our results also suggest that Ca2+-dependent inhibition of the IP3R needs to be slow to reproduce experimental results. Simulations suggest the experimentally observed optimal timing window of CF stimuli arises from the relative timing of CF influx of Ca2+ and IP3 production sensitizing IP3R for Ca2+-induced Ca2+ release. We also model ataxia, a loss of fine motor control assumed to be the result of malfunctioning information transmission at the granule to Purkinje cell synapse, resulting in a decrease or loss of Ca2+ transients. Finally, we propose possible ways of recovering Ca2+ transients under ataxia.
Collapse
|
34
|
Lee JH, Ryu SW, Ender NA, Paull TT. Poly-ADP-ribosylation drives loss of protein homeostasis in ATM and Mre11 deficiency. Mol Cell 2021; 81:1515-1533.e5. [PMID: 33571423 PMCID: PMC8026623 DOI: 10.1016/j.molcel.2021.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Loss of the ataxia-telangiectasia mutated (ATM) kinase causes cerebellum-specific neurodegeneration in humans. We previously demonstrated that deficiency in ATM activation via oxidative stress generates insoluble protein aggregates in human cells, reminiscent of protein dysfunction in common neurodegenerative disorders. Here, we show that this process is driven by poly-ADP-ribose polymerases (PARPs) and that the insoluble protein species arise from intrinsically disordered proteins associating with PAR-associated genomic sites in ATM-deficient cells. The lesions implicated in this process are single-strand DNA breaks dependent on reactive oxygen species, transcription, and R-loops. Human cells expressing Mre11 A-T-like disorder mutants also show PARP-dependent aggregation identical to ATM deficiency. Lastly, analysis of A-T patient cerebellum samples shows widespread protein aggregation as well as loss of proteins known to be critical in human spinocerebellar ataxias that is not observed in neocortex tissues. These results provide a hypothesis accounting for loss of protein integrity and cerebellum function in A-T.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Seung W Ryu
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Nicolette A Ender
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Tanya T Paull
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA.
| |
Collapse
|
35
|
Balancing ER-Mitochondrial Ca 2+ Fluxes in Health and Disease. Trends Cell Biol 2021; 31:598-612. [PMID: 33678551 DOI: 10.1016/j.tcb.2021.02.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
Organelles cooperate with each other to control cellular homeostasis and cell functions by forming close connections through membrane contact sites. Important contacts are present between the endoplasmic reticulum (ER), the main intracellular Ca2+-storage organelle, and the mitochondria, the organelle responsible not only for the majority of cellular ATP production but also for switching on cell death processes. Several Ca2+-transport systems focalize at these contact sites, thereby enabling the efficient transmission of Ca2+ signals from the ER toward mitochondria. This provides tight control of mitochondrial functions at the microdomain level. Here, we discuss how ER-mitochondrial Ca2+ transfers support cell function and how their dysregulation underlies, drives, or contributes to pathogenesis and pathophysiology, with a major focus on cancer and neurodegeneration but also with attention to other diseases such as diabetes and rare genetic diseases.
Collapse
|
36
|
Lagos-Cabré R, Ivanova A, Taylor CW. Ca 2+ Release by IP 3 Receptors Is Required to Orient the Mitotic Spindle. Cell Rep 2020; 33:108483. [PMID: 33326774 PMCID: PMC7758162 DOI: 10.1016/j.celrep.2020.108483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 09/23/2020] [Accepted: 11/13/2020] [Indexed: 11/30/2022] Open
Abstract
The mitotic spindle distributes chromosomes evenly to daughter cells during mitosis. The orientation of the spindle, guided by internal and external cues, determines the axis of cell division and thereby contributes to tissue morphogenesis. Progression through mitosis requires local Ca2+ signals at critical steps, and because store-operated Ca2+ entry is inhibited during mitosis, those signals probably require Ca2+ release through inositol 1,4,5-trisphosphate receptors (IP3Rs). In cells without IP3Rs, astral microtubules around the daughter centrosome are shorter than those at the mother centrosome, and the mitotic spindle fails to align with the substratum during metaphase. The misalignment is due to the spindle ineffectively detecting internal cues rather than a failure of cells to recognize the substratum. Expression of type 3 IP3R is sufficient to rescue spindle alignment, but only if the IP3R has a functional pore. We conclude that Ca2+ signals evoked by IP3Rs are required to orient the mitotic spindle. IP3 receptors are required for mitotic spindle orientation Only IP3 receptors with a functional channel restore spindle orientation Ca2+ release through IP3 receptors is required for spindle orientation
Collapse
Affiliation(s)
- Raul Lagos-Cabré
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Adelina Ivanova
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
37
|
Sharma A, Hasan G. Modulation of flight and feeding behaviours requires presynaptic IP 3Rs in dopaminergic neurons. eLife 2020; 9:e62297. [PMID: 33155978 PMCID: PMC7647402 DOI: 10.7554/elife.62297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/25/2020] [Indexed: 12/17/2022] Open
Abstract
Innate behaviours, although robust and hard wired, rely on modulation of neuronal circuits, for eliciting an appropriate response according to internal states and external cues. Drosophila flight is one such innate behaviour that is modulated by intracellular calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs). Cellular mechanism(s) by which IP3Rs modulate neuronal function for specific behaviours remain speculative, in vertebrates and invertebrates. To address this, we generated an inducible dominant negative form of the IP3R (IP3RDN). Flies with neuronal expression of IP3RDN exhibit flight deficits. Expression of IP3RDN helped identify key flight-modulating dopaminergic neurons with axonal projections in the mushroom body. Flies with attenuated IP3Rs in these presynaptic dopaminergic neurons exhibit shortened flight bouts and a disinterest in seeking food, accompanied by reduced excitability and dopamine release upon cholinergic stimulation. Our findings suggest that the same neural circuit modulates the drive for food search and for undertaking longer flight bouts.
Collapse
Affiliation(s)
- Anamika Sharma
- National Centre for Biological Sciences, TIFRBangaloreIndia
| | - Gaiti Hasan
- National Centre for Biological Sciences, TIFRBangaloreIndia
| |
Collapse
|
38
|
Hasan G, Sharma A. Regulation of neuronal physiology by Ca2+ release through the IP3R. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
39
|
Altered Organelle Calcium Transport in Ovarian Physiology and Cancer. Cancers (Basel) 2020; 12:cancers12082232. [PMID: 32785177 PMCID: PMC7464720 DOI: 10.3390/cancers12082232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022] Open
Abstract
Calcium levels have a huge impact on the physiology of the female reproductive system, in particular, of the ovaries. Cytosolic calcium levels are influenced by regulatory proteins (i.e., ion channels and pumps) localized in the plasmalemma and/or in the endomembranes of membrane-bound organelles. Imbalances between plasma membrane and organelle-based mechanisms for calcium regulation in different ovarian cell subtypes are contributing to ovarian pathologies, including ovarian cancer. In this review, we focused our attention on altered calcium transport and its role as a contributor to tumor progression in ovarian cancer. The most important proteins described as contributing to ovarian cancer progression are inositol trisphosphate receptors, ryanodine receptors, transient receptor potential channels, calcium ATPases, hormone receptors, G-protein-coupled receptors, and/or mitochondrial calcium uniporters. The involvement of mitochondrial and/or endoplasmic reticulum calcium imbalance in the development of resistance to chemotherapeutic drugs in ovarian cancer is also discussed, since Ca2+ channels and/or pumps are nowadays regarded as potential therapeutic targets and are even correlated with prognosis.
Collapse
|
40
|
Robinson KJ, Watchon M, Laird AS. Aberrant Cerebellar Circuitry in the Spinocerebellar Ataxias. Front Neurosci 2020; 14:707. [PMID: 32765211 PMCID: PMC7378801 DOI: 10.3389/fnins.2020.00707] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022] Open
Abstract
The spinocerebellar ataxias (SCAs) are a heterogeneous group of neurodegenerative diseases that share convergent disease features. A common symptom of these diseases is development of ataxia, involving impaired balance and motor coordination, usually stemming from cerebellar dysfunction and neurodegeneration. For most spinocerebellar ataxias, pathology can be attributed to an underlying gene mutation and the impaired function of the encoded protein through loss or gain-of-function effects. Strikingly, despite vast heterogeneity in the structure and function of disease-causing genes across the SCAs and the cellular processes affected, the downstream effects have considerable overlap, including alterations in cerebellar circuitry. Interestingly, aberrant function and degeneration of Purkinje cells, the major output neuronal population present within the cerebellum, precedes abnormalities in other neuronal populations within many SCAs, suggesting that Purkinje cells have increased vulnerability to cellular perturbations. Factors that are known to contribute to perturbed Purkinje cell function in spinocerebellar ataxias include altered gene expression resulting in altered expression or functionality of proteins and channels that modulate membrane potential, downstream impairments in intracellular calcium homeostasis and changes in glutamatergic input received from synapsing climbing or parallel fibers. This review will explore this enhanced vulnerability and the aberrant cerebellar circuitry linked with it in many forms of SCA. It is critical to understand why Purkinje cells are vulnerable to such insults and what overlapping pathogenic mechanisms are occurring across multiple SCAs, despite different underlying genetic mutations. Enhanced understanding of disease mechanisms will facilitate the development of treatments to prevent or slow progression of the underlying neurodegenerative processes, cerebellar atrophy and ataxic symptoms.
Collapse
Affiliation(s)
| | | | - Angela S. Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Science, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
41
|
SEPT7 regulates Ca 2+ entry through Orai channels in human neural progenitor cells and neurons. Cell Calcium 2020; 90:102252. [PMID: 32682163 DOI: 10.1016/j.ceca.2020.102252] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/25/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
Human neural progenitor cells (hNPCs) are self-renewing cells of neural lineage that can be differentiated into neurons of different subtypes. Here we show that SEPT7, a member of the family of filament-forming GTPases called septins, prevents constitutive Ca2+ entry through the store-operated Ca2+ entry channel, Orai in hNPCs and in differentiated neurons and is thus required for neuronal calcium homeostasis. Previous work in Drosophila neurons has shown that loss of one copy of the evolutionarily-conserved dSEPT7 gene leads to elevated Ca2+ entry via Orai, in the absence of ER-Ca2+ store depletion. We have identified an N-terminal polybasic region of SEPT7, known to interact with membrane-localized phospholipids, as essential for spontaneous calcium entry through Orai in hNPCs, whereas the GTPase domain of dSEPT7 is dispensable for this purpose. Re-organisation of Orai1 and the ER-Ca2+ sensor STIM1 observed near the plasma membrane in SEPT7 KD hNPCs, supports the idea that Septin7 containing heteromers prevent Ca2+ entry through a fraction of STIM-Orai complexes. Possible mechanisms by which SEPT7 reduction leads to opening of Orai channels in the absence of store-depletion are discussed.
Collapse
|
42
|
Chi TH, Hsieh BY, Liang PS, Han TH, Hsieh M. Expression and Functional Study of Single Mutations of Carbonic Anhydrase 8 in Neuronal Cells. Cell Mol Neurobiol 2020; 41:1355-1371. [PMID: 32583043 DOI: 10.1007/s10571-020-00907-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/18/2020] [Indexed: 11/27/2022]
Abstract
Carbonic anhydrase 8 (CA8), an isozyme of α-carbonic anhydrases, lacks the ability to catalyze the reversible hydration of CO2 to bicarbonate and proton. Previous studies have shown that single point mutations of CA8, CA8-S100P, and CA8-G162R, are associated with novel syndromes including congenital ataxia and mild cognitive impairment. Our previous results demonstrated that overexpression of wild type (WT) CA8 promoted cell proliferation, neurite outgrowth, anti-apoptosis, invasion and migration abilities in neuronal cells. In this study, we examined the expressions and functions of CA8-S100P and CA8-G162R in neuroblastoma cells lines, compared with those of WT CA8. Our results show that the protein expressions of mutant CA8-S100P and CA8-G162R were significantly decreased in Neuro-2a and SK-N-SH cells. Interestingly, CA8-S100P demonstrated a significant increase in cell proliferation in both Neuro-2a and SK-N-SH cells. However, both CA8 mutations showed significantly decreased effects on cell protection and migration in SK-N-SH cells. Surprisingly, a significant increase of invasive ability was observed in SK-N-SH cells with overexpression of CA8-S100P as compared with those with overexpression of WT CA8 under retinoic acid (RA) treatment. In addition, we found that Neuro-2a cells with overexpression of CA8-S100P and CA8-G162R showed significantly increased neurite outgrowth. Taken together, our data suggest that the expressions of CA8-S100P and CA8-G162R in neuronal cells alter cell morphology, proliferation, mobility and viability; indicating that the homozygous point mutations of CA8 lead to not only the loss of WT CA8 function, but also the gain of novel functions leading to neuromuscular dysfunction.
Collapse
Affiliation(s)
- Tang-Hao Chi
- Department of Life Science, Tunghai University, No.1727, Sec. 4, Taiwan Boulevard, Taichung, 407, Taiwan, ROC
| | - Benjamin Y Hsieh
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Pei-Shin Liang
- Department of Life Science, Tunghai University, No.1727, Sec. 4, Taiwan Boulevard, Taichung, 407, Taiwan, ROC
| | - Tien-Heng Han
- Department of Life Science, Tunghai University, No.1727, Sec. 4, Taiwan Boulevard, Taichung, 407, Taiwan, ROC
| | - Mingli Hsieh
- Department of Life Science, Tunghai University, No.1727, Sec. 4, Taiwan Boulevard, Taichung, 407, Taiwan, ROC. .,Life Science Research Center, Tunghai University, Taichung, Taiwan, ROC.
| |
Collapse
|
43
|
Rare CACNA1A mutations leading to congenital ataxia. Pflugers Arch 2020; 472:791-809. [DOI: 10.1007/s00424-020-02396-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/03/2023]
|
44
|
Inositol 1,4,5-Trisphosphate Receptors in Human Disease: A Comprehensive Update. J Clin Med 2020; 9:jcm9041096. [PMID: 32290556 PMCID: PMC7231134 DOI: 10.3390/jcm9041096] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/22/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (ITPRs) are intracellular calcium release channels located on the endoplasmic reticulum of virtually every cell. Herein, we are reporting an updated systematic summary of the current knowledge on the functional role of ITPRs in human disorders. Specifically, we are describing the involvement of its loss-of-function and gain-of-function mutations in the pathogenesis of neurological, immunological, cardiovascular, and neoplastic human disease. Recent results from genome-wide association studies are also discussed.
Collapse
|
45
|
Seki T. [Exploration of preventive drugs for spinocerebellar ataxia using cultured cerebellar Purkinje cells]. Nihon Yakurigaku Zasshi 2019; 154:310-314. [PMID: 31787682 DOI: 10.1254/fpj.154.310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Neurodegenerative diseases are caused by progressive degeneration of specific neurons. To overcome neurodegenerative diseases, the exploitation of preventive drugs is strongly expected, since impaired neurons are not regenerated by drugs. Spinocerebellar ataxia (SCA) is a group of dominantly inherited neurodegenerative diseases and is characterized by the progressive cerebellar ataxia. To date, SCA is classified into SCA1-48 by the variance of causal genes. Since SCA patients are commonly characterized by cerebellar ataxia and atrophy of cerebellum, it is possible that there are common pathogenic mechanisms in SCAs. However, there are not any shared functions among SCA-causing proteins. Cerebellar Purkinje cells (PCs) are sole output neurons from cerebellar cortexes, crucial for cerebellar functions and characterized highly branched dendrites. During the exploration of the molecular pathogenesis of several SCA-causing proteins, we found that several SCA-causing proteins commonly trigger the impairment of dendritic development of primary cultured cerebellar PCs. Dendritic shrinkage of cerebellar PCs has been observed and is considered to be related to the motor dysfunction in several SCA model mice. Therefore, we assume that impaired dendritic development of cultured cerebellar Purkinje cells is one of the common phenotypes of SCA in vitro and that cultured PCs expressing SCA-causing proteins could be in vitro SCA models. This SCA model would be useful for the efficient exploration of novel preventive drugs against various types of SCAs.
Collapse
Affiliation(s)
- Takahiro Seki
- Department of Chemico-Pharmacological Science, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
46
|
Abstract
In the body, extracellular stimuli produce inositol 1,4,5-trisphosphate (IP3), an intracellular chemical signal that binds to the IP3 receptor (IP3R) to release calcium ions (Ca2+) from the endoplasmic reticulum. In the past 40 years, the wide-ranging functions mediated by IP3R and its genetic defects causing a variety of disorders have been unveiled. Recent cryo-electron microscopy and X-ray crystallography have resolved IP3R structures and begun to integrate with concurrent functional studies, which can explicate IP3-dependent opening of Ca2+-conducting gates placed ∼90 Å away from IP3-binding sites and its regulation by Ca2+. This review highlights recent research progress on the IP3R structure and function. We also propose how protein plasticity within IP3R, which involves allosteric gating and assembly transformations accompanied by rapid and chronic structural changes, would enable it to regulate diverse functions at cellular microdomains in pathophysiological states.
Collapse
Affiliation(s)
- Kozo Hamada
- Laboratory of Cell Calcium Signaling, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China; ,
| | - Katsuhiko Mikoshiba
- Laboratory of Cell Calcium Signaling, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China; ,
| |
Collapse
|
47
|
Ma HL, Yu SJ, Chen J, Ding XF, Chen G, Liang Y, Pan JL. CA8 promotes RCC proliferation and migration though its expression level is lower in tumor compared to adjacent normal tissue. Biomed Pharmacother 2019; 121:109578. [PMID: 31715371 DOI: 10.1016/j.biopha.2019.109578] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy and radiotherapy are not as successful in the case of renal cell carcinoma (RCC) although some targeted drugs were approved for RCC therapy recently. Analysis of whole genomic data will lead to improvements in understanding RCC and identifying novel anticancer targets. Here, we found the differential mRNA expression and copy number variation (CNV) of Carbonic anhydrase-related protein VIII (CA8) gene in RCC through integrated bioinformatics analysis of TCGA database, which was confirmed in 5 cases of samples collected from RCC patients who underwent radical nephrectomy by analysis of CA8 mRNA and protein levels using RT-PCR immunohistochemical assay. However, we got a completely opposite result that CA8 promoted RCC progression, those are CA8 overexpression promoted the proliferative and migratory ability of Caki-1 and 769-P cells in vitro as determined with MTT and transwell assay, and CA8 overexpression could also promote Caki-1 xenograft growth in BALB/C‑nu/nu mice. On the contrary, CA8-knockdown reduced Caki-1 and 769-P cell proliferation and migration. Moreover, knockdown of CA8 decreased pAKT and MMP2 protein levels in Caki-1 cells while overexpressing CA8 increased pAKT and MMP2. In conclusion, we showed that CA8 promoted RCC cell proliferation and migration, but it was down-regulated in RCC, which requires an additional mechanism study.
Collapse
Affiliation(s)
- Huai-Lu Ma
- Department of Pharmacology, School of Clinical Medicine, Taizhou University, Taizhou, Zhejiang 318000, China; Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei 075000, China; School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Sheng-Jian Yu
- Department of Pharmacology, School of Clinical Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Jie Chen
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Xiao-Fei Ding
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Guang Chen
- Department of Pharmacology, School of Clinical Medicine, Taizhou University, Taizhou, Zhejiang 318000, China; School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China.
| | - Yong Liang
- Institute of Tumor, Taizhou University, School of Medicine, 1139 Shi-Fu Avenue, Taizhou, Zhejiang 318000, China.
| | - Jian-Li Pan
- Pharmacy Department, Eye Hospital of Wenzhou Medical University, NO. 618, Fengqi East Road, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
48
|
Ngo KJ, Poke G, Neas K, Fogel BL. Spinocerebellar Ataxia type 29 in a family of Māori descent. CEREBELLUM & ATAXIAS 2019; 6:14. [PMID: 31632679 PMCID: PMC6790028 DOI: 10.1186/s40673-019-0108-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 09/26/2019] [Indexed: 12/30/2022]
Abstract
Background Mutations in the Inositol 1,4,5-Trisphosphate Receptor Type 1 (ITPR1) gene cause spinocerebellar ataxia type 29 (SCA29), a rare congenital-onset autosomal dominant non-progressive cerebellar ataxia. The Māori, indigenous to New Zealand, are an understudied population for genetic ataxias. Case presentation We investigated the genetic origins of spinocerebellar ataxia in a family of Māori descent consisting of two affected sisters and their unaffected parents. Whole exome sequencing identified a pathogenic variant, p.Thr267Met, in ITPR1 in both sisters, establishing their diagnosis as SCA29. Conclusions We report the identification of a family of Māori descent with a mutation causing SCA29, extending the worldwide scope of this disease. Although this mutation has occurred de novo in other populations, suggesting a mutational hotspot, the children in this family inherited it from their unaffected mother who was germline mosaic.
Collapse
Affiliation(s)
- Kathie J Ngo
- 1Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Gonda Room 6554, Los Angeles, CA 90095 USA
| | - Gemma Poke
- Genetic Health Service NZ, Wellington, New Zealand
| | | | - Brent L Fogel
- 1Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Gonda Room 6554, Los Angeles, CA 90095 USA.,3Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
49
|
Wagner M, Osborn DPS, Gehweiler I, Nagel M, Ulmer U, Bakhtiari S, Amouri R, Boostani R, Hentati F, Hockley MM, Hölbling B, Schwarzmayr T, Karimiani EG, Kernstock C, Maroofian R, Müller-Felber W, Ozkan E, Padilla-Lopez S, Reich S, Reichbauer J, Darvish H, Shahmohammadibeni N, Tafakhori A, Vill K, Zuchner S, Kruer MC, Winkelmann J, Jamshidi Y, Schüle R. Bi-allelic variants in RNF170 are associated with hereditary spastic paraplegia. Nat Commun 2019; 10:4790. [PMID: 31636353 PMCID: PMC6803694 DOI: 10.1038/s41467-019-12620-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/18/2019] [Indexed: 12/11/2022] Open
Abstract
Alterations of Ca2+ homeostasis have been implicated in a wide range of neurodegenerative diseases. Ca2+ efflux from the endoplasmic reticulum into the cytoplasm is controlled by binding of inositol 1,4,5-trisphosphate to its receptor. Activated inositol 1,4,5-trisphosphate receptors are then rapidly degraded by the endoplasmic reticulum-associated degradation pathway. Mutations in genes encoding the neuronal isoform of the inositol 1,4,5-trisphosphate receptor (ITPR1) and genes involved in inositol 1,4,5-trisphosphate receptor degradation (ERLIN1, ERLIN2) are known to cause hereditary spastic paraplegia (HSP) and cerebellar ataxia. We provide evidence that mutations in the ubiquitin E3 ligase gene RNF170, which targets inositol 1,4,5-trisphosphate receptors for degradation, are the likely cause of autosomal recessive HSP in four unrelated families and functionally evaluate the consequences of mutations in patient fibroblasts, mutant SH-SY5Y cells and by gene knockdown in zebrafish. Our findings highlight inositol 1,4,5-trisphosphate signaling as a candidate key pathway for hereditary spastic paraplegias and cerebellar ataxias and thus prioritize this pathway for therapeutic interventions.
Collapse
Affiliation(s)
- Matias Wagner
- Institute of Human Genetics, Technische Universität München, Trogerstraße 32, 81675, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Daniel P S Osborn
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - Ina Gehweiler
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Maike Nagel
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Ulrike Ulmer
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Somayeh Bakhtiari
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, 85016, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Rim Amouri
- Neurology Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
- Neuroscience Department, Faculty of Medicine of Tunis, University Tunis El Manar, Tunis, Tunisia
| | | | - Faycal Hentati
- Neurology Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
- Neuroscience Department, Faculty of Medicine of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Maryam M Hockley
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Benedikt Hölbling
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Thomas Schwarzmayr
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Ehsan Ghayoor Karimiani
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
- Next Generation Genetic Clinic, Mashhad, Iran
| | - Christoph Kernstock
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Reza Maroofian
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - Wolfgang Müller-Felber
- Department of Pediatric Neurology and Developmental Medicine, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Ege Ozkan
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - Sergio Padilla-Lopez
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, 85016, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Selina Reich
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Jennifer Reichbauer
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany
| | - Hossein Darvish
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Katharina Vill
- Department of Pediatric Neurology and Developmental Medicine, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation, Department of Human Genetics, FL33136, Miami, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, FL33136, Miami, USA
| | - Michael C Kruer
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, 85016, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Juliane Winkelmann
- Institute of Human Genetics, Technische Universität München, Trogerstraße 32, 81675, Munich, Germany
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yalda Jamshidi
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - Rebecca Schüle
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Str. 27, 72076, Tübingen, Germany.
| |
Collapse
|
50
|
Bartok A, Weaver D, Golenár T, Nichtova Z, Katona M, Bánsághi S, Alzayady KJ, Thomas VK, Ando H, Mikoshiba K, Joseph SK, Yule DI, Csordás G, Hajnóczky G. IP 3 receptor isoforms differently regulate ER-mitochondrial contacts and local calcium transfer. Nat Commun 2019; 10:3726. [PMID: 31427578 PMCID: PMC6700175 DOI: 10.1038/s41467-019-11646-3] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 07/12/2019] [Indexed: 12/31/2022] Open
Abstract
Contact sites of endoplasmic reticulum (ER) and mitochondria locally convey calcium signals between the IP3 receptors (IP3R) and the mitochondrial calcium uniporter, and are central to cell survival. It remains unclear whether IP3Rs also have a structural role in contact formation and whether the different IP3R isoforms have redundant functions. Using an IP3R-deficient cell model rescued with each of the three IP3R isoforms and an array of super-resolution and ultrastructural approaches we demonstrate that IP3Rs are required for maintaining ER-mitochondrial contacts. This role is independent of calcium fluxes. We also show that, while each isoform can support contacts, type 2 IP3R is the most effective in delivering calcium to the mitochondria. Thus, these studies reveal a non-canonical, structural role for the IP3Rs and direct attention towards the type 2 IP3R that was previously neglected in the context of ER-mitochondrial calcium signaling.
Collapse
Affiliation(s)
- Adam Bartok
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Departent of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - David Weaver
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Tünde Golenár
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zuzana Nichtova
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Máté Katona
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Száva Bánsághi
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kamil J Alzayady
- Department of Physiology and Pharmacology, University of Rochester, Rochester, NY, USA
| | - V Kaye Thomas
- Department of Physiology and Pharmacology, University of Rochester, Rochester, NY, USA
| | - Hideaki Ando
- Lab for Developmental Neurobiology, RIKEN Brain Science Institute, Saitama, Japan
- Laboratory of Molecular Biomedicine for Pathogenesis, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Katsuhiko Mikoshiba
- Lab for Developmental Neurobiology, RIKEN Brain Science Institute, Saitama, Japan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Suresh K Joseph
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - David I Yule
- Department of Physiology and Pharmacology, University of Rochester, Rochester, NY, USA
| | - György Csordás
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|