1
|
Cao C, Nipu N, Johnston W, Adil S, Wei L, Mennigen JA. Chronic hypoxia induces alternative splicing of transcripts in the goldfish brain. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:92. [PMID: 40332630 DOI: 10.1007/s10695-025-01505-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025]
Abstract
Several species evolved mechanisms to tolerate periods of severe environmental hypoxia and anoxia. Among them, goldfish are unique as they do not enter a comatose state under such conditions. Taking advantage of the recently published and annotated goldfish genome, we had previously profiled the transcriptomic response of the goldfish brain under normoxic (21 kPa oxygen saturation, N) and hypoxic conditions (2.1 kPa oxygen saturation) after 1 and 4 weeks (1WH, 4WH). Using the RNA-Seq data, we report the occurrence of alternative mRNA splicing (skipped exon, retained intron, alternative 3' or 5' splice sites, and mutually exclusive exons). At 1WH/N, 1004 significant alternative splicing events on 769 gene loci were identified, increasing to 1187 on 963 loci at 4WH/N. There were 305 loci with alternatively spliced transcripts common to both 1WH/N and 4WH/N, 221 of which exhibited the same precise location and splicing mechanism. Specific gene transcripts affected by alternative splicing events were almost entirely different from previously identified differentially expressed genes under chronic hypoxia. GO-term enrichment analyses of gene loci of alternatively spliced transcripts, however, did include similar pathways as previously identified for DEGs. These include epigenetic machinery, ion channel activity (1WH/N), glutamate signaling (4WH/N), endothelial cell function, and ATP hydrolyzation pathways (1WH/N + 4WH/N). We describe selected examples of alternatively spliced transcripts to discuss possible functional relevance in the goldfish brain response to chronic hypoxia. Together, our data identified an additional layer of regulation in brain pathways relevant to hypoxia tolerance in goldfish, which complement previously reported gene expression changes.
Collapse
Affiliation(s)
- Catherine Cao
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada
| | - Niepukolie Nipu
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada
| | - William Johnston
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada
| | - Sally Adil
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada
| | - Lai Wei
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada
| | - Jan A Mennigen
- Department of Biology, University of Ottawa, K1 N6 N5 20 Marie Curie, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Quinodoz M, Rutz S, Peter V, Garavelli L, Innes AM, Lehmann EF, Kellenberger S, Peng Z, Barone A, Campos-Xavier B, Unger S, Rivolta C, Dutzler R, Superti-Furga A. De novo variants in LRRC8C resulting in constitutive channel activation cause a human multisystem disorder. EMBO J 2025; 44:413-436. [PMID: 39623139 PMCID: PMC11729881 DOI: 10.1038/s44318-024-00322-y] [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: 04/02/2024] [Revised: 08/23/2024] [Accepted: 11/08/2024] [Indexed: 01/15/2025] Open
Abstract
Volume-regulated anion channels (VRACs) are multimeric proteins composed of different paralogs of the LRRC8 family. They are activated in response to hypotonic swelling, but little is known about their specific functions. We studied two human individuals with the same congenital syndrome affecting blood vessels, brain, eyes, and bones. The LRRC8C gene harbored de novo variants in both patients, located in a region of the gene encoding the boundary between the pore and a cytoplasmic domain, which is depleted of sequence variations in control subjects. When studied by cryo-EM, both LRRC8C mutant proteins assembled as their wild-type counterparts, but showed increased flexibility, suggesting a destabilization of subunit interactions. When co-expressed with the obligatory LRRC8A subunit, the mutants exhibited enhanced activation, resulting in channel activity even at isotonic conditions in which wild-type channels are closed. We conclude that structural perturbations of LRRC8C impair channel gating and constitute the mechanistic basis of the dominant gain-of-function effect of these pathogenic variants. The pleiotropic phenotype of this novel clinical entity associated with monoallelic LRRC8C variants indicates the fundamental roles of VRACs in different tissues and organs.
Collapse
Affiliation(s)
- Mathieu Quinodoz
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031, Basel, Switzerland
- Department of Ophthalmology, University of Basel, 4031, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Sonja Rutz
- Department of Biochemistry University of Zurich, 8057, Zurich, Switzerland
| | - Virginie Peter
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031, Basel, Switzerland
- Department of Ophthalmology, University of Basel, 4031, Basel, Switzerland
| | - Livia Garavelli
- Clinical Genetics Unit, Azienda USL-IRCCS of Reggio Emilia, 42123, Reggio Emilia, Italy
| | - A Micheil Innes
- Department of Medical Genetics and Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T3B 6A8, Canada
| | - Elena F Lehmann
- Department of Biochemistry University of Zurich, 8057, Zurich, Switzerland
| | - Stephan Kellenberger
- Department of biomedical Sciences, University of, Lausanne, 1011, Lausanne, Switzerland
| | - Zhong Peng
- Department of biomedical Sciences, University of, Lausanne, 1011, Lausanne, Switzerland
| | - Angelica Barone
- Pediatric Onco-Hematology Unit, Children's Hospital, Parma University Hospital, Parma, Italy
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), and University of Lausanne, 1011, Lausanne, Switzerland
| | - Sheila Unger
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), and University of Lausanne, 1011, Lausanne, Switzerland
- Genetica AG, Zurich and Lausanne, Switzerland
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031, Basel, Switzerland.
- Department of Ophthalmology, University of Basel, 4031, Basel, Switzerland.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK.
| | - Raimund Dutzler
- Department of Biochemistry University of Zurich, 8057, Zurich, Switzerland.
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), and University of Lausanne, 1011, Lausanne, Switzerland.
- Genetica AG, Zurich and Lausanne, Switzerland.
| |
Collapse
|
3
|
Scotton E, Ziani PR, Wilges RLB, da Rosa Correa PH, Giordano LA, Goularte JF, Schons T, Almeida FB, Stein DJ, de Castro JM, de Bastiani MA, de Oliveira Soares EG, Paixão DB, da Silva CDG, Schneider PH, Colombo R, Rosa AR. Molecular signature underlying (R)-ketamine rapid antidepressant response on anhedonic-like behavior induced by sustained exposure to stress. Pharmacol Biochem Behav 2024; 245:173882. [PMID: 39488299 DOI: 10.1016/j.pbb.2024.173882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 11/04/2024]
Abstract
Anhedonia induced by sustained stress exposure is a hallmark symptom of major depressive disorder (MDD) and in rodents, it can be accessed through the sucrose preference test (SPT). (R)-ketamine is a fast-acting antidepressant with less detrimental side effects and abuse liability compared to racemic ketamine. The present study combined high-throughput proteomics and network analysis to identify molecular mechanisms involved in chronic variable stress (CVS)-induced anhedonia and promising targets underlying (R)-ketamine rapid antidepressant response. Male Wistar rats were subjected to CVS for five weeks. Based on the SPT, animals were clustered into resilient or anhedonic-like (ANH) groups. ANH rats received a single dose of saline or (R)-ketamine (20 mg/kg, i.p.), which was proceeded by treatment response evaluation. After prefrontal cortex collection, proteomic analysis was performed to uncover the differentially expressed proteins (DEPs) related to both anhedonic-like behavior and pharmacological response. The behavioral assessment showed that the ANH animals had a significant decrease in SPT, and that (R)-ketamine responders showed a reversal of anhedonic-like behavior. On a molecular level, anhedonia-like behavior was associated with the downregulation of Neuronal Pentraxin Receptor (Nptxr) and Galectin-1 (Gal-1). These data reinforce a disruption in the inflammatory response, neurotransmitter receptor activity, and glutamatergic synapses in chronic stress-induced anhedonia. (R)-ketamine response-associated DEPs included novel potential targets involved in the modulation of oxidative stress, energetic metabolism, synaptogenesis, dendritic arborization, neuroinflammation, gene expression, and telomere length, converging to biological themes extensively documented in MDD physiopathology. Our data provide valuable insights into the molecular mechanisms underlying the response to (R)-ketamine and highlight these pathways as potential therapeutic targets for anhedonia. By addressing proteins involved in oxidative stress, energy metabolism, synaptogenesis, dendritic arborization, neuroinflammation, gene expression, and telomere length, we can target multiple key factors involved in the pathophysiology of MDD. Modulating these proteins could open avenues for novel therapeutic strategies and deepen our understanding of anhedonia, offering hope for improved outcomes in individuals facing this challenging condition. However, additional studies will be essential to validate these findings and further explore their therapeutic implications.
Collapse
Affiliation(s)
- Ellen Scotton
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Paola Rampelotto Ziani
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Renata Luiza Boff Wilges
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Pedro Henrique da Rosa Correa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Lucas Azambuja Giordano
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Jéferson Ferraz Goularte
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Tainá Schons
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Felipe Borges Almeida
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Dirson João Stein
- Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Josimar Macedo de Castro
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; School of Medicine and Post-Graduate Program in Medical Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Marco Antônio de Bastiani
- Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | - Douglas Bernardo Paixão
- Institute of Chemistry, Laboratory of Molecular Catalysis, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Caren Daniele Galeano da Silva
- Institute of Chemistry, Laboratory of Molecular Catalysis, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Paulo Henrique Schneider
- Institute of Chemistry, Laboratory of Molecular Catalysis, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rafael Colombo
- Institute of Biotechnology, University of Caxias do Sul (UCS), Caxias do Sul, RS, Brazil..
| | - Adriane R Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
4
|
Bell TA, Luce BE, Hakim P, Ananda VY, Dardari H, Nguyen TH, Monshizadeh A, Chao LH. Prominin 1 and Tweety Homology 1 both induce extracellular vesicle formation. eLife 2024; 13:e100061. [PMID: 39136554 PMCID: PMC11405016 DOI: 10.7554/elife.100061] [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: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024] Open
Abstract
Prominin 1 (Prom1) is a five-transmembrane pass integral membrane protein that associates with curved regions of the plasma membrane. Prom1 interacts with membrane cholesterol and actively remodels the plasma membrane. Membrane-bending activity is particularly evident in photoreceptors, where Prom1 loss-of-function mutations cause failure of outer segment homeostasis, leading to cone-rod retinal dystrophy (CRRD). The Tweety Homology (Ttyh) protein family has been proposed to be homologous to Prominin, but it is not known whether Ttyh proteins have an analogous membrane-bending function. Here, we characterize the membrane-bending activity of human Prom1 and Ttyh1 in native bilayer membranes. We find that Prom1 and Ttyh1 both induce formation of extracellular vesicles (EVs) in cultured mammalian cells and that the EVs produced are physically similar. Ttyh1 is more abundant in EV membranes than Prom1 and produces EVs with membranes that are more tubulated than Prom1 EVs. We further show that Prom1 interacts more stably with membrane cholesterol than Ttyh1 and that this may contribute to membrane-bending inhibition in Prom1 EVs. Intriguingly, a loss-of-function mutation in Prom1 associated with CRRD induces particularly stable cholesterol binding. These experiments provide mechanistic insight into Prominin function in CRRD and suggest that Prom and Ttyh belong to a single family of functionally related membrane-bending, EV-generating proteins.
Collapse
Affiliation(s)
- Tristan A Bell
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Bridget E Luce
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Pusparanee Hakim
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Virly Y Ananda
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Hiba Dardari
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Tran H Nguyen
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Arezu Monshizadeh
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Luke H Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
5
|
Lu P, Deng S, Liu J, Xiao Q, Zhou Z, Li S, Xin J, Shu G, Yi B, Yin G. Tweety homolog 3 promotes colorectal cancer progression through mutual regulation of histone deacetylase 7. MedComm (Beijing) 2024; 5:e576. [PMID: 38827027 PMCID: PMC11141500 DOI: 10.1002/mco2.576] [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: 07/24/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 06/04/2024] Open
Abstract
Colorectal cancer (CRC) is one of the leading cancers worldwide, with metastasis being a major cause of high mortality rates among patients. In this study, dysregulated gene Tweety homolog 3 (TTYH3) was identified by Gene Expression Omnibus database. Public databases were used to predict potential competing endogenous RNAs (ceRNAs) for TTYH3. Quantitative real-time polymerase chain reaction, western blot, and immunohistochemistry were utilized to analyze TTYH3 and histone deacetylase 7 (HDAC7) levels. Luciferase assays confirmed miR-1271-5p directly targeting the 3' untranslated regions of TTYH3 and HDAC7. In vitro experiments such as transwell and human umbilical vein endothelial cell tube formation, as well as in vivo mouse models, were conducted to assess the biological functions of TTYH3 and HDAC7. We discovered that upregulation of TTYH3 in CRC promotes cell migration by affecting the Epithelial-mesenchymal transition pathway, which was independent of its ion channel activity. Mechanistically, TTYH3 and HDAC7 functioned as ceRNAs, reciprocally regulating each other's expression. TTYH3 competes for binding miR-1271-5p, increasing HDAC7 expression, facilitating CRC metastasis and angiogenesis. This study reveals the critical role of TTYH3 in promoting CRC metastasis through ceRNA crosstalk, offering new insights into potential therapeutic targets for clinical intervention.
Collapse
Affiliation(s)
- Pengyan Lu
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Shumin Deng
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Jiaxin Liu
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Qing Xiao
- Department of PathologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Zhengwei Zhou
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Shuojie Li
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Jiaxuan Xin
- Department of Gastrointestinal SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Guang Shu
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
| | - Bo Yi
- Department of PathologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Gang Yin
- Department of PathologyXiangya Hospital, School of Basic Medical Sciences, Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
- China‐Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
6
|
Bell TA, Luce BE, Hakim P, Ananda VY, Dardari H, Nguyen TH, Monshizadeh A, Chao LH. Prominin 1 and Tweety Homology 1 both induce extracellular vesicle formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.08.566258. [PMID: 37986829 PMCID: PMC10659291 DOI: 10.1101/2023.11.08.566258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Prominin-1 (Prom1) is a five-transmembrane-pass integral membrane protein that associates with curved regions of the plasma membrane. Prom1 interacts with membrane cholesterol and actively remodels the plasma membrane. Membrane bending activity is particularly evident in photoreceptors, where Prom1 loss-of-function mutations cause failure of outer segment homeostasis, leading to cone-rod retinal dystrophy (CRRD). The Tweety Homology (Ttyh) protein family has been proposed to be homologous to Prominin, but it is not known whether Ttyh proteins have an analogous membrane-bending function. Here, we characterize the membrane-bending activity of human Prom1 and Ttyh1 in native bilayer membranes. We find that Prom1 and Ttyh1 both induce formation of extracellular vesicles (EVs) in cultured mammalian cells and that the EVs produced are physically similar. Ttyh1 is more abundant in EV membranes than Prom1 and produces EVs with membranes that are more tubulated than Prom1 EVs. We further show that Prom1 interacts more stably with membrane cholesterol than Ttyh1 and that this may contribute to membrane bending inhibition in Prom1 EVs. Intriguingly, a loss-of-function mutation in Prom1 associated with CRRD induces particularly stable cholesterol binding. These experiments provide mechanistic insight into Prominin function in CRRD and suggest that Prom and Ttyh belong to a single family of functionally related membrane-bending, EV-generating proteins.
Collapse
Affiliation(s)
- Tristan A. Bell
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115
- Current Address: Generate Biomedicines, 101 South St, Somerville, MA, 02143
| | - Bridget E. Luce
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Pusparanee Hakim
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Virly Y. Ananda
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Hiba Dardari
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Tran H. Nguyen
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Arezu Monshizadeh
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
| | - Luke H. Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115
| |
Collapse
|
7
|
Leung HH, Mansour C, Rousseau M, Nakhla A, Kiselyov K, Venkatachalam K, Wong CO. Drosophila tweety facilitates autophagy to regulate mitochondrial homeostasis and bioenergetics in Glia. Glia 2024; 72:433-451. [PMID: 37870193 PMCID: PMC10842981 DOI: 10.1002/glia.24484] [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: 06/02/2023] [Revised: 09/12/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
Mitochondria support the energetic demands of the cells. Autophagic turnover of mitochondria serves as a critical pathway for mitochondrial homeostasis. It is unclear how bioenergetics and autophagy are functionally connected. Here, we identify an endolysosomal membrane protein that facilitates autophagy to regulate ATP production in glia. We determined that Drosophila tweety (tty) is highly expressed in glia and localized to endolysosomes. Diminished fusion between autophagosomes and endolysosomes in tty-deficient glia was rescued by expressing the human Tweety Homolog 1 (TTYH1). Loss of tty in glia attenuated mitochondrial turnover, elevated mitochondrial oxidative stress, and impaired locomotor functions. The cellular and organismal defects were partially reversed by antioxidant treatment. We performed live-cell imaging of genetically encoded metabolite sensors to determine the impact of tty and autophagy deficiencies on glial bioenergetics. We found that tty-deficient glia exhibited reduced mitochondrial pyruvate consumption accompanied by a shift toward glycolysis for ATP production. Likewise, genetic inhibition of autophagy in glia resulted in a similar glycolytic shift in bioenergetics. Furthermore, the survival of mutant flies became more sensitive to starvation, underlining the significance of tty in the crosstalk between autophagy and bioenergetics. Together, our findings uncover the role for tty in mitochondrial homeostasis via facilitating autophagy, which determines bioenergetic balance in glia.
Collapse
Affiliation(s)
- Ho Hang Leung
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
- Present address: South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Christina Mansour
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Morgan Rousseau
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center (UTHealth), Houston, TX 77030, USA
| | - Anwar Nakhla
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, PA 15260, USA
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center (UTHealth), Houston, TX 77030, USA
| | - Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
8
|
Okada Y. Physiology of the volume-sensitive/regulatory anion channel VSOR/VRAC. Part 1: from its discovery and phenotype characterization to the molecular entity identification. J Physiol Sci 2024; 74:3. [PMID: 38238667 PMCID: PMC10795261 DOI: 10.1186/s12576-023-00897-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024]
Abstract
The volume-sensitive outwardly rectifying or volume-regulated anion channel, VSOR/VRAC, which was discovered in 1988, is expressed in most vertebrate cell types and is essentially involved in cell volume regulation after swelling and in the induction of cell death. This series of review articles describes what is already known and what remains to be uncovered about the functional and molecular properties as well as the physiological and pathophysiological roles of VSOR/VRAC. This Part 1 review article describes, from the physiological standpoint, first its discovery and significance in cell volume regulation, second its phenotypical properties, and third its molecular identification. Although the pore-forming core molecules and the volume-sensing subcomponent of VSOR/VRAC were identified as LRRC8 members and TRPM7 in 2014 and 2021, respectively, it is stressed that the identification of the molecular entity of VSOR/VRAC is still not complete enough to explain the full set of phenotypical properties.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan.
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan.
- Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan.
| |
Collapse
|
9
|
Yoshie S, Murono S, Hazama A. Approach for Elucidating the Molecular Mechanism of Epithelial to Mesenchymal Transition in Fibrosis of Asthmatic Airway Remodeling Focusing on Cl - Channels. Int J Mol Sci 2023; 25:289. [PMID: 38203460 PMCID: PMC10779031 DOI: 10.3390/ijms25010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Airway remodeling caused by asthma is characterized by structural changes of subepithelial fibrosis, goblet cell metaplasia, submucosal gland hyperplasia, smooth muscle cell hyperplasia, and angiogenesis, leading to symptoms such as dyspnea, which cause marked quality of life deterioration. In particular, fibrosis exacerbated by asthma progression is reportedly mediated by epithelial-mesenchymal transition (EMT). It is well known that the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling is closely associated with several signaling pathways, including the TGF-β1/Smad, TGF-β1/non-Smad, and Wnt/β-catenin signaling pathways. However, the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling has not yet been fully clarified. Given that Cl- transport through Cl- channels causes passive water flow and consequent changes in cell volume, these channels may be considered to play a key role in EMT, which is characterized by significant morphological changes. In the present article, we highlight how EMT, which causes fibrosis and carcinogenesis in various tissues, is strongly associated with activation or inactivation of Cl- channels and discuss whether Cl- channels can lead to elucidation of the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Susumu Yoshie
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeyuki Murono
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akihiro Hazama
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
10
|
Chen GL, Li J, Zhang J, Zeng B. To Be or Not to Be an Ion Channel: Cryo-EM Structures Have a Say. Cells 2023; 12:1870. [PMID: 37508534 PMCID: PMC10378246 DOI: 10.3390/cells12141870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Ion channels are the second largest class of drug targets after G protein-coupled receptors. In addition to well-recognized ones like voltage-gated Na/K/Ca channels in the heart and neurons, novel ion channels are continuously discovered in both excitable and non-excitable cells and demonstrated to play important roles in many physiological processes and diseases such as developmental disorders, neurodegenerative diseases, and cancer. However, in the field of ion channel discovery, there are an unignorable number of published studies that are unsolid and misleading. Despite being the gold standard of a functional assay for ion channels, electrophysiological recordings are often accompanied by electrical noise, leak conductance, and background currents of the membrane system. These unwanted signals, if not treated properly, lead to the mischaracterization of proteins with seemingly unusual ion-conducting properties. In the recent ten years, the technical revolution of cryo-electron microscopy (cryo-EM) has greatly advanced our understanding of the structures and gating mechanisms of various ion channels and also raised concerns about the pore-forming ability of some previously identified channel proteins. In this review, we summarize cryo-EM findings on ion channels with molecular identities recognized or disputed in recent ten years and discuss current knowledge of proposed channel proteins awaiting cryo-EM analyses. We also present a classification of ion channels according to their architectures and evolutionary relationships and discuss the possibility and strategy of identifying more ion channels by analyzing structures of transmembrane proteins of unknown function. We propose that cross-validation by electrophysiological and structural analyses should be essentially required for determining molecular identities of novel ion channels.
Collapse
Affiliation(s)
- Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jian Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
11
|
Liu Y, Wang XR, Jiang YH, Li T, Ling S, Wang HY, Yu JW, Jia SW, Liu XY, Hou CM, Parpura V, Wang YF. Interactions between the Astrocytic Volume-Regulated Anion Channel and Aquaporin 4 in Hyposmotic Regulation of Vasopressin Neuronal Activity in the Supraoptic Nucleus. Cells 2023; 12:1723. [PMID: 37443757 PMCID: PMC10341125 DOI: 10.3390/cells12131723] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
We assessed interactions between the astrocytic volume-regulated anion channel (VRAC) and aquaporin 4 (AQP4) in the supraoptic nucleus (SON). Acute SON slices and cultures of hypothalamic astrocytes prepared from rats received hyposmotic challenge (HOC) with/without VRAC or AQP4 blockers. In acute slices, HOC caused an early decrease with a late rebound in the neuronal firing rate of vasopressin neurons, which required activity of astrocytic AQP4 and VRAC. HOC also caused a persistent decrease in the excitatory postsynaptic current frequency, supported by VRAC and AQP4 activity in early HOC; late HOC required only VRAC activity. These events were associated with the dynamics of glial fibrillary acidic protein (GFAP) filaments, the late retraction of which was mediated by VRAC activity; this activity also mediated an HOC-evoked early increase in AQP4 expression and late subside in GFAP-AQP4 colocalization. AQP4 activity supported an early HOC-evoked increase in VRAC levels and its colocalization with GFAP. In cultured astrocytes, late HOC augmented VRAC currents, the activation of which depended on AQP4 pre-HOC/HOC activity. HOC caused an early increase in VRAC expression followed by a late rebound, requiring AQP4 and VRAC, or only AQP4 activity, respectively. Astrocytic swelling in early HOC depended on AQP4 activity, and so did the early extension of GFAP filaments. VRAC and AQP4 activity supported late regulatory volume decrease, the retraction of GFAP filaments, and subside in GFAP-VRAC colocalization. Taken together, astrocytic morphological plasticity relies on the coordinated activities of VRAC and AQP4, which are mutually regulated in the astrocytic mediation of HOC-evoked modulation of vasopressin neuronal activity.
Collapse
Affiliation(s)
- Yang Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Xiao-Ran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Yun-Hao Jiang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Tong Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
- Neuroscience Laboratory for Translational Medicine, School of Mental Health, Qiqihar Medical University, Qiqihar 161006, China
| | - Shuo Ling
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Hong-Yang Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Jia-Wei Yu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Shu-Wei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Xiao-Yu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Chun-Mei Hou
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| | - Vladimir Parpura
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China (H.-Y.W.)
| |
Collapse
|
12
|
Lee JM, Gadhe CG, Kang H, Pae AN, Lee CJ. Glutamate Permeability of Chicken Best1. Exp Neurobiol 2022; 31:277-288. [PMID: 36351838 PMCID: PMC9659495 DOI: 10.5607/en22038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 07/28/2023] Open
Abstract
Bestrophin-1 (Best1) is a calcium (Ca2+)-activated chloride (Cl-) channel which has a phylogenetically conserved channel structure with an aperture and neck in the ion-conducting pathway. Mammalian mouse Best1 (mBest1) has been known to have a permeability for large organic anions including gluconate, glutamate, and D-serine, in addition to several small monovalent anions, such as Cl‑, bromine (Br-), iodine (I-), and thiocyanate (SCN-). However, it is still unclear whether non-mammalian Best1 has a glutamate permeability through the ion-conducting pathway. Here, we report that chicken Best1 (cBest1) is permeable to glutamate in a Ca2+-dependent manner. The molecular docking and molecular dynamics simulation showed a glutamate binding at the aperture and neck of cBest1 and a glutamate permeation through the ion-conducting pore, respectively. Moreover, through electrophysiological recordings, we calculated the permeability ratio of glutamate to Cl- (PGlutamate/PCl) as 0.28 based on the reversal potential shift by ion substitution from Cl- to glutamate in the internal solution. Finally, we directly detected the Ca2+-dependent glutamate release through cBest1 using the ultrasensitive two-cell sniffer patch technique. Our results propose that Best1 homologs from non-mammalian (cBest1) to mammalian (mBest1) have a conserved permeability for glutamate.
Collapse
Affiliation(s)
- Jung Moo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | | | - Hyunji Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34113, Korea
| | - Ae Nim Pae
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
- KIST School, University of Science and Technology, Seoul 02792, Korea
| | - C. Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
13
|
TTYH family members form tetrameric complexes at the cell membrane. Commun Biol 2022; 5:886. [PMID: 36042377 PMCID: PMC9427776 DOI: 10.1038/s42003-022-03862-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 08/18/2022] [Indexed: 11/08/2022] Open
Abstract
The conserved Tweety homolog (TTYH) family consists of three paralogs in vertebrates, displaying a ubiquitous expression pattern. Although considered as ion channels for almost two decades, recent structural and functional analyses refuted this role. Intriguingly, while all paralogs shared a dimeric stoichiometry following detergent solubilization, their structures revealed divergence in their relative subunit orientation. Here, we determined the stoichiometry of intact mouse TTYH (mTTYH) complexes in cells. Using cross-linking and single-molecule fluorescence microscopy, we demonstrate that mTTYH1 and mTTYH3 form tetramers at the plasma membrane, stabilized by interactions between their extracellular domains. Using blue-native PAGE, fluorescence-detection size-exclusion chromatography, and hydrogen/deuterium exchange mass spectrometry (HDX-MS), we reveal that detergent solubilization results in tetramers destabilization, leading to their dissolution into dimers. Moreover, HDX-MS demonstrates that the extracellular domains are stabilized in the context of the tetrameric mTTYH complex. Together, our results expose the innate tetrameric organization of TTYH complexes at the cell membrane. Future structural analyses of these assemblies in native membranes are required to illuminate their long-sought cellular function.
Collapse
|
14
|
Brignone MS, Lanciotti A, Michelucci A, Mallozzi C, Camerini S, Catacuzzeno L, Sforna L, Caramia M, D’Adamo MC, Ceccarini M, Molinari P, Macioce P, Macchia G, Petrucci TC, Pessia M, Visentin S, Ambrosini E. The CaMKII/MLC1 Axis Confers Ca2+-Dependence to Volume-Regulated Anion Channels (VRAC) in Astrocytes. Cells 2022; 11:cells11172656. [PMID: 36078064 PMCID: PMC9454758 DOI: 10.3390/cells11172656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Astrocytes, the main glial cells of the central nervous system, play a key role in brain volume control due to their intimate contacts with cerebral blood vessels and the expression of a distinctive equipment of proteins involved in solute/water transport. Among these is MLC1, a protein highly expressed in perivascular astrocytes and whose mutations cause megalencephalic leukoencephalopathy with subcortical cysts (MLC), an incurable leukodystrophy characterized by macrocephaly, chronic brain edema, cysts, myelin vacuolation, and astrocyte swelling. Although, in astrocytes, MLC1 mutations are known to affect the swelling-activated chloride currents (ICl,swell) mediated by the volume-regulated anion channel (VRAC), and the regulatory volume decrease, MLC1′s proper function is still unknown. By combining molecular, biochemical, proteomic, electrophysiological, and imaging techniques, we here show that MLC1 is a Ca2+/Calmodulin-dependent protein kinase II (CaMKII) target protein, whose phosphorylation, occurring in response to intracellular Ca2+ release, potentiates VRAC-mediated ICl,swell. Overall, these findings reveal that MLC1 is a Ca2+-regulated protein, linking volume regulation to Ca2+ signaling in astrocytes. This knowledge provides new insight into the MLC1 protein function and into the mechanisms controlling ion/water exchanges in the brain, which may help identify possible molecular targets for the treatment of MLC and other pathological conditions caused by astrocyte swelling and brain edema.
Collapse
Affiliation(s)
| | - Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Antonio Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Serena Camerini
- Core Facilities (FAST), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Martino Caramia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Maria Cristina D’Adamo
- Department of Medicine and Surgery, LUM Giuseppe Degennaro University, 70010 Bari, Italy
| | - Marina Ceccarini
- National Centre for Rare Diseases, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Paola Molinari
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | | | | | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Sergio Visentin
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
- Correspondence: ; Tel.: +39-06-4990-2037
| |
Collapse
|
15
|
Wei Z, Zeng X, Lei Y, He H, Jamal M, Zhang C, Tan H, Xie S, Zhang Q. TTYH3, a potential prognosis biomarker associated with immune infiltration and immunotherapy response in lung cancer. Int Immunopharmacol 2022; 110:108999. [PMID: 35858518 DOI: 10.1016/j.intimp.2022.108999] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE The recognition of new diagnostic and prognostic biological markers for lung cancer is an essential and eager study. It's shown that ion channels play important roles in regulating various cellular processes and have been suggested to be associated with patient survival. However, tweety family member 3 (TTYH3), as a maxi-Cl- channel, its role in lung cancer remains elusive. METHODS The expression, diagnostic and prognostic efficacy of TTYH3 were analyzed by public databases and clinical samples. Cell functional experiments were used to explore the effects of TTYH3 on cell viability. GO and KEGG enrichment analysis revealed underlying pathways that TTYH3 and its co-expressed genes were enriched in. TIMER, TIDE and R language analyses were used to detect the correlation between TTYH3 and immune infiltration cell and immunotherapy response. RESULTS TTYH3 was up-regulated in lung cancer tissues compared to normal tissues and possessed a prominent diagnostic and prognostic value. TTYH3 knockdown significantly inhibited the proliferation of lung cancer cells. Enrichment analyses showed that TTYH3 and its co-expressed genes were mainly involved in immune related signaling pathways. Further investigation clarified that TTYH3 had a positive correlation with the infiltration of TAMs, Treg infiltration as well as T cell exhaustion and high TTYH3 expression indicated worse immunotherapy response and shorter survival after immune checkpoint blockade treatment. CONCLUSION This study not only revealed the diagnostic and prognostic value of TTYH3 but also provided TTYH3-based estimation of immunotherapy response for lung cancer patients, which might provide new strategies like anti-TTYH3 combined with immune therapy for the treatment of lung cancer.
Collapse
Affiliation(s)
- Zimeng Wei
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xingruo Zeng
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yufei Lei
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hengjing He
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Muhammad Jamal
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chengjie Zhang
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Haiyan Tan
- Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiuping Zhang
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, China.
| |
Collapse
|
16
|
Wang Y, Xie Y, Dong B, Xue W, Chen S, Mitsuo S, Zou H, Feng Y, Ma K, Dong Q, Cao J, Zhu C. The TTYH3/MK5 Positive Feedback Loop regulates Tumor Progression via GSK3-β/β-catenin signaling in HCC. Int J Biol Sci 2022; 18:4053-4070. [PMID: 35844789 PMCID: PMC9274494 DOI: 10.7150/ijbs.73009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/05/2022] [Indexed: 12/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide, and identification of novel targets is necessary for its diagnosis and treatment. This study aimed to investigate the biological function and clinical significance of tweety homolog 3 (TTYH3) in HCC. TTYH3 overexpression promoted cell proliferation, migration, and invasion and inhibited HCCM3 and Hep3B cell apoptosis. TTYH3 promoted tumor formation and metastasis in vivo. TTYH3 upregulated calcium influx and intracellular chloride concentration, thereby promoting cellular migration and regulating epithelial-mesenchymal transition-related protein expression. The interaction between TTYH3 and MK5 was identified through co-immunoprecipitation assays and protein docking. TTYH3 promoted the expression of MK5, which then activated the GSK3β/β-catenin signaling pathway. MK5 knockdown attenuated the activation of GSK3β/β-catenin signaling by TTYH3. TTYH3 expression was regulated in a positive feedback manner. In clinical HCC samples, TTYH3 was upregulated in the HCC tissues compared to nontumor tissues. Furthermore, high TTYH3 expression was significantly correlated with poor patient survival. The CpG islands were hypomethylated in the promoter region of TTYH3 in HCC tissues. In conclusion, we identified TTYH3 regulates tumor development and progression via MK5/GSK3-β/β-catenin signaling in HCC and promotes itself expression in a positive feedback loop.
Collapse
Affiliation(s)
- Yixiu Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Yuwei Xie
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Bingzi Dong
- Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Weijie Xue
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Shuhai Chen
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Shimada Mitsuo
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Hao Zou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Yujie Feng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Kai Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Qian Dong
- Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, China
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Chengzhan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
- Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, China
| |
Collapse
|
17
|
Koh W, Park M, Chun YE, Lee J, Shim HS, Park MG, Kim S, Sa M, Joo J, Kang H, Oh SJ, Woo J, Chun H, Lee SE, Hong J, Feng J, Li Y, Ryu H, Cho J, Lee CJ. Astrocytes Render Memory Flexible by Releasing D-Serine and Regulating NMDA Receptor Tone in the Hippocampus. Biol Psychiatry 2022; 91:740-752. [PMID: 34952697 DOI: 10.1016/j.biopsych.2021.10.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND NMDA receptor (NMDAR) hypofunction has been implicated in several psychiatric disorders with impairment of cognitive flexibility. However, the molecular mechanism of how NMDAR hypofunction with decreased NMDAR tone causes the impairment of cognitive flexibility has been minimally understood. Furthermore, it has been unclear whether hippocampal astrocytes regulate NMDAR tone and cognitive flexibility. METHODS We employed cell type-specific genetic manipulations, ex vivo electrophysiological recordings, sniffer patch recordings, cutting-edge biosensor for norepinephrine, and behavioral assays to investigate whether astrocytes can regulate NMDAR tone by releasing D-serine and glutamate. Subsequently, we further investigated the role of NMDAR tone in heterosynaptic long-term depression, metaplasticity, and cognitive flexibility. RESULTS We found that hippocampal astrocytes regulate NMDAR tone via BEST1-mediated corelease of D-serine and glutamate. Best1 knockout mice exhibited reduced NMDAR tone and impairments of homosynaptic and α1 adrenergic receptor-dependent heterosynaptic long-term depression, which leads to defects in metaplasticity and cognitive flexibility. These impairments in Best1 knockout mice can be rescued by hippocampal astrocyte-specific BEST1 expression or enhanced NMDAR tone through D-serine supplement. D-serine injection in Best1 knockout mice during initial learning rescues subsequent reversal learning. CONCLUSIONS These findings indicate that NMDAR tone during initial learning is important for subsequent learning, and hippocampal NMDAR tone regulated by astrocytic BEST1 is critical for heterosynaptic long-term depression, metaplasticity, and cognitive flexibility.
Collapse
Affiliation(s)
- Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; Department of Neuroscience, Division of BioMedical Science & Technology, Korea Institute of Science and Technology School, Korea University of Science and Technology, Seoul, South Korea; Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Mijeong Park
- Department of Neuroscience, Division of BioMedical Science & Technology, Korea Institute of Science and Technology School, Korea University of Science and Technology, Seoul, South Korea; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Ye Eun Chun
- Department of Neuroscience, Division of BioMedical Science & Technology, Korea Institute of Science and Technology School, Korea University of Science and Technology, Seoul, South Korea; Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jaekwang Lee
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Hyun Soo Shim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Sunpil Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea; Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Jinhyeong Joo
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; IBS School, Korea University of Science and Technology, Daejeon, South Korea
| | - Hyunji Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; IBS School, Korea University of Science and Technology, Daejeon, South Korea
| | - Soo-Jin Oh
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Junsung Woo
- Department of Neuroscience, Division of BioMedical Science & Technology, Korea Institute of Science and Technology School, Korea University of Science and Technology, Seoul, South Korea; Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Heejung Chun
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea; Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jinpyo Hong
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Hoon Ryu
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jeiwon Cho
- Brain and Cognitive Science, Scranton College, Ewha Womans University, Seoul, South Korea
| | - C Justin Lee
- Department of Neuroscience, Division of BioMedical Science & Technology, Korea Institute of Science and Technology School, Korea University of Science and Technology, Seoul, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea; IBS School, Korea University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
18
|
Kolobkova Y, Pervaiz S, Stauber T. The expanding toolbox to study the LRRC8-formed volume-regulated anion channel VRAC. CURRENT TOPICS IN MEMBRANES 2021; 88:119-163. [PMID: 34862024 DOI: 10.1016/bs.ctm.2021.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The volume-regulated anion channel (VRAC) is activated upon cell swelling and facilitates the passive movement of anions across the plasma membrane in cells. VRAC function underlies many critical homeostatic processes in vertebrate cells. Among them are the regulation of cell volume and membrane potential, glutamate release and apoptosis. VRAC is also permeable for organic osmolytes and metabolites including some anti-cancer drugs and antibiotics. Therefore, a fundamental understanding of VRAC's structure-function relationships, its physiological roles, its utility for therapy of diseases, and the development of compounds modulating its activity are important research frontiers. Here, we describe approaches that have been applied to study VRAC since it was first described more than 30 years ago, providing an overview of the recent methodological progress. The diverse applications reflecting a compromise between the physiological situation, biochemical definition, and biophysical resolution range from the study of VRAC activity using a classic electrophysiology approach, to the measurement of osmolytes transport by various means and the investigation of its activation using a novel biophysical approach based on fluorescence resonance energy transfer.
Collapse
Affiliation(s)
- Yulia Kolobkova
- Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, Germany
| | - Sumaira Pervaiz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Tobias Stauber
- Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany.
| |
Collapse
|
19
|
Structures of tweety homolog proteins TTYH2 and TTYH3 reveal a Ca 2+-dependent switch from intra- to intermembrane dimerization. Nat Commun 2021; 12:6913. [PMID: 34824283 PMCID: PMC8617170 DOI: 10.1038/s41467-021-27283-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023] Open
Abstract
Tweety homologs (TTYHs) comprise a conserved family of transmembrane proteins found in eukaryotes with three members (TTYH1-3) in vertebrates. They are widely expressed in mammals including at high levels in the nervous system and have been implicated in cancers and other diseases including epilepsy, chronic pain, and viral infections. TTYHs have been reported to form Ca2+- and cell volume-regulated anion channels structurally distinct from any characterized protein family with potential roles in cell adhesion, migration, and developmental signaling. To provide insight into TTYH family structure and function, we determined cryo-EM structures of Mus musculus TTYH2 and TTYH3 in lipid nanodiscs. TTYH2 and TTYH3 adopt a previously unobserved fold which includes an extended extracellular domain with a partially solvent exposed pocket that may be an interaction site for hydrophobic molecules. In the presence of Ca2+, TTYH2 and TTYH3 form homomeric cis-dimers bridged by extracellularly coordinated Ca2+. Strikingly, in the absence of Ca2+, TTYH2 forms trans-dimers that span opposing membranes across a ~130 Å intermembrane space as well as a monomeric state. All TTYH structures lack ion conducting pathways and we do not observe TTYH2-dependent channel activity in cells. We conclude TTYHs are not pore forming subunits of anion channels and their function may involve Ca2+-dependent changes in quaternary structure, interactions with hydrophobic molecules near the extracellular membrane surface, and/or association with additional protein partners.
Collapse
|
20
|
Cao Y, Wu HN, Cao XL, Yue KY, Han WJ, Cao ZP, Zhang YF, Gao XY, Luo C, Jiang XF, Han H, Zheng MH. Transmembrane Protein Ttyh1 Maintains the Quiescence of Neural Stem Cells Through Ca 2+/NFATc3 Signaling. Front Cell Dev Biol 2021; 9:779373. [PMID: 34869383 PMCID: PMC8635056 DOI: 10.3389/fcell.2021.779373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
The quiescence, activation, and subsequent neurogenesis of neural stem cells (NSCs) play essential roles in the physiological homeostasis and pathological repair of the central nervous system. Previous studies indicate that transmembrane protein Ttyh1 is required for the stemness of NSCs, whereas the exact functions in vivo and precise mechanisms are still waiting to be elucidated. By constructing Ttyh1-promoter driven reporter mice, we determined the specific expression of Ttyh1 in quiescent NSCs and niche astrocytes. Further evaluations on Ttyh1 knockout mice revealed that Ttyh1 ablation leads to activated neurogenesis and enhanced spatial learning and memory in adult mice (6-8 weeks). Correspondingly, Ttyh1 deficiency results in accelerated exhaustion of NSC pool and impaired neurogenesis in aged mice (12 months). By RNA-sequencing, bioinformatics and molecular biological analysis, we found that Ttyh1 is involved in the regulation of calcium signaling in NSCs, and transcription factor NFATc3 is a critical effector in quiescence versus cell cycle entry regulated by Ttyh1. Our research uncovered new endogenous mechanisms that regulate quiescence versus activation of NSCs, therefore provide novel targets for the intervention to activate quiescent NSCs to participate in injury repair during pathology and aging.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hai-ning Wu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Xiu-li Cao
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Kang-yi Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wen-juan Han
- Department of Neurobiology, Fourth Military Medical University, Xi’an, China
| | - Zi-peng Cao
- Department of Occupational and Environmental Health, Fourth Military Medical University, Xi’an, China
| | - Yu-fei Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Xiang-yu Gao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ceng Luo
- Department of Neurobiology, Fourth Military Medical University, Xi’an, China
| | - Xiao-fan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Min-hua Zheng
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
21
|
Gorniak-Walas M, Nizinska K, Lukasiuk K. Cloning and Functional Analysis of Rat Tweety-Homolog 1 Gene Promoter. Neurochem Res 2021; 46:2463-2472. [PMID: 34173119 PMCID: PMC8302521 DOI: 10.1007/s11064-021-03374-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 11/06/2022]
Abstract
Tweety-homolog 1 protein (Ttyh1) is abundantly expressed in neurons in the healthy brain, and its expression is induced under pathological conditions. In hippocampal neurons in vitro, Ttyh1 was implicated in the regulation of primary neuron morphology. However, the mechanisms that underlie transcriptional regulation of the Ttyh1 gene in neurons remain elusive. The present study sought to identify the promoter of the Ttyh1 gene and functionally characterize cis-regulatory elements that are potentially involved in the transcriptional regulation of Ttyh1 expression in rat dissociated hippocampal neurons in vitro. We cloned a 592 bp rat Ttyh1 promoter sequence and designed deletion constructs of the transcription factors specificity protein 1 (Sp1), E2F transcription factor 3 (E2f3), and achaete-scute homolog 1 (Ascl1) that were fused upstream of a luciferase reporter gene in pGL4.10[luc2]. The luciferase reporter gene assay showed the possible involvement of Ascl1, Sp1, and responsive cis-regulatory elements in Ttyh1 expression. These findings provide novel information about Ttyh1 gene regulation in neurons.
Collapse
Affiliation(s)
- Malgorzata Gorniak-Walas
- Laboratory of Epileptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Karolina Nizinska
- Laboratory of Epileptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Katarzyna Lukasiuk
- Laboratory of Epileptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| |
Collapse
|
22
|
Sukalskaia A, Straub MS, Deneka D, Sawicka M, Dutzler R. Cryo-EM structures of the TTYH family reveal a novel architecture for lipid interactions. Nat Commun 2021; 12:4893. [PMID: 34385445 PMCID: PMC8361169 DOI: 10.1038/s41467-021-25106-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022] Open
Abstract
The Tweety homologs (TTYHs) are members of a conserved family of eukaryotic membrane proteins that are abundant in the brain. The three human paralogs were assigned to function as anion channels that are either activated by Ca2+ or cell swelling. To uncover their unknown architecture and its relationship to function, we have determined the structures of human TTYH1–3 by cryo-electron microscopy. All structures display equivalent features of a dimeric membrane protein that contains five transmembrane segments and an extended extracellular domain. As none of the proteins shows attributes reminiscent of an anion channel, we revisited functional experiments and did not find any indication of ion conduction. Instead, we find density in an extended hydrophobic pocket contained in the extracellular domain that emerges from the lipid bilayer, which suggests a role of TTYH proteins in the interaction with lipid-like compounds residing in the membrane. The human Tweety homologue (TTYH) family of transmembrane proteins have been suggested to act as chloride channels. Here the authors present cryo-EM structures of the 3 human TTYH paralogs that do not display the expected features of an anion channel, and instead appear to interact with lipid-like compounds residing in the membrane; suggesting an involvement in lipid-associated processes.
Collapse
Affiliation(s)
| | - Monique S Straub
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Dawid Deneka
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Marta Sawicka
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| | - Raimund Dutzler
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Attwood MM, Schiöth HB. Characterization of Five Transmembrane Proteins: With Focus on the Tweety, Sideroflexin, and YIP1 Domain Families. Front Cell Dev Biol 2021; 9:708754. [PMID: 34350187 PMCID: PMC8327215 DOI: 10.3389/fcell.2021.708754] [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: 05/12/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
Transmembrane proteins are involved in many essential cell processes such as signal transduction, transport, and protein trafficking, and hence many are implicated in different disease pathways. Further, as the structure and function of proteins are correlated, investigating a group of proteins with the same tertiary structure, i.e., the same number of transmembrane regions, may give understanding about their functional roles and potential as therapeutic targets. This analysis investigates the previously unstudied group of proteins with five transmembrane-spanning regions (5TM). More than half of the 58 proteins identified with the 5TM architecture belong to 12 families with two or more members. Interestingly, more than half the proteins in the dataset function in localization activities through movement or tethering of cell components and more than one-third are involved in transport activities, particularly in the mitochondria. Surprisingly, no receptor activity was identified within this dataset in large contrast with other TM groups. The three major 5TM families, which comprise nearly 30% of the dataset, include the tweety family, the sideroflexin family and the Yip1 domain (YIPF) family. We also analyzed the evolutionary origin of these three families. The YIPF family appears to be the most ancient with presence in bacteria and archaea, while the tweety and sideroflexin families are first found in eukaryotes. We found no evidence of common decent for these three families. About 30% of the 5TM proteins have prominent expression in the brain, liver, or testis. Importantly, 60% of these proteins are identified as cancer prognostic markers, where they are associated with clinical outcomes of various tumor types. Nearly 10% of the 5TMs are still not fully characterized and further investigation of their functional activities and expression is warranted. This study provides the first comprehensive analysis of proteins with the 5TM architecture, providing details of their unique characteristics.
Collapse
Affiliation(s)
- Misty M Attwood
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
24
|
Nalamalapu RR, Yue M, Stone AR, Murphy S, Saha MS. The tweety Gene Family: From Embryo to Disease. Front Mol Neurosci 2021; 14:672511. [PMID: 34262434 PMCID: PMC8273234 DOI: 10.3389/fnmol.2021.672511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
The tweety genes encode gated chloride channels that are found in animals, plants, and even simple eukaryotes, signifying their deep evolutionary origin. In vertebrates, the tweety gene family is highly conserved and consists of three members—ttyh1, ttyh2, and ttyh3—that are important for the regulation of cell volume. While research has elucidated potential physiological functions of ttyh1 in neural stem cell maintenance, proliferation, and filopodia formation during neural development, the roles of ttyh2 and ttyh3 are less characterized, though their expression patterns during embryonic and fetal development suggest potential roles in the development of a wide range of tissues including a role in the immune system in response to pathogen-associated molecules. Additionally, members of the tweety gene family have been implicated in various pathologies including cancers, particularly pediatric brain tumors, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease. Here, we review the current state of research using information from published articles and open-source databases on the tweety gene family with regard to its structure, evolution, expression during development and adulthood, biochemical and cellular functions, and role in human disease. We also identify promising areas for further research to advance our understanding of this important, yet still understudied, family of genes.
Collapse
Affiliation(s)
- Rithvik R Nalamalapu
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Michelle Yue
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Aaron R Stone
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Samantha Murphy
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Margaret S Saha
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| |
Collapse
|
25
|
Liu Y, Xu Y, Jiang W, Ji H, Wang ZW, Zhu X. Discovery of key genes as novel biomarkers specifically associated with HPV-negative cervical cancer. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:492-506. [PMID: 33997099 PMCID: PMC8091489 DOI: 10.1016/j.omtm.2021.03.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cervical cancer is a common female malignancy that is mainly caused by human papillomavirus (HPV) infection. However, the incidence of HPV-negative cervical cancer has shown an increasing trend in recent years. Because the mechanism of HPV-negative cervical cancer development is unclear, this study aims to find the pattern of differential gene expression in HPV-negative cervical cancer and verify the underlying potential mechanism. Differentially expressed genes were compared among HPV-positive cervical cancer, HPV-negative cervical cancer, and normal cervical tissues retrieved from TCGA. Subsequently, dysregulated differentially expressed genes specifically existed in HPV-negative cervical cancer tissues and HPV-negative cell lines were validated by qRT-PCR, western blotting, and immunohistochemical staining. We found seventeen highly expressed genes that were particularly associated with HPV-negative cervical cancer from analysis of TCGA database. Among the 17 novel genes, 7 genes (preferentially expressed antigen in melanoma [PRAME], HMGA2, ETS variant 4 [ETV4], MEX3A, TM7SF2, SLC19A1, and tweety-homologs 3 [TTYH3]) displayed significantly elevated expression in HPV-negative cervical cancer cells and HPV-negative cervical cancer tissues. Additionally, higher expression of MEX3A and TTYH3 was associated with a shorter overall survival of patients with HPV-negative cervical cancer. Our study implies that these seven genes are more likely to provide novel insights into the occurrence and progression of HPV-negative cervical cancer.
Collapse
Affiliation(s)
- Yi Liu
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yichi Xu
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenxiao Jiang
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huihui Ji
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhi-Wei Wang
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xueqiong Zhu
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
26
|
Okada Y, Sabirov RZ, Sato-Numata K, Numata T. Cell Death Induction and Protection by Activation of Ubiquitously Expressed Anion/Cation Channels. Part 1: Roles of VSOR/VRAC in Cell Volume Regulation, Release of Double-Edged Signals and Apoptotic/Necrotic Cell Death. Front Cell Dev Biol 2021; 8:614040. [PMID: 33511120 PMCID: PMC7835517 DOI: 10.3389/fcell.2020.614040] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022] Open
Abstract
Cell volume regulation (CVR) is essential for survival and functions of animal cells. Actually, normotonic cell shrinkage and swelling are coupled to apoptotic and necrotic cell death and thus called the apoptotic volume decrease (AVD) and the necrotic volume increase (NVI), respectively. A number of ubiquitously expressed anion and cation channels are involved not only in CVD but also in cell death induction. This series of review articles address the question how cell death is induced or protected with using ubiquitously expressed ion channels such as swelling-activated anion channels, acid-activated anion channels and several types of TRP cation channels including TRPM2 and TRPM7. The Part 1 focuses on the roles of the volume-sensitive outwardly rectifying anion channels (VSOR), also called the volume-regulated anion channel (VRAC), which is activated by cell swelling or reactive oxygen species (ROS) in a manner dependent on intracellular ATP. First we describe phenotypical properties, the molecular identity, and physical pore dimensions of VSOR/VRAC. Second, we highlight the roles of VSOR/VRAC in the release of organic signaling molecules, such as glutamate, glutathione, ATP and cGAMP, that play roles as double-edged swords in cell survival. Third, we discuss how VSOR/VRAC is involved in CVR and cell volume dysregulation as well as in the induction of or protection from apoptosis, necrosis and regulated necrosis under pathophysiological conditions.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences, Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ravshan Z. Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Japan Society for the Promotion of Science, Tokyo, Japan
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tomohiro Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
27
|
Tweety-Homolog 1 Facilitates Pain via Enhancement of Nociceptor Excitability and Spinal Synaptic Transmission. Neurosci Bull 2020; 37:478-496. [PMID: 33355899 DOI: 10.1007/s12264-020-00617-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Tweety-homolog 1 (Ttyh1) is expressed in neural tissue and has been implicated in the generation of several brain diseases. However, its functional significance in pain processing is not understood. By disrupting the gene encoding Ttyh1, we found a loss of Ttyh1 in nociceptors and their central terminals in Ttyh1-deficient mice, along with a reduction in nociceptor excitability and synaptic transmission at identified synapses between nociceptors and spinal neurons projecting to the periaqueductal grey (PAG) in the basal state. More importantly, the peripheral inflammation-evoked nociceptor hyperexcitability and spinal synaptic potentiation recorded in spinal-PAG projection neurons were compromised in Ttyh1-deficient mice. Analysis of the paired-pulse ratio and miniature excitatory postsynaptic currents indicated a role of presynaptic Ttyh1 from spinal nociceptor terminals in the regulation of neurotransmitter release. Interfering with Ttyh1 specifically in nociceptors produces a comparable pain relief. Thus, in this study we demonstrated that Ttyh1 is a critical determinant of acute nociception and pain sensitization caused by peripheral inflammation.
Collapse
|
28
|
Kwak H, Koh W, Kim S, Song K, Shin JI, Lee JM, Lee EH, Bae JY, Ha GE, Oh JE, Park YM, Kim S, Feng J, Lee SE, Choi JW, Kim KH, Kim YS, Woo J, Lee D, Son T, Kwon SW, Park KD, Yoon BE, Lee J, Li Y, Lee H, Bae YC, Lee CJ, Cheong E. Astrocytes Control Sensory Acuity via Tonic Inhibition in the Thalamus. Neuron 2020; 108:691-706.e10. [PMID: 32905785 DOI: 10.1016/j.neuron.2020.08.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/05/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022]
Abstract
Sensory discrimination is essential for survival. However, how sensory information is finely controlled in the brain is not well defined. Here, we show that astrocytes control tactile acuity via tonic inhibition in the thalamus. Mechanistically, diamine oxidase (DAO) and the subsequent aldehyde dehydrogenase 1a1 (Aldh1a1) convert putrescine into GABA, which is released via Best1. The GABA from astrocytes inhibits synaptically evoked firing at the lemniscal synapses to fine-tune the dynamic range of the stimulation-response relationship, the precision of spike timing, and tactile discrimination. Our findings reveal a novel role of astrocytes in the control of sensory acuity through tonic GABA release.
Collapse
Affiliation(s)
- Hankyul Kwak
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Wuhyun Koh
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea
| | - Sangwoo Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Kiyeong Song
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jeong-Im Shin
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jung Moo Lee
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea
| | - Elliot H Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41566, South Korea
| | - Go Eun Ha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Ju-Eun Oh
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yongmin Mason Park
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea
| | - Sunpil Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Ji Won Choi
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Ki Hun Kim
- Doping Control Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, College of Natural Science, Dankook University, Cheonan 31116, South Korea
| | - Junsung Woo
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Taehwang Son
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, South Korea
| | - Soon Woo Kwon
- Radiation Medicine Clinical Research Division, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Ki Duk Park
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, South Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, College of Natural Science, Dankook University, Cheonan 31116, South Korea
| | - Jaeick Lee
- Doping Control Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Hyunbeom Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41566, South Korea
| | - C Justin Lee
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea.
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea; POSTECH Biotech Center, POSTECH, Pohang, South Korea.
| |
Collapse
|
29
|
Lambo S, von Hoff K, Korshunov A, Pfister SM, Kool M. ETMR: a tumor entity in its infancy. Acta Neuropathol 2020; 140:249-266. [PMID: 32601913 PMCID: PMC7423804 DOI: 10.1007/s00401-020-02182-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/04/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Abstract
Embryonal tumor with Multilayered Rosettes (ETMR) is a relatively rare but typically deadly type of brain tumor that occurs mostly in infants. Since the discovery of the characteristic chromosome 19 miRNA cluster (C19MC) amplification a decade ago, the methods for diagnosing this entity have improved and many new insights in the molecular landscape of ETMRs have been acquired. All ETMRs, despite their highly heterogeneous histology, are characterized by specific high expression of the RNA-binding protein LIN28A, which is, therefore, often used as a diagnostic marker for these tumors. ETMRs have few recurrent genetic aberrations, mainly affecting the miRNA pathway and including amplification of C19MC (embryonal tumor with multilayered rosettes, C19MC-altered) and mutually exclusive biallelic DICER1 mutations of which the first hit is typically inherited through the germline (embryonal tumor with multilayered rosettes, DICER1-altered). Identification of downstream pathways affected by the deregulated miRNA machinery has led to several proposed potential therapeutical vulnerabilities including targeting the WNT, SHH, or mTOR pathways, MYCN or chromosomal instability. However, despite those findings, treatment outcomes have only marginally improved, since the initial description of this tumor entity. Many patients do not survive longer than a year after diagnosis and the 5-year overall survival rate is still lower than 30%. Thus, there is an urgent need to translate the new insights in ETMR biology into more effective treatments. Here, we present an overview of clinical and molecular characteristics of ETMRs and the current progress on potential targeted therapies.
Collapse
Affiliation(s)
- Sander Lambo
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Katja von Hoff
- Department of Pediatric Oncology/Hematology, Charité University Medicine, Berlin, Germany
| | - Andrey Korshunov
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Volume-regulated anion channel as a novel cancer therapeutic target. Int J Biol Macromol 2020; 159:570-576. [DOI: 10.1016/j.ijbiomac.2020.05.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/23/2020] [Accepted: 05/17/2020] [Indexed: 01/01/2023]
|
31
|
Woo J, Jang MW, Lee J, Koh W, Mikoshiba K, Lee CJ. The molecular mechanism of synaptic activity-induced astrocytic volume transient. J Physiol 2020; 598:4555-4572. [PMID: 32706443 DOI: 10.1113/jp279741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Neuronal activity causes astrocytic volume change via K+ uptake through TREK-1 containing two-pore domain potassium channels. The volume transient is terminated by Cl- efflux through the Ca2+ -activated anion channel BEST1. The source of the Ca2+ required to open BEST1 appears to be the stretch-activated TRPA1 channel. Intense neuronal activity is synaptically coupled with a physical change in astrocytes via volume transients. ABSTRACT The brain volume changes dynamically and transiently upon intense neuronal activity through a tight regulation of ion concentrations and water movement across the plasma membrane of astrocytes. We have recently demonstrated that an intense neuronal activity and subsequent astrocytic AQP4-dependent volume transient are critical for synaptic plasticity and memory. We have also pharmacologically demonstrated a functional coupling between synaptic activity and the astrocytic volume transient. However, the precise molecular mechanisms of how intense neuronal activity and the astrocytic volume transient are coupled remain unclear. Here we utilized an intrinsic optical signal imaging technique combined with fluorescence imaging using ion sensitive dyes and molecular probes and electrophysiology to investigate the detailed molecular mechanisms in genetically modified mice. We report that a brief synaptic activity induced by a train stimulation (20 Hz, 1 s) causes a prolonged astrocytic volume transient (80 s) via K+ uptake through TREK-1 containing two-pore domain potassium (K2P) channels, but not Kir4.1 or NKCC1. This volume change is terminated by Cl- efflux through the Ca2+ -activated anion channel BEST1, but not the volume-regulated anion channel TTYH. The source of the Ca2+ required to open BEST1 appears to be the stretch-activated TRPA1 channel in astrocytes, but not IP3 R2. In summary, our study identifies several important astrocytic ion channels (AQP4, TREK-1, BEST1, TRPA1) as the key molecules leading to the neuronal activity-dependent volume transient in astrocytes. Our findings reveal new molecular and cellular mechanisms for the synaptic coupling of intense neuronal activity with a physical change in astrocytes via volume transients.
Collapse
Affiliation(s)
- Junsung Woo
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Minwoo Wendy Jang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Jaekwang Lee
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.,Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Biology, RIKEN Brain Science Institute, Hirosawa 2-1, Wako, Saitama, 351-0198, Japan
| | - C Justin Lee
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.,Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| |
Collapse
|
32
|
Wilke BU, Kummer KK, Leitner MG, Kress M. Chloride - The Underrated Ion in Nociceptors. Front Neurosci 2020; 14:287. [PMID: 32322187 PMCID: PMC7158864 DOI: 10.3389/fnins.2020.00287] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/12/2020] [Indexed: 01/06/2023] Open
Abstract
In contrast to pain processing neurons in the spinal cord, where the importance of chloride conductances is already well established, chloride homeostasis in primary afferent neurons has received less attention. Sensory neurons maintain high intracellular chloride concentrations through balanced activity of Na+-K+-2Cl- cotransporter 1 (NKCC1) and K+-Cl- cotransporter 2 (KCC2). Whereas in other cell types activation of chloride conductances causes hyperpolarization, activation of the same conductances in primary afferent neurons may lead to inhibitory or excitatory depolarization depending on the actual chloride reversal potential and the total amount of chloride efflux during channel or transporter activation. Dorsal root ganglion (DRG) neurons express a multitude of chloride channel types belonging to different channel families, such as ligand-gated, ionotropic γ-aminobutyric acid (GABA) or glycine receptors, Ca2+-activated chloride channels of the anoctamin/TMEM16, bestrophin or tweety-homolog family, CLC chloride channels and transporters, cystic fibrosis transmembrane conductance regulator (CFTR) as well as volume-regulated anion channels (VRACs). Specific chloride conductances are involved in signal transduction and amplification at the peripheral nerve terminal, contribute to excitability and action potential generation of sensory neurons, or crucially shape synaptic transmission in the spinal dorsal horn. In addition, chloride channels can be modified by a plethora of inflammatory mediators affecting them directly, via protein-protein interaction, or through signaling cascades. Since chloride channels as well as mediators that modulate chloride fluxes are regulated in pain disorders and contribute to nociceptor excitation and sensitization it is timely and important to emphasize their critical role in nociceptive primary afferents in this review.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
33
|
Barabas P, Augustine J, Fernández JA, McGeown JG, McGahon MK, Curtis TM. Ion channels and myogenic activity in retinal arterioles. CURRENT TOPICS IN MEMBRANES 2020; 85:187-226. [PMID: 32402639 DOI: 10.1016/bs.ctm.2020.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Retinal pressure autoregulation is an important mechanism that protects the retina by stabilizing retinal blood flow during changes in arterial or intraocular pressure. Similar to other vascular beds, retinal pressure autoregulation is thought to be mediated largely through the myogenic response of small arteries and arterioles which constrict when transmural pressure increases or dilate when it decreases. Over recent years, we and others have investigated the signaling pathways underlying the myogenic response in retinal arterioles, with particular emphasis on the involvement of different ion channels expressed in the smooth muscle layer of these vessels. Here, we review and extend previous work on the expression and spatial distribution of the plasma membrane and sarcoplasmic reticulum ion channels present in retinal vascular smooth muscle cells (VSMCs) and discuss their contribution to pressure-induced myogenic tone in retinal arterioles. This includes new data demonstrating that several key players and modulators of the myogenic response show distinctively heterogeneous expression along the length of the retinal arteriolar network, suggesting differences in myogenic signaling between larger and smaller pre-capillary arterioles. Our immunohistochemical investigations have also highlighted the presence of actin-containing microstructures called myobridges that connect the retinal VSMCs to one another. Although further work is still needed, studies to date investigating myogenic mechanisms in the retina have contributed to a better understanding of how blood flow is regulated in this tissue. They also provide a basis to direct future research into retinal diseases where blood flow changes contribute to the pathology.
Collapse
Affiliation(s)
- Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - José A Fernández
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Mary K McGahon
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom.
| |
Collapse
|
34
|
Kushwah N, Jain V, Yadav D. Osmolytes: A Possible Therapeutic Molecule for Ameliorating the Neurodegeneration Caused by Protein Misfolding and Aggregation. Biomolecules 2020; 10:biom10010132. [PMID: 31941036 PMCID: PMC7022651 DOI: 10.3390/biom10010132] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 12/20/2022] Open
Abstract
Most of the neurological disorders in the brain are caused by the abnormal buildup of misfolded or aggregated proteins. Osmolytes are low molecular weight organic molecules usually built up in tissues at a quite high amount during stress or any pathological condition. These molecules help in providing stability to the aggregated proteins and protect these proteins from misfolding. Alzheimer’s disease (AD) is the uttermost universal neurological disorder that can be described by the deposition of neurofibrillary tangles, aggregated/misfolded protein produced by the amyloid β-protein (Aβ). Osmolytes provide stability to the folded, functional form of a protein and alter the folding balance away from aggregation and/or degradation of the protein. Moreover, they are identified as chemical chaperones. Brain osmolytes enhance the pace of Aβ aggregation, combine with the nearby water molecules more promptly, and avert the aggregation/misfolding of proteins by providing stability to them. Therefore, osmolytes can be employed as therapeutic targets and may assist in potential drug design for many neurodegenerative and other diseases.
Collapse
Affiliation(s)
- Neetu Kushwah
- Functional Genomics Unit, CSIR-institute of genomics and integrative biology, Mall road, Delhi 110007, India;
| | - Vishal Jain
- Department of ECE, Carnegie Mellon University, Pittsburgh, PA 15289, USA
- Correspondence: (V.J.); (D.Y.); Tel.: +1-412-251-3042 (V.J.); +82-10-2202-1191(D.Y.)
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Korea
- Correspondence: (V.J.); (D.Y.); Tel.: +1-412-251-3042 (V.J.); +82-10-2202-1191(D.Y.)
| |
Collapse
|
35
|
Nikolic L, Nobili P, Shen W, Audinat E. Role of astrocyte purinergic signaling in epilepsy. Glia 2019; 68:1677-1691. [DOI: 10.1002/glia.23747] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Ljiljana Nikolic
- Institute for Biological Research Siniša Stanković, University of Belgrade Serbia
| | | | - Weida Shen
- Zhejiang University City College Zhejiang Hangzhou China
| | - Etienne Audinat
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM Montpellier France
| |
Collapse
|
36
|
Weinberg F, Griffin R, Fröhlich M, Heining C, Braun S, Spohr C, Iconomou M, Hollek V, Röring M, Horak P, Kreutzfeldt S, Warsow G, Hutter B, Uhrig S, Neumann O, Reuss D, Heiland DH, von Kalle C, Weichert W, Stenzinger A, Brors B, Glimm H, Fröhling S, Brummer T. Identification and characterization of a BRAF fusion oncoprotein with retained autoinhibitory domains. Oncogene 2019; 39:814-832. [PMID: 31558800 DOI: 10.1038/s41388-019-1021-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Fusion proteins involving the BRAF serine/threonine kinase occur in many cancers. The oncogenic potential of BRAF fusions has been attributed to the loss of critical N-terminal domains that mediate BRAF autoinhibition. We used whole-exome and RNA sequencing in a patient with glioblastoma multiforme to identify a rearrangement between TTYH3, encoding a membrane-resident, calcium-activated chloride channel, and BRAF intron 1, resulting in a TTYH3-BRAF fusion protein that retained all features essential for BRAF autoinhibition. Accordingly, the BRAF moiety of the fusion protein alone, which represents full-length BRAF without the amino acids encoded by exon 1 (BRAFΔE1), did not induce MEK/ERK phosphorylation or transformation. Likewise, neither the TTYH3 moiety of the fusion protein nor full-length TTYH3 provoked ERK pathway activity or transformation. In contrast, TTYH3-BRAF displayed increased MEK phosphorylation potential and transforming activity, which were caused by TTYH3-mediated tethering of near-full-length BRAF to the (endo)membrane system. Consistent with this mechanism, a synthetic approach, in which BRAFΔE1 was tethered to the membrane by fusing it to the cytoplasmic tail of CD8 also induced transformation. Furthermore, we demonstrate that TTYH3-BRAF signals largely independent of a functional RAS binding domain, but requires an intact BRAF dimer interface and activation loop phosphorylation sites. Cells expressing TTYH3-BRAF exhibited increased MEK/ERK signaling, which was blocked by clinically achievable concentrations of sorafenib, trametinib, and the paradox breaker PLX8394. These data provide the first example of a fully autoinhibited BRAF protein whose oncogenic potential is dictated by a distinct fusion partner and not by a structural change in BRAF itself.
Collapse
Affiliation(s)
- Florian Weinberg
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Ricarda Griffin
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martina Fröhlich
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Christoph Heining
- Department of Translational Medical Oncology, NCT Dresden, Dresden, and DKFZ, Heidelberg, Germany.,University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Sandra Braun
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Corinna Spohr
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Mary Iconomou
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Viola Hollek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Röring
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Gregor Warsow
- Omics IT and Data Management Core Facility, DKFZ, Heidelberg, Germany.,Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany
| | - Barbara Hutter
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Sebastian Uhrig
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Olaf Neumann
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Reuss
- DKTK, Heidelberg, Germany.,Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Translational NeuroOncology Research Group, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany.,DKTK, Munich, Germany
| | - Albrecht Stenzinger
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, NCT Dresden, Dresden, and DKFZ, Heidelberg, Germany.,University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany. .,DKTK, Heidelberg, Germany.
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany. .,Comprehensive Cancer Centre Freiburg, University of Freiburg, Freiburg, Germany. .,DKTK Partner Site Freiburg and DKFZ, Heidelberg, Germany.
| |
Collapse
|
37
|
Okada Y. Tweety Homologs (TTYH) Freshly Join the Journey of Molecular Identification of the VRAC/VSOR Channel Pore. Exp Neurobiol 2019; 28:131-133. [PMID: 31138984 PMCID: PMC6526112 DOI: 10.5607/en.2019.28.2.131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yasunobu Okada
- Department of Molecular Cellular Physiologye, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
| |
Collapse
|