1
|
Tetorou K, Aghaeipour A, Singh S, Morgan JE, Muntoni F. The role of dystrophin isoforms and interactors in the brain. Brain 2025; 148:1081-1098. [PMID: 39673425 PMCID: PMC11967788 DOI: 10.1093/brain/awae384] [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: 04/24/2024] [Revised: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 12/16/2024] Open
Abstract
Dystrophin is a protein crucial for maintaining the structural integrity of skeletal muscle. So far, attention has been focused on the role of dystrophin in muscle, in view of the devastating progression of weakness and early death that characterizes Duchenne muscular dystrophy. However, in the last few years, the role of shorter dystrophin isoforms, including development and adult expression-specific mechanisms, has been a greater focus. Within the cerebral landscape, various cell types, such as glia, oligodendrocytes and Purkinje, cerebellar granule and vascular-associated cells express a spectrum of dystrophin isoforms, including Dp427, Dp140, Dp71 and Dp40. The interaction of these isoforms with a multitude of proteins suggests their involvement in neurotransmission, influencing several circuit functions. This review presents the intricate interactions among dystrophin isoforms and diverse protein complexes across different cell types and brain regions, as well as the associated clinical complications. We focus on studies investigating protein interactions with dystrophin in the past 30 years at a biochemical level. In essence, the brain's dystrophin landscape is a thrilling exploration of diversity, challenging preconceptions and opening new avenues for understanding CNS physiology. It also holds potential therapeutic implications for neurological complications involving brain dystrophin deficiency. By revealing the molecular complexities related to dystrophin, this review paves the way for future investigations and therapeutic interventions for this CNS aspect of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Konstantina Tetorou
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Artadokht Aghaeipour
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Simran Singh
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Jennifer E Morgan
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Francesco Muntoni
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
2
|
Horvat S, Fonović UP, Mitrović A, Zidar N, Kos J, Pišlar A. The α- to γ-enolase switch: The role and regulation of γ-enolase during oligodendrocyte differentiation. Int J Biol Macromol 2025; 301:140464. [PMID: 39884600 DOI: 10.1016/j.ijbiomac.2025.140464] [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: 10/22/2024] [Revised: 01/11/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The glycolytic enzyme γ-enolase is a highly specific neuronal marker that is known to replace ubiquitously expressed α-enolase in the brain. Moreover, γ-enolase has been shown to exert neurotrophic activity, which is regulated by cathepsin X, a lysosomal peptidase. This study investigates the role of γ-enolase and its regulation by cathepsin X during the differentiation of oligodendrocytes, which are essential for normal brain function. We established a differentiation protocol for the human oligodendroglioma (HOG) cell line and demonstrated for the first time that an α- to γ-enolase switch occurs during HOG cell differentiation. This switch was confirmed by the expression of specific markers underscoring the role of γ-enolase in oligodendrocyte differentiation. Moreover, γ-enolase overexpression enhanced oligodendrocyte differentiation, while silencing of γ-enolase by siRNA significantly decreased maturation marker. Further, the regulatory role of cysteine peptidase cathepsin X on γ-enolase function was found. Silencing cathepsin X significantly changed cell morphology, enhanced oligodendrocyte differentiation, altered the expression of oligodendrocyte markers, and increased levels of the active form of γ-enolase. Inhibiting cathepsin X similarly changed cell morphology and enhanced oligodendrocyte differentiation. These findings suggest that cathepsin X modulates γ-enolase activity and thereby influences oligodendrocyte differentiation and thus neuronal function.
Collapse
Affiliation(s)
- Selena Horvat
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Urša Pečar Fonović
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Ana Mitrović
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.
| | - Nace Zidar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Janko Kos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.
| | - Anja Pišlar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
3
|
Horvat S, Kos J, Pišlar A. Multifunctional roles of γ-enolase in the central nervous system: more than a neuronal marker. Cell Biosci 2024; 14:61. [PMID: 38735971 PMCID: PMC11089681 DOI: 10.1186/s13578-024-01240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/27/2024] [Indexed: 05/14/2024] Open
Abstract
Enolase, a multifunctional protein with diverse isoforms, has generally been recognized for its primary roles in glycolysis and gluconeogenesis. The shift in isoform expression from α-enolase to neuron-specific γ-enolase extends beyond its enzymatic role. Enolase is essential for neuronal survival, differentiation, and the maturation of neurons and glial cells in the central nervous system. Neuron-specific γ-enolase is a critical biomarker for neurodegenerative pathologies and neurological conditions, not only indicating disease but also participating in nerve cell formation and neuroprotection and exhibiting neurotrophic-like properties. These properties are precisely regulated by cysteine peptidase cathepsin X and scaffold protein γ1-syntrophin. Our findings suggest that γ-enolase, specifically its C-terminal part, may offer neuroprotective benefits against neurotoxicity seen in Alzheimer's and Parkinson's disease. Furthermore, although the therapeutic potential of γ-enolase seems promising, the effectiveness of enolase inhibitors is under debate. This paper reviews the research on the roles of γ-enolase in the central nervous system, especially in pathophysiological events and the regulation of neurodegenerative diseases.
Collapse
Affiliation(s)
- Selena Horvat
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Janko Kos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
- Department of Biotechnology, Jožef Stefan Institute, Jamova Cesta 39, 1000, Ljubljana, Slovenia
| | - Anja Pišlar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
4
|
Zhang H, Li Z, Jiang J, Lei Y, Xie J, Liu Y, Yi B. SNTB1 regulates colorectal cancer cell proliferation and metastasis through YAP1 and the WNT/β-catenin pathway. Cell Cycle 2023; 22:1865-1883. [PMID: 37592763 PMCID: PMC10599191 DOI: 10.1080/15384101.2023.2244778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 08/19/2023] Open
Abstract
Colorectal cancer is a common type of digestive tract cancer with a significant morbidity and death rate across the world, partially attributing to the metastasis-associated problems. In this study, integrative bioinformatics analyses were performed to identify genes that might contribute to colorectal cancer metastasis, and 293 genes were dramatically increased and 369 genes were decreased within colon cancer samples. Among up-regulated genes, top five genes correlated with colorectal cancer patient's prognosis were verified for expression in clinical samples and syntrophin beta 1 (SNTB1) was the most up-regulated. In vitro, SNTB1 knockdown suppresses the malignant behaviors of colorectal cancer cells, including cell viability, colony formation capacity, as well as the abilities to migrate and invade. Furthermore, SNTB1 knockdown decreased the levels of Wnt1, C-Jun, C-Myc, TCF7, and cyclin D1, and inhibited EMT in both cell lines. In vivo, SNTB1 knockdown inhibited tumor growth and metastasis in nude mice models. SNTB1 positively regulated Yes1 associated transcriptional regulator (YAP1) expression; YAP1 partially reversed the effects of SNTB1 on colorectal cancer cell phenotypes and the Wnt/β-catenin/MYC signaling. In conclusion, SNTB1 knockdown inhibits colorectal cancer cell aggressiveness in vitro and tumor growth and metastasis in vivo through the Wnt/β-catenin/MYC signaling; YAP1 might mediate SNTB1 functions on colorectal cancer.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Li
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Juan Jiang
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yang Lei
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingmao Xie
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yihui Liu
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Bo Yi
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Liu L, Chen Y, Lin X, Wu M, Li J, Xie Q, Sferra TJ, Han Y, Liu H, Cao L, Yao M, Peng J, Shen A. Upregulation of SNTB1 correlates with poor prognosis and promotes cell growth by negative regulating PKN2 in colorectal cancer. Cancer Cell Int 2021; 21:547. [PMID: 34663329 PMCID: PMC8524951 DOI: 10.1186/s12935-021-02246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/05/2021] [Indexed: 11/12/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most highly malignant tumors and has a complicated pathogenesis. A preliminary study identified syntrophin beta 1 (SNTB1) as a potential oncogene in CRC. However, the clinical significance, biological function, and underlying mechanisms of SNTB1 in CRC remain largely unknown. Thus, the present study aimed to investigate the role of SNTB1 in CRC. Methods The expression profile of SNTB1 in CRC samples was evaluated by database analysis, cDNA array, tissue microarray, quantitative real-time PCR (qPCR), and immunohistochemistry. SNTB1 expression in human CRC cells was silenced using short hairpin RNAs (shRNA)/small interfering RNAs (siRNA) and its mRNA and protein levels were assessed by qPCR and/or western blotting. Cell viability, survival, cell cycle, and apoptosis were determined by the CCK-8 assay, colony formation, and flow cytometry assays, respectively. A xenograft nude mouse model of CRC was established to validate the roles of SNTB1 in vivo. Immunohistochemistry and TUNEL staining were used to determine the expression of SNTB1, PCNA, and cell apoptosis in tissue samples. Isobaric tag for relative and absolute quantification (iTRAQ) was used to analyze the differentially expressed proteins after knockdown of SNTB1 in CRC cells. Silence of protein kinase N2 (PKN2) using si-PNK2 was performed for rescue experiments. Results SNTB1 expression was increased in CRC tissues compared with adjacent noncancerous tissues and the increased SNTB1 expression was associated with shorter overall survival of CRC patients. Silencing of SNTB1 suppressed cell viability and survival, induced cell cycle arrest and apoptosis in vitro, and inhibited the growth of CRC cells in vivo. Further elucidation of the regulation of STNB1 on CRC growth by iTRAQ analysis identified 210 up-regulated and 55 down-regulated proteins in CRC cells after SNTB knockdown. A PPI network analysis identified PKN2 as a hub protein and was up-regulated in CRC cells after SNTB1 knockdown. Western-blot analysis further confirmed that SNTB1 knockdown significantly up-regulated PKN2 protein expression in CRC cells and decreased the phosphorylation of both ERK1/2 and AKT. Moreover, rescue experiments indicated that PKN2 knockdown significantly rescued SNTB1 knockdown-mediated decrease in cell viability, survival, and increase of cell cycle arrest at G0/G1 phase and apoptosis of CRC cells. Conclusions These findings indicate that SNTB1 is overexpressed in CRC. Elevated SNTB1 levels are correlated with shorter patient survival. Importantly, SNTB1 promotes tumor growth and progression of CRC, possibly by reducing the expression of PKN2 and activating the ERK and AKT signaling pathway. Our study highlights the potential of SNTB1 as a new prognostic factor and therapeutic target for CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02246-7.
Collapse
Affiliation(s)
- Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, UH Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA
| | - Xiaoying Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Jiapeng Li
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Thomas J Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, UH Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA
| | - Yuying Han
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Huixin Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Liujing Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Mengying Yao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China. .,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China. .,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
6
|
Kim MJ, Whitehead NP, Bible KL, Adams ME, Froehner SC. Mice lacking α-, β1- and β2-syntrophins exhibit diminished function and reduced dystrophin expression in both cardiac and skeletal muscle. Hum Mol Genet 2019; 28:386-395. [PMID: 30256963 DOI: 10.1093/hmg/ddy341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/21/2018] [Indexed: 01/16/2023] Open
Abstract
Syntrophins are a family of modular adaptor proteins that are part of the dystrophin protein complex, where they recruit and anchor a variety of signaling proteins. Previously we generated mice lacking α- and/or β2-syntrophin but showed that in the absence of one isoform, other syntrophin isoforms can partially compensate. Therefore, in the current study, we generated mice that lacked α, β1 and β2-syntrophins [triple syntrophin knockout (tKO) mice] and assessed skeletal and cardiac muscle function. The tKO mice showed a profound reduction in voluntary wheel running activity at both 6 and 12 months of age. Function of the tibialis anterior was assessed in situ and we found that the specific force of tKO muscle was decreased by 20-25% compared with wild-type mice. This decrease was accompanied by a shift in fiber-type composition from fast 2B to more oxidative fast 2A fibers. Using echocardiography to measure cardiac function, it was revealed that tKO hearts had left ventricular cardiac dysfunction and were hypertrophic, with a thicker left ventricular posterior wall. Interestingly, we also found that membrane-localized dystrophin expression was lower in both skeletal and cardiac muscles of tKO mice. Since dystrophin mRNA levels were not different in tKO, this finding suggests that syntrophins may regulate dystrophin trafficking to, or stabilization at, the sarcolemma. These results show that the loss of all three major muscle syntrophins has a profound effect on exercise performance, and skeletal and cardiac muscle dysfunction contributes to this deficiency.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nicholas P Whitehead
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Kenneth L Bible
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Adams ME, Odom GL, Kim MJ, Chamberlain JS, Froehner SC. Syntrophin binds directly to multiple spectrin-like repeats in dystrophin and mediates binding of nNOS to repeats 16-17. Hum Mol Genet 2019; 27:2978-2985. [PMID: 29790927 DOI: 10.1093/hmg/ddy197] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/14/2018] [Indexed: 11/12/2022] Open
Abstract
Mutation of the gene encoding dystrophin leads to Duchenne and Becker muscular dystrophy (DMD and BMD). Currently, dystrophin is thought to function primarily as a structural protein, connecting the muscle cell actin cytoskeleton to the extra-cellular matrix. In addition to this structural role, dystrophin also plays an important role as a scaffold that organizes an array of signaling proteins including sodium, potassium, and calcium channels, kinases, and nitric oxide synthase (nNOS). Many of these signaling proteins are linked to dystrophin via syntrophin, an adapter protein that is known to bind directly to two sites in the carboxyl terminal region of dystrophin. A search of the dystrophin sequence revealed three additional potential syntrophin binding sites (SBSs) within the spectrin-like repeat (SLR) region of dystrophin. Binding assays revealed that the site at SLR 17 bound specifically to the α isoform of syntrophin while the site at SLR 22 bound specifically to the β-syntrophins. The SLR 17 α-SBS contained the core sequence known to be required for nNOS-dystrophin interaction. In vitro and in vivo assays indicate that α-syntrophin facilitates the nNOS-dystrophin interaction at this site rather than nNOS binding directly to dystrophin as previously reported. The identification of multiple SBSs within the SLR region of dystrophin demonstrates that this region functions as a signaling scaffold. The signaling role of the SLR region of dystrophin will need to be considered for effective gene replacement or exon skipping based DMD/BMD therapies.
Collapse
Affiliation(s)
- Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA
| | - Guy L Odom
- Department of Neurology, University of Washington, Seattle, WA 98195-7290, USA.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA 98195-7720, USA
| | - Min Jeong Kim
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington, Seattle, WA 98195-7290, USA.,Department of Biochemistry, University of Washington, Seattle, WA 98195-7290, USA.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA 98195-7720, USA
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA
| |
Collapse
|
8
|
Bhat SS, Ali R, Khanday FA. Syntrophins entangled in cytoskeletal meshwork: Helping to hold it all together. Cell Prolif 2019; 52:e12562. [PMID: 30515904 PMCID: PMC6496184 DOI: 10.1111/cpr.12562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/23/2018] [Accepted: 11/08/2018] [Indexed: 01/10/2023] Open
Abstract
Syntrophins are a family of 59 kDa peripheral membrane-associated adapter proteins, containing multiple protein-protein and protein-lipid interaction domains. The syntrophin family consists of five isoforms that exhibit specific tissue distribution, distinct sub-cellular localization and unique expression patterns implying their diverse functional roles. These syntrophin isoforms form multiple functional protein complexes and ensure proper localization of signalling proteins and their binding partners to specific membrane domains and provide appropriate spatiotemporal regulation of signalling pathways. Syntrophins consist of two PH domains, a PDZ domain and a conserved SU domain. The PH1 domain is split by the PDZ domain. The PH2 and the SU domain are involved in the interaction between syntrophin and the dystrophin-glycoprotein complex (DGC). Syntrophins recruit various signalling proteins to DGC and link extracellular matrix to internal signalling apparatus via DGC. The different domains of the syntrophin isoforms are responsible for modulation of cytoskeleton. Syntrophins associate with cytoskeletal proteins and lead to various cellular responses by modulating the cytoskeleton. Syntrophins are involved in many physiological processes which involve cytoskeletal reorganization like insulin secretion, blood pressure regulation, myogenesis, cell migration, formation and retraction of focal adhesions. Syntrophins have been implicated in various pathologies like Alzheimer's disease, muscular dystrophy, cancer. Their role in cytoskeletal organization and modulation makes them perfect candidates for further studies in various cancers and other ailments that involve cytoskeletal modulation. The role of syntrophins in cytoskeletal organization and modulation has not yet been comprehensively reviewed till now. This review focuses on syntrophins and highlights their role in cytoskeletal organization, modulation and dynamics via its involvement in different cell signalling networks.
Collapse
Affiliation(s)
- Sahar S. Bhat
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of KashmirSrinagarIndia
| | - Roshia Ali
- Department of BiotechnologyUniversity of KashmirSrinagarIndia
- Department of BiochemistryUniversity of KashmirSrinagarIndia
| | | |
Collapse
|
9
|
Rao SB, Katoozi S, Skauli N, Froehner SC, Ottersen OP, Adams ME, Amiry-Moghaddam M. Targeted deletion of β1-syntrophin causes a loss of K ir 4.1 from Müller cell endfeet in mouse retina. Glia 2019; 67:1138-1149. [PMID: 30803043 DOI: 10.1002/glia.23600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 02/02/2023]
Abstract
Proper function of the retina depends heavily on a specialized form of retinal glia called Müller cells. These cells carry out important homeostatic functions that are contingent on their polarized nature. Specifically, the Müller cell endfeet that contact retinal microvessels and the corpus vitreum show a tenfold higher concentration of the inwardly rectifying potassium channel Kir 4.1 than other Müller cell plasma membrane domains. This highly selective enrichment of Kir 4.1 allows K+ to be siphoned through endfoot membranes in a special form of spatial buffering. Here, we show that Kir 4.1 is enriched in endfoot membranes through an interaction with β1-syntrophin. Targeted disruption of this syntrophin caused a loss of Kir 4.1 from Müller cell endfeet without affecting the total level of Kir 4.1 expression in the retina. Targeted disruption of α1-syntrophin had no effect on Kir 4.1 localization. Our findings show that the Kir 4.1 aggregation that forms the basis for K+ siphoning depends on a specific syntrophin isoform that colocalizes with Kir 4.1 in Müller endfoot membranes.
Collapse
Affiliation(s)
- Shreyas B Rao
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Shirin Katoozi
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nadia Skauli
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, Western Australia
| | - Ole Petter Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, Western Australia
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Ye R, Onodera T, Blanchard PG, Kusminski CM, Esser V, Brekken RA, Scherer PE. β1 Syntrophin Supports Autophagy Initiation and Protects against Cerulein-Induced Acute Pancreatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:813-825. [PMID: 30653956 DOI: 10.1016/j.ajpath.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 01/23/2023]
Abstract
Syntrophins are a family of proteins forming membrane-anchored scaffolds and serving as adaptors for various transmembrane and intracellular signaling molecules. To understand the physiological roles of β1 syntrophin, one of the least characterized members, we generated mouse models to eliminate β1 syntrophin specifically in the endocrine or exocrine pancreas. β1 syntrophin is dispensable for the morphology and function of insulin-producing β cells. However, mice with β1 syntrophin deletion in exocrine acinar cells exhibit increased severity of cerulein-induced acute pancreatitis. Reduced expression of cystic fibrosis transmembrane conductance regulator and dilation of acinar lumen are potential predisposition factors. During the disease progression, a relative lack of autophagy is associated with deficiencies in both actin assembly and endoplasmic reticulum nucleation. Our findings reveal, for the first time, that β1 syntrophin is a critical regulator of actin cytoskeleton and autophagy in pancreatic acinar cells and is potently protective against cerulein-induced acute pancreatitis.
Collapse
Affiliation(s)
- Risheng Ye
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Medical Education, Texas Tech University Health Sciences Center Paul L. Foster School of Medicine, El Paso, Texas
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Pierre-Gilles Blanchard
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Victoria Esser
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
11
|
Bhat HF, Mir SS, Dar KB, Bhat ZF, Shah RA, Ganai NA. ABC of multifaceted dystrophin glycoprotein complex (DGC). J Cell Physiol 2017; 233:5142-5159. [DOI: 10.1002/jcp.25982] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/01/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Hina F. Bhat
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Saima S. Mir
- Department of BiotechnologyUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Khalid B. Dar
- Department of BiochemistryUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Zuhaib F. Bhat
- Division of Livestock Products and TechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST‐J), R.S. PoraJammuJammu and KashmirIndia
| | - Riaz A. Shah
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Nazir A. Ganai
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| |
Collapse
|
12
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
13
|
Lee HJ, Kwon MH, Lee S, Hall RA, Yun CC, Choi I. Systematic family-wide analysis of sodium bicarbonate cotransporter NBCn1/SLC4A7 interactions with PDZ scaffold proteins. Physiol Rep 2014; 2:2/5/e12016. [PMID: 24844638 PMCID: PMC4098744 DOI: 10.14814/phy2.12016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
NBCn1 (SLC4A7) plays a role in transepithelial HCO3 (-) movement and intracellular pH maintenance in many tissues. In this study, we searched PDZ proteins capable of binding to NBCn1. We screened a protein array membrane, on which 96 different class I PDZ protein peptides were blotted, with the C-terminal domain of NBCn1 fused to GST. Thirteen proteins were identified in these screens: MAGI-3, NHERF-1, NHERF-2, PSD-95, chapsyn-110, ERBIN, MALS-1, densin-180, syntrophins α1, β2, γ2, MUPP1, and PDZK1. After determining these binding partners, we analyzed the database of known and predicted protein interactions to obtain an NBCn1 interaction network. The network shows NBCn1 being physically and functionally associated with a variety of membrane and cytosolic proteins via the binding partners. We then focused on syntrophin γ2 to examine the molecular and functional interaction between NBCn1 and one of the identified binding partners in the Xenopus oocyte expression system. GST/NBCn1 pulled down syntrophin γ2 and conversely GST/syntrophin γ2 pulled down NBCn1. Moreover, syntrophin γ2 increased intracellular pH recovery, from acidification, mediated by NBCn1's Na/HCO3 cotransport. Syntrophin γ2 also increased an ionic conductance produced by NBCn1 channel-like activity. Thus, syntrophin γ2 regulates NBCn1 activity. In conclusion, this study demonstrates that NBCn1 binds to many PDZ proteins, which in turn may allow the transporter to associate with other physiologically important proteins.
Collapse
Affiliation(s)
- Hye Jeong Lee
- Department of Pediatrics, Division of Hematology and Oncology, Vanderbilt University, Nashville, Tennessee, USA
| | - Min Hyung Kwon
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Soojung Lee
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Randy A Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - C Chris Yun
- Department of Medicine, Division of Digestive Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Inyeong Choi
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Edeleva EV, Shcherbata HR. Stress-induced ECM alteration modulates cellular microRNAs that feedback to readjust the extracellular environment and cell behavior. Front Genet 2013; 4:305. [PMID: 24427166 PMCID: PMC3876577 DOI: 10.3389/fgene.2013.00305] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/16/2013] [Indexed: 12/14/2022] Open
Abstract
The extracellular environment is a complex entity comprising of the extracellular matrix (ECM) and regulatory molecules. It is highly dynamic and under cell-extrinsic stress, transmits the stressed organism’s state to each individual ECM-connected cell. microRNAs (miRNAs) are regulatory molecules involved in virtually all the processes in the cell, especially under stress. In this review, we analyse how miRNA expression is regulated downstream of various signal transduction pathways induced by changes in the extracellular environment. In particular, we focus on the muscular dystrophy-associated cell adhesion molecule dystroglycan capable of signal transduction. Then we show how exactly the same miRNAs feedback to regulate the extracellular environment. The ultimate goal of this bi-directional signal transduction process is to change cell behavior under cell-extrinsic stress in order to respond to it accordingly.
Collapse
Affiliation(s)
- Evgeniia V Edeleva
- Max Planck Research Group for Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry Göttingen, Germany
| | - Halyna R Shcherbata
- Max Planck Research Group for Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry Göttingen, Germany
| |
Collapse
|
15
|
Janke A, Upadhaya R, Snow WM, Anderson JE. A new look at cytoskeletal NOS-1 and β-dystroglycan changes in developing muscle and brain in control and mdx dystrophic mice. Dev Dyn 2013; 242:1369-81. [PMID: 23940011 DOI: 10.1002/dvdy.24031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/17/2013] [Accepted: 07/25/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Loss of dystrophin profoundly affects muscle function and cognition. Changes in the dystrophin-glycoprotein complex (DGC) including disruption of nitric oxide synthase (NOS-1) may result from loss of dystrophin or secondarily after muscle damage. Disruptions in NOS-1 and beta-dystroglycan (bDG) were examined in developing diaphragm, quadriceps, and two brain regions between control and mdx mice at embryonic day E18 and postnatal days P1, P10, and P28. Age-dependent differential muscle loading allowed us to test the hypothesis that DGC changes are dependent on muscle use. RESULTS Muscle development, including loss of central nucleation and the localization of NOS-1 and bDG, was earlier in diaphragm than quadriceps; these features were differentially disrupted in dystrophic muscles. The NOS-1/bDG ratio, an index of DGC stability, was higher in dystrophic diaphragm (P10-P28) and quadriceps (P28) than controls. There were also distinct regional differences in NOS-1 and bDG in brain tissues with age and strain. NOS-1 increased with age in control forebrain and cerebellum, and in mdx cerebellum; NOS-1 and bDG were higher in control than mdx mouse forebrain. CONCLUSIONS Important developmental changes in structure and muscle DGC preceded the hallmarks of dystrophy, and are consistent with the impact of muscle-specific differential loading during maturation.
Collapse
Affiliation(s)
- Alyssa Janke
- Faculty of Science, Department of Biological Sciences, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | | | | | | |
Collapse
|
16
|
Bhat HF, Adams ME, Khanday FA. Syntrophin proteins as Santa Claus: role(s) in cell signal transduction. Cell Mol Life Sci 2013; 70:2533-54. [PMID: 23263165 PMCID: PMC11113789 DOI: 10.1007/s00018-012-1233-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 11/21/2012] [Accepted: 12/03/2012] [Indexed: 11/30/2022]
Abstract
Syntrophins are a family of cytoplasmic membrane-associated adaptor proteins, characterized by the presence of a unique domain organization comprised of a C-terminal syntrophin unique (SU) domain and an N-terminal pleckstrin homology (PH) domain that is split by insertion of a PDZ domain. Syntrophins have been recognized as an important component of many signaling events, and they seem to function more like the cell's own personal 'Santa Claus' that serves to 'gift' various signaling complexes with precise proteins that they 'wish for', and at the same time care enough for the spatial, temporal control of these signaling events, maintaining overall smooth functioning and general happiness of the cell. Syntrophins not only associate various ion channels and signaling proteins to the dystrophin-associated protein complex (DAPC), via a direct interaction with dystrophin protein but also serve as a link between the extracellular matrix and the intracellular downstream targets and cell cytoskeleton by interacting with F-actin. They play an important role in regulating the postsynaptic signal transduction, sarcolemmal localization of nNOS, EphA4 signaling at the neuromuscular junction, and G-protein mediated signaling. In our previous work, we reported a differential expression pattern of alpha-1-syntrophin (SNTA1) protein in esophageal and breast carcinomas. Implicated in several other pathologies, like cardiac dys-functioning, muscular dystrophies, diabetes, etc., these proteins provide a lot of scope for further studies. The present review focuses on the role of syntrophins in membrane targeting and regulation of cellular proteins, while highlighting their relevance in possible development and/or progression of pathologies including cancer which we have recently demonstrated.
Collapse
Affiliation(s)
- Hina F Bhat
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India.
| | | | | |
Collapse
|
17
|
Marrone AK, Edeleva EV, Kucherenko MM, Hsiao NH, Shcherbata HR. Dg-Dys-Syn1 signaling in Drosophila regulates the microRNA profile. BMC Cell Biol 2012; 13:26. [PMID: 23107381 PMCID: PMC3551632 DOI: 10.1186/1471-2121-13-26] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 10/22/2012] [Indexed: 11/30/2022] Open
Abstract
Background The Dystrophin Glycoprotein Complex (DGC) is at the center of significant inheritable diseases, such as muscular dystrophies that can be fatal and impair neuronal function in addition to muscle degeneration. Recent evidence has shown that it can control cellular homeostasis and work via Dystrophin signaling to regulate microRNA gene expression which implies that disease phenotypes hide an entourage of regulatory and homeostatic anomalies. Uncovering these hidden processes could shed new light on the importance of proper DGC function for an organism’s overall welfare and bring forth new ideas for treatments. Results To better understand a role for the DGC in these processes, we used the genetically advantageous Drosophila muscular dystrophy model to conduct a whole animal microarray screen. Since we have recently found that dystrophic symptoms can be caused by stress even in wild type animals and are enhanced in mutants, we screened stressed animals for microRNA misregulation as well. We were able to define microRNAs misregulated due to stress and/or dystrophy. Our results support the hypothesis that there is a Dystrophin and Dystroglycan dependent circuitry of processes linking stress response, dystrophic conditions and cellular signaling and that microRNAs play an important role in this network. Verification of a subset of our results was conducted via q-PCR and revealed that miR-956, miR-980 and miR-252 are regulated via a Dystroglycan-Dystrophin-Syntrophin dependent pathway. Conclusions The results presented in this study support the hypothesis that there is a Dystrophin and Dystroglycan dependent circuitry of processes that includes regulation of microRNAs. Dystrophin signaling has already been found to occur in mammalian musculature; however, our data reveals that this regulation is evolutionarily conserved and also present in at least neuronal tissues. Our data imply that Dystroglycan-Dystrophin-Syntrophin signaling through control of multiple microRNAs is involved in highly managed regulation of gene expression required to adapt cellular homeostasis that is compromised under stress and dystrophic conditions.
Collapse
Affiliation(s)
- April K Marrone
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for biophysical chemistry, Am Fassberg 11, Goettingen 37077, Germany
| | | | | | | | | |
Collapse
|
18
|
Leblond CS, Heinrich J, Delorme R, Proepper C, Betancur C, Huguet G, Konyukh M, Chaste P, Ey E, Rastam M, Anckarsäter H, Nygren G, Gillberg IC, Melke J, Toro R, Regnault B, Fauchereau F, Mercati O, Lemière N, Skuse D, Poot M, Holt R, Monaco AP, Järvelä I, Kantojärvi K, Vanhala R, Curran S, Collier DA, Bolton P, Chiocchetti A, Klauck SM, Poustka F, Freitag CM, Waltes R, Kopp M, Duketis E, Bacchelli E, Minopoli F, Ruta L, Battaglia A, Mazzone L, Maestrini E, Sequeira AF, Oliveira B, Vicente A, Oliveira G, Pinto D, Scherer SW, Zelenika D, Delepine M, Lathrop M, Bonneau D, Guinchat V, Devillard F, Assouline B, Mouren MC, Leboyer M, Gillberg C, Boeckers TM, Bourgeron T. Genetic and functional analyses of SHANK2 mutations suggest a multiple hit model of autism spectrum disorders. PLoS Genet 2012; 8:e1002521. [PMID: 22346768 PMCID: PMC3276563 DOI: 10.1371/journal.pgen.1002521] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 12/11/2011] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental disorders with a complex inheritance pattern. While many rare variants in synaptic proteins have been identified in patients with ASD, little is known about their effects at the synapse and their interactions with other genetic variations. Here, following the discovery of two de novo SHANK2 deletions by the Autism Genome Project, we identified a novel 421 kb de novo SHANK2 deletion in a patient with autism. We then sequenced SHANK2 in 455 patients with ASD and 431 controls and integrated these results with those reported by Berkel et al. 2010 (n = 396 patients and n = 659 controls). We observed a significant enrichment of variants affecting conserved amino acids in 29 of 851 (3.4%) patients and in 16 of 1,090 (1.5%) controls (P = 0.004, OR = 2.37, 95% CI = 1.23-4.70). In neuronal cell cultures, the variants identified in patients were associated with a reduced synaptic density at dendrites compared to the variants only detected in controls (P = 0.0013). Interestingly, the three patients with de novo SHANK2 deletions also carried inherited CNVs at 15q11-q13 previously associated with neuropsychiatric disorders. In two cases, the nicotinic receptor CHRNA7 was duplicated and in one case the synaptic translation repressor CYFIP1 was deleted. These results strengthen the role of synaptic gene dysfunction in ASD but also highlight the presence of putative modifier genes, which is in keeping with the "multiple hit model" for ASD. A better knowledge of these genetic interactions will be necessary to understand the complex inheritance pattern of ASD.
Collapse
Affiliation(s)
- Claire S. Leblond
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Jutta Heinrich
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Richard Delorme
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | | | - Catalina Betancur
- INSERM, U952, Paris, France
- CNRS, UMR 7224, Paris, France
- UPMC Univ Paris 06, Paris, France
| | - Guillaume Huguet
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Marina Konyukh
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Pauline Chaste
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Elodie Ey
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Maria Rastam
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | | | - Gudrun Nygren
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
| | - I. Carina Gillberg
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
| | - Jonas Melke
- Institute of Neuroscience and Physiology, Department of Pharmacology, Gothenburg University, Göteborg, Sweden
| | - Roberto Toro
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Beatrice Regnault
- Eukaryote Genotyping Platform, Genopole, Institut Pasteur, Paris, France
| | - Fabien Fauchereau
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Oriane Mercati
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - David Skuse
- Behavioural and Brain Sciences Unit, Institute of Child Health, University College London, London, United Kingdom
| | - Martin Poot
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Richard Holt
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Irma Järvelä
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Katri Kantojärvi
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Raija Vanhala
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Sarah Curran
- Academic Department of Child and Adolescent Psychiatry, Institute of Psychiatry, King's College London, London, United Kingdom
| | - David A. Collier
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Patrick Bolton
- Academic Department of Child and Adolescent Psychiatry, Institute of Psychiatry, King's College London, London, United Kingdom
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Andreas Chiocchetti
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine M. Klauck
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fritz Poustka
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Christine M. Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Regina Waltes
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Marnie Kopp
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Eftichia Duketis
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Elena Bacchelli
- Department of Biology, University of Bologna, Bologna, Italy
| | | | - Liliana Ruta
- Division of Child Neurology and Psychiatry, Department of Paediatrics, University of Catania, Catania, Italy
| | - Agatino Battaglia
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, Pisa, Italy
| | - Luigi Mazzone
- Division of Child Neurology and Psychiatry, Department of Pediatrics, University of Catania, Catania, Italy
| | - Elena Maestrini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Ana F. Sequeira
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Barbara Oliveira
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Astrid Vicente
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Guiomar Oliveira
- Unidade Neurodesenvolvimento e Autismo, Centro Investigação e Formação Clinica, Hospital Pediátrico Coimbra e Faculdade Medicina, Universidade Coimbra, Coimbra, Portugal
| | - Dalila Pinto
- The Centre for Applied Genomics and Program in Genetics and Genomic Biology, The Hospital for Sick Children, Toronto, Canada
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Program in Genetics and Genomic Biology, The Hospital for Sick Children, Toronto, Canada
| | | | | | | | - Dominique Bonneau
- INSERM U771 and CNRS 6214, Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
| | - Vincent Guinchat
- CADIPA–Centre de Ressources Autisme Rhône-Alpes, Saint Egrève, France
| | | | | | - Marie-Christine Mouren
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - Marion Leboyer
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- AP-HP, Hôpital H. Mondor–A. Chenevier, Département de Psychiatrie, Créteil, France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
- Institute of Child Health, University College London, London, United Kingdom
| | | | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| |
Collapse
|
19
|
Hafner A, Obermajer N, Kos J. γ-1-syntrophin mediates trafficking of γ-enolase towards the plasma membrane and enhances its neurotrophic activity. Neurosignals 2011; 18:246-58. [PMID: 21358174 DOI: 10.1159/000324292] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 01/13/2011] [Indexed: 12/29/2022] Open
Abstract
Syntrophins are scaffold proteins that can bind several signaling molecules and localize them to the plasma membrane. We demonstrate here that in neuroblastoma SH-SY5Y cells, brain-specific γ1-syntrophin binds the neurotrophic factor γ-enolase through its PDZ domain, and translocates it to the plasma membrane, as shown by immunoprecipitation, surface plasmon resonance, fluorescence colocalization and flow cytometry. Extensive colocalization of γ1-syntrophin and γ-enolase was observed in neurite growth cones in differentiated SH-SY5Y cells. Silencing of the γ1-syntrophin gene by RNA interference significantly reduced the re-distribution of γ-enolase to the plasma membrane and impaired its neurotrophic effects. We demonstrated that an intact C-terminal end of γ-enolase is essential for its γ1-syntrophin-assisted trafficking. The cleavage of two amino acids at the C-terminal end of γ-enolase by the carboxypeptidase cathepsin X prevents binding with the γ1-syntrophin PDZ domain. Collectively, these data demonstrate that γ1-syntrophin participates in γ-enolase translocation towards the plasma membrane, a pre-requisite for its neurotrophic activity. By disrupting this γ1-syntrophin-guided subcellular distribution, cathepsin X reduces γ-enolase-induced neurotrophic signaling.
Collapse
Affiliation(s)
- Anja Hafner
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | | |
Collapse
|
20
|
Abstract
The dystrophin protein complex, an important regulator of muscle membrane integrity, also maintains neural organization through interactions with the L1CAM family member SAX-7. The dystrophin protein complex (DPC), composed of dystrophin and associated proteins, is essential for maintaining muscle membrane integrity. The link between mutations in dystrophin and the devastating muscle failure of Duchenne’s muscular dystrophy (DMD) has been well established. Less well appreciated are the accompanying cognitive impairment and neuropsychiatric disorders also presented in many DMD patients, which suggest a wider role for dystrophin in membrane–cytoskeleton function. This study provides genetic evidence of a novel role for DYS-1/dystrophin in maintaining neural organization in Caenorhabditis elegans. This neuronal function is distinct from the established role of DYS-1/dystrophin in maintaining muscle integrity and regulating locomotion. SAX-7, an L1 cell adhesion molecule (CAM) homologue, and STN-2/γ-syntrophin also function to maintain neural integrity in C. elegans. This study provides biochemical data that show that SAX-7 associates with DYS-1 in an STN-2/γ-syntrophin–dependent manner. These results reveal a recruitment of L1CAMs to the DPC to ensure neural integrity is maintained.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Genetics, Cell Biology, and Development, Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
21
|
Adams ME, Anderson KNE, Froehner SC. The alpha-syntrophin PH and PDZ domains scaffold acetylcholine receptors, utrophin, and neuronal nitric oxide synthase at the neuromuscular junction. J Neurosci 2010; 30:11004-10. [PMID: 20720107 PMCID: PMC2936458 DOI: 10.1523/jneurosci.1930-10.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 05/25/2010] [Accepted: 06/27/2010] [Indexed: 11/21/2022] Open
Abstract
At the neuromuscular junction (NMJ), the dystrophin protein complex provides a scaffold that functions to stabilize acetylcholine receptor (AChR) clusters. Syntrophin, a key component of that scaffold, is a multidomain adapter protein that links a variety of signaling proteins and ion channels to the dystrophin protein complex. Without syntrophin, utrophin and neuronal nitric oxide synthase mu (nNOSmu) fail to localize to the NMJ and the AChRs are distributed abnormally. Here we investigate the contribution of syntrophin domains to AChR distribution and to localization of utrophin and nNOSmu at the NMJ. Transgenic mice expressing alpha-syntrophin lacking portions of the first pleckstrin homology (PH) domain (DeltaPH1a or DeltaPH1b) or the entire PDZ domain (DeltaPDZ) were bred onto the alpha-syntrophin null background. As expected the DeltaPDZ transgene did not restore the NMJ localization of nNOS. The DeltaPH1a transgene did restore postsynaptic nNOS but surprisingly did not restore sarcolemmal nNOS (although sarcolemmal aquaporin-4 was restored). Mice lacking the alpha-syntrophin PDZ domain or either half of the PH1 domain were able to restore utrophin to the NMJ but did not correct the aberrant AChR distribution of the alpha-syntrophin knock-out mice. However, mice expressing both the transgenic DeltaPDZ and the transgenic DeltaPH1a constructs did restore normal AChR distribution, demonstrating that both domains are required but need not be confined within the same protein to function. We conclude that the PH1 and PDZ domains of alpha-syntrophin work in concert to facilitate the localization of AChRs and nNOS at the NMJ.
Collapse
Affiliation(s)
- Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195-78290, USA.
| | | | | |
Collapse
|
22
|
Nagai R, Hashimoto R, Yamaguchi M. Drosophila Syntrophins are involved in locomotion and regulation of synaptic morphology. Exp Cell Res 2010; 316:2313-21. [PMID: 20632467 DOI: 10.1016/j.yexcr.2010.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Syntrophin components of the dystrophin glycoprotein complex (DGC) feature multiple protein interaction domains that may act in molecular scaffolding, recruiting signaling proteins to membranes and the DGC. Drosophila Syntrophin-1 (Syn1) and Syntrophin-2 (Syn2) are counterparts of human alpha1/beta1/beta2-syntrophins and gamma1/gamma2-syntrophins, respectively. alpha1/beta1/beta2-syntrophins are well documented, while little is known about gamma1/gamma2-syntrophins. Here, we performed immunohistochemical analyses with a specific antibody to Syn2 and demonstrated predominant expression in the larval and adult central nervous system. To investigate the in vivo functions of Syn2, we have generated Drosophila Syn2 deficiency mutants. Although the Syn2 mutants exhibit no overt phenotype, the combination of Syn1 knockdown and Syn2(37) mutation dramatically shortened life span, synergistically reduced locomotion ability and synergistically enhanced overgrowth of neuromuscular junctions in N-ethylmaleimide sensitive factor 2 mutants. From these data we conclude that Syn1 and Syn2 are required for locomotion and are involved in regulation of synaptic morphology. In addition, the two syntrophins can at least partially compensate for each other's functions.
Collapse
Affiliation(s)
- Rika Nagai
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | | | | |
Collapse
|
23
|
Abstract
Dystrophin and the alpha-dystrobrevins bind directly to the adapter protein syntrophin to form membrane-associated scaffolds. At the blood-brain barrier, alpha-syntrophin colocalizes with dystrophin and the alpha-dystrobrevins in perivascular glial endfeet and is required for localization of the water channel aquaporin-4. Earlier we have shown that localization of the scaffolding proteins gamma2-syntrophin, alpha-dystrobrevin-2, and dystrophin to glial endfeet is also dependent on the presence of alpha-syntrophin. In this study, we show that the expression levels of alpha-syntrophin, gamma2-syntrophin, and dystrophin at the blood-brain barrier are reduced in alpha-dystrobrevin-null mice. This is the first demonstration in which assembly of an astroglial protein scaffold containing syntrophin and dystrophin in perivascular astrocytes is dependent on the presence of alpha-dystrobrevin.
Collapse
Affiliation(s)
| | | | - Stanley C. Froehner
- Department of Physiology & Biophysics, University of Washington, Seattle, Washington 98195 USA
| |
Collapse
|
24
|
Böhm SV, Constantinou P, Tan S, Jin H, Roberts RG. Profound human/mouse differences in alpha-dystrobrevin isoforms: a novel syntrophin-binding site and promoter missing in mouse and rat. BMC Biol 2009; 7:85. [PMID: 19961569 PMCID: PMC2796648 DOI: 10.1186/1741-7007-7-85] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 12/04/2009] [Indexed: 11/29/2022] Open
Abstract
Background The dystrophin glycoprotein complex is disrupted in Duchenne muscular dystrophy and many other neuromuscular diseases. The principal heterodimeric partner of dystrophin at the heart of the dystrophin glycoprotein complex in the main clinically affected tissues (skeletal muscle, heart and brain) is its distant relative, α-dystrobrevin. The α-dystrobrevin gene is subject to complex transcriptional and post-transcriptional regulation, generating a substantial range of isoforms by alternative promoter use, alternative polyadenylation and alternative splicing. The choice of isoform is understood, amongst other things, to determine the stoichiometry of syntrophins (and their ligands) in the dystrophin glycoprotein complex. Results We show here that, contrary to the literature, most α-dystrobrevin genes, including that of humans, encode three distinct syntrophin-binding sites, rather than two, resulting in a greatly enhanced isoform repertoire. We compare in detail the quantitative tissue-specific expression pattern of human and mouse α-dystrobrevin isoforms, and show that two major gene features (the novel syntrophin-binding site-encoding exon and the internal promoter and first exon of brain-specific isoforms α-dystrobrevin-4 and -5) are present in most mammals but specifically ablated in mouse and rat. Conclusion Lineage-specific mutations in the murids mean that the mouse brain has fewer than half of the α-dystrobrevin isoforms found in the human brain. Our finding that there are likely to be fundamental functional differences between the α-dystrobrevins (and therefore the dystrophin glycoprotein complexes) of mice and humans raises questions about the current use of the mouse as the principal model animal for studying Duchenne muscular dystrophy and other related disorders, especially the neurological aspects thereof.
Collapse
Affiliation(s)
- Sabrina V Böhm
- Division of Medical & Molecular Genetics, King's College London, London, UK.
| | | | | | | | | |
Collapse
|
25
|
Syntrophin-2 is required for eye development in Drosophila. Exp Cell Res 2009; 316:272-85. [PMID: 19836389 DOI: 10.1016/j.yexcr.2009.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/07/2009] [Accepted: 10/07/2009] [Indexed: 11/23/2022]
Abstract
Syntrophins are components of the dystrophin glycoprotein complex (DGC), which is encoded by causative genes of muscular dystrophies. The DGC is thought to play roles not only in linking the actin cytoskeleton to the extracellular matrix, providing stability to the cell membrane, but also in signal transduction. Because of their binding to a variety of different molecules, it has been suggested that syntrophins are adaptor proteins recruiting signaling proteins to membranes and the DGC. However, critical roles in vivo remain elusive. Drosophila Syntrophin-2 (Syn2) is an orthologue of human gamma 1/gamma 2-syntrophins. Western immunoblot analysis here showed Syn2 to be expressed throughout development, with especially high levels in the adult head. Morphological aberrations were observed in Syn2 knockdown adult flies, with lack of retinal elongation and malformation of rhabdomeres. Furthermore, Syn2 knockdown flies exhibited excessive apoptosis in third instar larvae and alterations in the actin localization in the pupal retinae. Genetic crosses with a collection of Drosophila deficiency stocks allowed us to identify seven genomic regions, deletions of which caused enhancement of the rough eye phenotype induced by Syn2 knockdown. This information should facilitate identification of Syn2 regulators in Drosophila and clarification of roles of Syn2 in eye development.
Collapse
|
26
|
Charych EI, Liu F, Moss SJ, Brandon NJ. GABA(A) receptors and their associated proteins: implications in the etiology and treatment of schizophrenia and related disorders. Neuropharmacology 2009; 57:481-95. [PMID: 19631671 PMCID: PMC2836902 DOI: 10.1016/j.neuropharm.2009.07.027] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 07/02/2009] [Accepted: 07/13/2009] [Indexed: 02/05/2023]
Abstract
Gamma-aminobutyric acid type A (GABA(A)) receptors play an important role in mediating fast synaptic inhibition in the brain. They are ubiquitously expressed in the CNS and also represent a major site of action for clinically relevant drugs. Recent technological advances have greatly clarified the molecular and cellular roles played by distinct GABA(A) receptor subunit classes and isoforms in normal brain function. At the same time, postmortem and genetic studies have linked neuropsychiatric disorders including schizophrenia and bipolar disorder with GABAergic neurotransmission and various specific GABA(A) receptor subunits, while evidence implicating GABA(A)R-associated proteins is beginning to emerge. In this review we discuss the mounting genetic, molecular, and cellular evidence pointing toward a role for GABA(A) receptor heterogeneity in both schizophrenia etiology and therapeutic development. Finally, we speculate on the relationship between schizophrenia-related disorders and selected GABA(A) receptor associated proteins, key regulators of GABA(A) receptor trafficking, targeting, clustering, and anchoring that often carry out these functions in a subtype-specific manner.
Collapse
Affiliation(s)
- Erik I Charych
- Wyeth Research, Neuroscience Discovery, Princeton NJ 08852, USA.
| | | | | | | |
Collapse
|
27
|
Kim K, Yang J, Zhong XP, Kim MH, Kim YS, Lee HW, Han S, Choi J, Han K, Seo J, Prescott SM, Topham MK, Bae YC, Koretzky G, Choi SY, Kim E. Synaptic removal of diacylglycerol by DGKzeta and PSD-95 regulates dendritic spine maintenance. EMBO J 2009; 28:1170-9. [PMID: 19229292 PMCID: PMC2683696 DOI: 10.1038/emboj.2009.44] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Accepted: 01/27/2009] [Indexed: 12/22/2022] Open
Abstract
Diacylglycerol (DAG) is an important lipid signalling molecule that exerts an effect on various effector proteins including protein kinase C. A main mechanism for DAG removal is to convert it to phosphatidic acid (PA) by DAG kinases (DGKs). However, it is not well understood how DGKs are targeted to specific subcellular sites and tightly regulates DAG levels. The neuronal synapse is a prominent site of DAG production. Here, we show that DGKzeta is targeted to excitatory synapses through its direct interaction with the postsynaptic PDZ scaffold PSD-95. Overexpression of DGKzeta in cultured neurons increases the number of dendritic spines, which receive the majority of excitatory synaptic inputs, in a manner requiring its catalytic activity and PSD-95 binding. Conversely, DGKzeta knockdown reduces spine density. Mice deficient in DGKzeta expression show reduced spine density and excitatory synaptic transmission. Time-lapse imaging indicates that DGKzeta is required for spine maintenance but not formation. We propose that PSD-95 targets DGKzeta to synaptic DAG-producing receptors to tightly couple synaptic DAG production to its conversion to PA for the maintenance of spine density.
Collapse
Affiliation(s)
- Karam Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jinhee Yang
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Xiao-Ping Zhong
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Myoung-Hwan Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yun Sook Kim
- BK21 Program, Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun Woo Lee
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seungnam Han
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jeonghoon Choi
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kihoon Han
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jinsoo Seo
- BK21 Program, Department of Physiology, College of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Stephen M Prescott
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Matthew K Topham
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Yong Chul Bae
- BK21 Program, Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Gary Koretzky
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Se-Young Choi
- BK21 Program, Department of Physiology, College of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Eunjoon Kim
- National Creative Research Initiative Center for Synaptogenesis and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
28
|
Lyssand JS, DeFino MC, Tang XB, Hertz AL, Feller DB, Wacker JL, Adams ME, Hague C. Blood pressure is regulated by an alpha1D-adrenergic receptor/dystrophin signalosome. J Biol Chem 2008; 283:18792-800. [PMID: 18468998 DOI: 10.1074/jbc.m801860200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypertension is a cardiovascular disease associated with increased plasma catecholamines, overactivation of the sympathetic nervous system, and increased vascular tone and total peripheral resistance. A key regulator of sympathetic nervous system function is the alpha(1D)-adrenergic receptor (AR), which belongs to the adrenergic family of G-protein-coupled receptors (GPCRs). Endogenous catecholamines norepinephrine and epinephrine activate alpha(1D)-ARs on vascular smooth muscle to stimulate vasoconstriction, which increases total peripheral resistance and mean arterial pressure. Indeed, alpha(1D)-AR KO mice display a hypotensive phenotype and are resistant to salt-induced hypertension. Unfortunately, little information exists about how this important GPCR functions because of an inability to obtain functional expression in vitro. Here, we identified the dystrophin proteins, syntrophin, dystrobrevin, and utrophin as essential GPCR-interacting proteins for alpha(1D)-ARs. We found that dystrophins complex with alpha(1D)-AR both in vitro and in vivo to ensure proper functional expression. More importantly, we demonstrate that knock-out of multiple syntrophin isoforms results in the complete loss of alpha(1D)-AR function in mouse aortic smooth muscle cells and abrogation of alpha(1D)-AR-mediated increases in blood pressure. Our findings demonstrate that syntrophin and utrophin associate with alpha(1D)-ARs to create a functional signalosome, which is essential for alpha(1D)-AR regulation of vascular tone and blood pressure.
Collapse
Affiliation(s)
- John S Lyssand
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Albrecht DE, Sherman DL, Brophy PJ, Froehner SC. The ABCA1 cholesterol transporter associates with one of two distinct dystrophin-based scaffolds in Schwann cells. Glia 2008; 56:611-8. [PMID: 18286648 PMCID: PMC4335170 DOI: 10.1002/glia.20636] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cytoskeletal scaffolding complexes help organize specialized membrane domains with unique functions on the surface of cells. In this study, we define the scaffolding potential of the Schwann cell dystrophin glycoprotein complex (DGC) by establishing the presence of four syntrophin isoforms, (alpha1, beta1, beta2, and gamma2), and one dystrobrevin isoform, (alpha-dystrobrevin-1), in the abaxonal membrane. Furthermore, we demonstrate the existence of two separate DGCs in Schwann cells that divide the abaxonal membrane into spatially distinct domains, the DRP2/periaxin rich plaques and the Cajal bands that contain Dp116, utrophin, alpha-dystrobrevin-1 and four syntrophin isoforms. Finally, we show that the two different DGCs can scaffold unique accessory molecules in distinct areas of the Schwann cell membrane. Specifically, the cholesterol transporter ABCA1, associates with the Dp116/syntrophin complex in Cajal bands and is excluded from the DRP2/periaxin rich plaques.
Collapse
Affiliation(s)
- Douglas E Albrecht
- Department of Physiology and Biophysics, University of Washington, 1959 NE Pacific St, Box 357290, Seattle WA 98195-7290, USA
| | | | | | | |
Collapse
|
30
|
Pawlikowski BT, Maimone MM. alpha-Dystrobrevin isoforms differ in their colocalization with and stabilization of agrin-induced acetylcholine receptor clusters. Neuroscience 2008; 154:582-94. [PMID: 18468804 DOI: 10.1016/j.neuroscience.2008.01.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 12/21/2007] [Accepted: 01/12/2008] [Indexed: 11/19/2022]
Abstract
In skeletal muscle, alpha-dystrobrevin (alphaDB) is expressed throughout the sarcolemma with high concentrations at the neuromuscular junction. Mice lacking alphaDB display a mild muscular dystrophy and perturbations at the neuromuscular junction that include disruptions to acetylcholine receptor (AChR) cluster stability and patterning. In adult skeletal muscle, three alternatively spliced isoforms (alphaDB1, alphaDB2, alphaDB3) are expressed, while two other splice variants (alphaDB1(-) and alphaDB2(-)) are expressed only during early development. alphaDB is clearly important in AChR stabilization; however, the degree to which individual alphaDB isoforms and their specific functional domains contribute to AChR cluster stability is not fully understood. To investigate this, we established a primary muscle cell culture system from alphaDB knockout mice and stably expressed individual alphaDB isoforms using retroviral infection. A comparison between wild-type and alphaDB knockout muscle cells showed that in the absence of alphaDB, fewer AChR clusters formed in response to agrin treatment, and these AChR clusters were very unstable. Retroviral expression studies revealed that the largest isoforms (alphaDB1, alphaDB1(-), alphaDB2, alphaDB2(-)) colocalized with agrin-induced AChR clusters and rescued the AChR cluster formation defects back to wild-type levels, while only the first three isoforms fully rescued AChR cluster stability back to wild-type levels. alphaDB2(-) conferred an intermediate level of stability to the AChR clusters. In contrast, alphaDB3 showed no specific colocalization with AChR clusters and little effect on AChR cluster formation or stabilization. Twice as much syntrophin was found associated with alphaDB2 compared with alphaDB2(-) in myotubes suggesting that increased recruitment of syntrophin by alphaDB may enhance the stability of AChR clusters. Taken together, these data demonstrate that different alphaDB isoforms have different functional capabilities in the formation and maintenance of AChR clusters in muscle cells, and that these differences are likely due to the presence of different functional domains in each isoform.
Collapse
MESH Headings
- Agrin/pharmacology
- Animals
- Cells, Cultured
- DNA, Complementary/biosynthesis
- DNA, Complementary/genetics
- Dystrophin-Associated Proteins/genetics
- Dystrophin-Associated Proteins/metabolism
- Genetic Vectors
- Isomerism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Myoblasts/drug effects
- Myoblasts/enzymology
- Plasmids/genetics
- Receptors, Cholinergic/drug effects
- Receptors, Cholinergic/metabolism
- Retroviridae/genetics
Collapse
Affiliation(s)
- B T Pawlikowski
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
31
|
Okumura A, Nagai K, Okumura N. Interaction of α1-syntrophin with multiple isoforms of heterotrimeric G protein α subunits. FEBS J 2007; 275:22-33. [DOI: 10.1111/j.1742-4658.2007.06174.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Rees MLJ, Lien CF, Górecki DC. Dystrobrevins in muscle and non-muscle tissues. Neuromuscul Disord 2007; 17:123-34. [PMID: 17251025 DOI: 10.1016/j.nmd.2006.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 10/26/2006] [Accepted: 11/20/2006] [Indexed: 01/23/2023]
Abstract
The alpha- and beta-dystrobrevins belong to the family of dystrophin-related and dystrophin-associated proteins. As constituents of the dystrophin-associated protein complex, alpha-dystrobrevin was believed to have a role predominantly in muscles and beta-dystrobrevin in non-muscle tissues. Recent reports described novel localisations and molecular characteristics of alpha-dystrobrevin isoforms in non-muscle tissues (developing and adult). While single and double knockout studies have revealed distinct functions of dystrobrevin in some tissues, these also suggested a strong compensatory mechanism, where dystrobrevins displaying overlapping tissue expression pattern and structure/function similarity can substitute each other. No human disease has been unequivocally associated within mutations of dystrobrevin genes. However, some significant exceptions to these overlapping expression patterns, mainly in the brain, suggest that dystrobrevin mutations might underlie some specific motor, behavioural or cognitive defects. Dystrobrevin binding partner DTNBP1 (dysbindin) is a probable susceptibility gene for schizophrenia and bipolar affective disorder in some populations. As dysbindin abnormality is linked to Hermansky-Pudlak syndrome, dystrobrevins and/or their binding partners may also be required for proper function of other non-muscle tissues.
Collapse
Affiliation(s)
- Melissa L J Rees
- Department of Molecular Medicine, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | | | | |
Collapse
|