1
|
Xiong X, Lee HC, Lu T. Impact of Sorbs2 dysfunction on cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167813. [PMID: 40139410 PMCID: PMC12037213 DOI: 10.1016/j.bbadis.2025.167813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Despite significant advancements in prevention and treatment over the past decades, cardiovascular diseases (CVDs) remain the leading cause of death worldwide. CVDs involve multifactorial inheritance, but our understanding of the genetic impact on these diseases is still incomplete. Sorbin and SH3 domain-containing protein 2 (Sorbs2) is ubiquitously expressed in various tissues, including the cardiovascular system. Increasing evidence suggests that Sorbs2 malfunction contributes to CVDs. This manuscript will review our current understanding of the potential mechanisms underlying Sorbs2 dysregulation in the development of CVDs.
Collapse
Affiliation(s)
- Xiaowei Xiong
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Hon-Chi Lee
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Tong Lu
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
2
|
Zhao G, Li C, Liu W, Wu J, Yang X. Understanding the Molecular Mechanisms of SORBS2 in TNBC Lung Metastasis. Biochem Biophys Res Commun 2025; 762:151762. [PMID: 40199126 DOI: 10.1016/j.bbrc.2025.151762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025]
Abstract
Metastasis is the leading cause of recurrence and mortality in triple-negative breast cancer (TNBC), an aggressive subtype that predominantly spreads to the lungs, brain, bones, and liver, with lung metastasis being particularly prevalent. Despite the clinical significance of TNBC metastasis, the molecular mechanisms that drive lung-specific metastasis remain poorly understood. RNA-binding proteins (RBPs) are crucial regulators of post-transcriptional gene expression and are frequently dysregulated in cancers. This study identifies SORBS2 as a critical RBP implicated in TNBC lung metastasis. Using RNA sequencing (RNA-seq) and LACE-seq, we demonstrate that SORBS2 regulates a specific set of genes through direct binding to coding sequences (CDS), introns, and 3' untranslated regions (UTRs), and its binding targets are linked to various pathways, including a possible association with Wnt/β-catenin signaling, among others. Functional assays confirm that SORBS2 knockdown inhibits proliferation, migration, and invasion in TNBC cells. These findings highlight SORBS2 as a key regulator of TNBC lung metastasis, with a context-dependent role that promotes metastatic behavior in highly metastatic TNBC cells, providing potential avenues for novel therapeutic strategies.
Collapse
Affiliation(s)
- Gongke Zhao
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Chunzheng Li
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Wan Liu
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Jianing Wu
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Xianguang Yang
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China.
| |
Collapse
|
3
|
Veloso A, Bleuart A, Conrard L, Orban T, Bruyr J, Cabochette P, Germano RFV, Schevenels G, Bernard A, Zindy E, Demeyer S, Vanhollebeke B, Dequiedt F, Martin M. The cytoskeleton adaptor protein Sorbs1 controls the development of lymphatic and venous vessels in zebrafish. BMC Biol 2024; 22:51. [PMID: 38414014 PMCID: PMC10900589 DOI: 10.1186/s12915-024-01850-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Lymphangiogenesis, the formation of lymphatic vessels, is tightly linked to the development of the venous vasculature, both at the cellular and molecular levels. Here, we identify a novel role for Sorbs1, the founding member of the SoHo family of cytoskeleton adaptor proteins, in vascular and lymphatic development in the zebrafish. RESULTS We show that Sorbs1 is required for secondary sprouting and emergence of several vascular structures specifically derived from the axial vein. Most notably, formation of the precursor parachordal lymphatic structures is affected in sorbs1 mutant embryos, severely impacting the establishment of the trunk lymphatic vessel network. Interestingly, we show that Sorbs1 interacts with the BMP pathway and could function outside of Vegfc signaling. Mechanistically, Sorbs1 controls FAK/Src signaling and subsequently impacts on the cytoskeleton processes regulated by Rac1 and RhoA GTPases. Inactivation of Sorbs1 altered cell-extracellular matrix (ECM) contacts rearrangement and cytoskeleton dynamics, leading to specific defects in endothelial cell migratory and adhesive properties. CONCLUSIONS Overall, using in vitro and in vivo assays, we identify Sorbs1 as an important regulator of venous and lymphatic angiogenesis independently of the Vegfc signaling axis. These results provide a better understanding of the complexity found within context-specific vascular and lymphatic development.
Collapse
Affiliation(s)
- Alexandra Veloso
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Anouk Bleuart
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Louise Conrard
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Tanguy Orban
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Jonathan Bruyr
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Pauline Cabochette
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
- Present Address: Laboratory of Developmental Genetics, ULB Neuroscience Institute, Université Libre de Bruxelles, B-6041, Gosselies, Belgium
| | - Raoul F V Germano
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Giel Schevenels
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Alice Bernard
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, University of Liège (ULiège), Liège, Belgium
| | - Egor Zindy
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Sofie Demeyer
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Benoit Vanhollebeke
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Franck Dequiedt
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Maud Martin
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium.
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium.
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium.
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium.
- WEL Research Institute (WELBIO Department), Avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
4
|
Lee SE, Chang S. nArgBP2 together with GKAP and SHANK3 forms a dynamic layered structure. Front Cell Neurosci 2024; 18:1354900. [PMID: 38440150 PMCID: PMC10909995 DOI: 10.3389/fncel.2024.1354900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
nArgBP2, a protein whose disruption is implicated in intellectual disability, concentrates in excitatory spine-synapses. By forming a triad with GKAP and SHANK, it regulates spine structural rearrangement. We here find that GKAP and SHANK3 concentrate close to the synaptic contact, whereas nArgBP2 concentrates more centrally in the spine. The three proteins collaboratively form biomolecular condensates in living fibroblasts, exhibiting distinctive layered localizations. nArgBP2 concentrates in the inner phase, SHANK3 in the outer phase, and GKAP partially in both. Upon co-expression of GKAP and nArgBP2, they evenly distribute within condensates, with a notable peripheral localization of SHANK3 persisting when co-expressed with either GKAP or nArgBP2. Co-expression of SHANK3 and GKAP with CaMKIIα results in phase-in-phase condensates, with CaMKIIα at the central locus and SHANK3 and GKAP exhibiting peripheral localization. Additional co-expression of nArgBP2 maintains the layered organizational structure within condensates. Subsequent CaMKIIα activation disperses a majority of the condensates, with an even distribution of all proteins within the extant deformed condensates. Our findings suggest that protein segregation via phase separation may contribute to establishing layered organization in dendritic spines.
Collapse
Affiliation(s)
- Sang-Eun Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Kutsuna S, Sugiyama G, Komiyama T, Kamohara H, Ohyama Y, Kumamaru W, Yamada T. TNF-α-induced Inhibition of Protein Myristoylation Via Binding Between NMT1 and Sorbs2 in Osteoblasts. In Vivo 2024; 38:107-113. [PMID: 38148048 PMCID: PMC10756471 DOI: 10.21873/invivo.13416] [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: 10/04/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND/AIM Bone resolution due to tumor invasion often occurs on the surface of the jaw and is important for clinical prognosis. Although cytokines, such as TNF-α are known to impair osteoblasts, the underlying mechanism remains unclear. Protein myristoylation, a post-translational modification, plays an important role in the development of immune responses and cancerization of cells. A clear understanding of the mechanisms underlying this involvement will provide insights into molecular-targeted therapies. N-myristoyltransferase1 (NMT1), a specific enzyme involved in myristoylation, is expressed in cancer cells and in other normal cells, suggesting that changes in myristoylation may result from the regulation of NMT1 in cancer cells. MATERIALS AND METHODS Using newly emerging state-of-the-art techniques such as the Click-it assay, RNA interference, mass spectrometry, immunoprecipitation, immunocytochemistry, and western blotting, the expression of myristoylated proteins and the role of TNF-α stimulation on NMT1 and Sorbs2 binding were evaluated in a murine osteoblastic cell line (MC3T3-E1). RESULTS The expression of myristoylated proteins was detected; however, TNF-α stimulation resulted in their inhibition in MC3T3-E1 cells. The expression of NMT1 also increased. Immunoprecipitation and mass spectrometry identified Sorbs2 as a novel binding protein of NMT1, which upon TNF-α stimulation, inhibited myristoylation. CONCLUSION The binding between NMT1 and Sorbs2 can regulate myristoylation, and NMT1 can be considered as a potential target molecule for tumor invasion.
Collapse
Affiliation(s)
- Shigehiko Kutsuna
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Goro Sugiyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takuma Komiyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hanae Kamohara
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yukiko Ohyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Wataru Kumamaru
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomohiro Yamada
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Wang T, Bai Y, Zheng X, Liu X, Xing S, Wang L, Wang H, Feng G, Li C. Sapap4 deficiency leads to postsynaptic defects and abnormal behaviors relevant to hyperkinetic neuropsychiatric disorder in mice. Cereb Cortex 2023; 33:1104-1118. [PMID: 35368073 DOI: 10.1093/cercor/bhac123] [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: 06/18/2020] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Postsynaptic proteins play critical roles in synaptic development, function, and plasticity. Dysfunction of postsynaptic proteins is strongly linked to neurodevelopmental and psychiatric disorders. SAP90/PSD95-associated protein 4 (SAPAP4; also known as DLGAP4) is a key component of the PSD95-SAPAP-SHANK excitatory postsynaptic scaffolding complex, which plays important roles at synapses. However, the exact function of the SAPAP4 protein in the brain is poorly understood. Here, we report that Sapap4 knockout (KO) mice have reduced spine density in the prefrontal cortex and abnormal compositions of key postsynaptic proteins in the postsynaptic density (PSD) including reduced PSD95, GluR1, and GluR2 as well as increased SHANK3. These synaptic defects are accompanied by a cluster of abnormal behaviors including hyperactivity, impulsivity, reduced despair/depression-like behavior, hypersensitivity to low dose of amphetamine, memory deficits, and decreased prepulse inhibition, which are reminiscent of mania. Furthermore, the hyperactivity of Sapap4 KO mice could be partially rescued by valproate, a mood stabilizer used for mania treatment in humans. Together, our findings provide evidence that SAPAP4 plays an important role at synapses and reinforce the view that dysfunction of the postsynaptic scaffolding protein SAPAP4 may contribute to the pathogenesis of hyperkinetic neuropsychiatric disorder.
Collapse
Affiliation(s)
- Tianhua Wang
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Yunxia Bai
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Xianjie Zheng
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Xinxia Liu
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Shuang Xing
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Linbin Wang
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Huimin Wang
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China.,NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts, 02139, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts, 02142, USA
| | - Chunxia Li
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), School of Psychology and Cognitive Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| |
Collapse
|
7
|
Biomolecular condensate assembly of nArgBP2 tunes its functionality to manifest the structural plasticity of dendritic spines. Exp Mol Med 2023; 55:108-119. [PMID: 36599935 PMCID: PMC9898538 DOI: 10.1038/s12276-022-00918-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 01/05/2023] Open
Abstract
nArgBP2, a candidate gene for intellectual disability, is a postsynaptic protein critical for dendritic spine development and morphogenesis, and its knockdown (KD) in developing neurons severely impairs spine-bearing excitatory synapse formation. Surprisingly, nArgBP2 KD in mature neurons did not cause morphological defects in the existing spines at rest, raising questions of how it functions in mature neurons. We found that unlike its inaction at rest, nArgBP2 KD completely inhibited the enlargement of dendritic spines during chemically induced long-term potentiation (cLTP) in mature neurons. We further found that nArgBP2 forms condensates in dendritic spines and that these condensates are dispersed by cLTP, which spatiotemporally coincides with spine head enlargement. Condensates with CaMKII phosphorylation-deficient mutant or CaMKII inhibition are neither dispersed nor accompanied by spine enlargement during cLTP. We found that nArgBP2 condensates in spines exhibited liquid-like properties, and in heterologous and in vitro expression systems, nArgBP2 undergoes liquid-liquid phase separation via multivalent intermolecular interactions between SH3 domains and proline-rich domains. It also forms coacervates with CaMKIIα, which is rapidly dissembled by calcium/CaMKIIα-dependent phosphorylation. We further showed that the interaction between nArgBP2 and WAVE1 competes with nArgBP2 phase separation and that blocking the nArgBP2-WAVE1 interaction prevents spine enlargement during cLTP. Together, our results suggest that nArgBP2 at rest is confined to the condensates but is released by CaMKIIα-mediated phosphorylation during synaptic plasticity, which regulates its timely interaction with WAVE1 to induce spine head enlargement in mature neurons.
Collapse
|
8
|
Li H, Zhang J, Ke JR, Yu Z, Shi R, Gao SS, Li JF, Gao ZX, Ke CS, Han HX, Xu J, Leng Q, Wu GR, Li Y, Tao L, Zhang X, Sy MS, Li C. Pro-prion, as a membrane adaptor protein for E3 ligase c-Cbl, facilitates the ubiquitination of IGF-1R, promoting melanoma metastasis. Cell Rep 2022; 41:111834. [PMID: 36543142 DOI: 10.1016/j.celrep.2022.111834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/13/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Aberrant activation of receptor tyrosine kinase (RTK) is usually a result of mutation and plays important roles in tumorigenesis. How RTK without mutation affects tumorigenesis remains incompletely understood. Here we show that in human melanomas pro-prion (pro-PrP) is an adaptor protein for an E3 ligase c-Cbl, enabling it to polyubiquitinate activated insulin-like growth factor-1 receptor (IGF-1R), leading to enhanced melanoma metastasis. All human melanoma cell lines studied here express pro-PrP, retaining its glycosylphosphatidylinositol-peptide signal sequence (GPI-PSS). The sequence, PVILLISFLI in the GPI-PSS of pro-PrP, binds c-Cbl, docking c-Cbl to the inner cell membrane, forming a pro-PrP/c-Cbl/IGF-1R trimeric complex. Subsequently, IGF-1R polyubiquitination and degradation are augmented, which increases autophagy and tumor metastasis. Importantly, the synthetic peptide PVILLISFLI disrupts the pro-PrP/c-Cbl/IGF-1R complex, reducing cancer cell autophagy and mitigating tumor aggressiveness in vitro and in vivo. Targeting cancer-associated GPI-PSS may provide a therapeutic approach for treating human cancers expressing pro-PrP.
Collapse
Affiliation(s)
- Huan Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China; Wuhan Institute of Virology, Chinese Academy of Sciences, 44 Xiao Hong Shan Zhong Qu, Wuhan 430030, China
| | - Jie Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Jing-Ru Ke
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ze Yu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Run Shi
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Shan-Shan Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Jing-Feng Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Zhen-Xing Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Chang-Shu Ke
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Hui-Xia Han
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, No. 1023-1063 Shatai South Road, Guangzhou 510515, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, No. 107 North 2nd Road, Shihezi 832008, China
| | - Qibin Leng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Gui-Ru Wu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Yingqiu Li
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, 135 West Xingang Road, Guangzhou 510275, China
| | - Lin Tao
- Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832008, China
| | - Xianghui Zhang
- Department of Public Health, Shihezi University School of Medicine, Shihezi 832000, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chaoyang Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China.
| |
Collapse
|
9
|
Bai Y, Wang H, Li C. SAPAP Scaffold Proteins: From Synaptic Function to Neuropsychiatric Disorders. Cells 2022; 11:cells11233815. [PMID: 36497075 PMCID: PMC9740047 DOI: 10.3390/cells11233815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Excitatory (glutamatergic) synaptic transmission underlies many aspects of brain activity and the genesis of normal human behavior. The postsynaptic scaffolding proteins SAP90/PSD-95-associated proteins (SAPAPs), which are abundant components of the postsynaptic density (PSD) at excitatory synapses, play critical roles in synaptic structure, formation, development, plasticity, and signaling. The convergence of human genetic data with recent in vitro and in vivo animal model data indicates that mutations in the genes encoding SAPAP1-4 are associated with neurological and psychiatric disorders, and that dysfunction of SAPAP scaffolding proteins may contribute to the pathogenesis of various neuropsychiatric disorders, such as schizophrenia, autism spectrum disorders, obsessive compulsive disorders, Alzheimer's disease, and bipolar disorder. Here, we review recent major genetic, epigenetic, molecular, behavioral, electrophysiological, and circuitry studies that have advanced our knowledge by clarifying the roles of SAPAP proteins at the synapses, providing new insights into the mechanistic links to neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yunxia Bai
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
| | - Huimin Wang
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai 200062, China
| | - Chunxia Li
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
- Correspondence:
| |
Collapse
|
10
|
Advances in the previous two decades in our understanding of the post-translational modifications, functions, and drug perspectives of ArgBP2 and its family members. Biomed Pharmacother 2022; 155:113853. [DOI: 10.1016/j.biopha.2022.113853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022] Open
|
11
|
Svec KV, Howe AK. Protein Kinase A in cellular migration-Niche signaling of a ubiquitous kinase. Front Mol Biosci 2022; 9:953093. [PMID: 35959460 PMCID: PMC9361040 DOI: 10.3389/fmolb.2022.953093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cell migration requires establishment and maintenance of directional polarity, which in turn requires spatial heterogeneity in the regulation of protrusion, retraction, and adhesion. Thus, the signaling proteins that regulate these various structural processes must also be distinctly regulated in subcellular space. Protein Kinase A (PKA) is a ubiquitous serine/threonine kinase involved in innumerable cellular processes. In the context of cell migration, it has a paradoxical role in that global inhibition or activation of PKA inhibits migration. It follows, then, that the subcellular regulation of PKA is key to bringing its proper permissive and restrictive functions to the correct parts of the cell. Proper subcellular regulation of PKA controls not only when and where it is active but also specifies the targets for that activity, allowing the cell to use a single, promiscuous kinase to exert distinct functions within different subcellular niches to facilitate cell movement. In this way, understanding PKA signaling in migration is a study in context and in the elegant coordination of distinct functions of a single protein in a complex cellular process.
Collapse
Affiliation(s)
- Kathryn V. Svec
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, V T, United States
- University of Vermont Cancer Center, University of Vermont, Burlington, VT, United States
| |
Collapse
|
12
|
McLendon JM, Zhang X, Matasic DS, Kumar M, Koval OM, Grumbach IM, Sadayappan S, London B, Boudreau RL. Knockout of Sorbin And SH3 Domain Containing 2 (Sorbs2) in Cardiomyocytes Leads to Dilated Cardiomyopathy in Mice. J Am Heart Assoc 2022; 11:e025687. [PMID: 35730644 PMCID: PMC9333371 DOI: 10.1161/jaha.122.025687] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Sorbin and SH3 domain containing 2 (Sorbs2) protein is a cytoskeletal adaptor with an emerging role in cardiac biology and disease; yet, its potential relevance to adult‐onset cardiomyopathies remains underexplored. Sorbs2 global knockout mice display lethal arrhythmogenic cardiomyopathy; however, the causative mechanisms remain unclear. Herein, we examine Sorbs2 dysregulation in heart failure, characterize novel Sorbs2 cardiomyocyte‐specific knockout mice (Sorbs2‐cKO), and explore associations between Sorbs2 genetic variations and human cardiovascular disease. Methods and Results Bioinformatic analyses show myocardial Sorbs2 mRNA is consistently upregulated in humans with adult‐onset cardiomyopathies and in heart failure models. We generated Sorbs2‐cKO mice and report that they develop progressive systolic dysfunction and enlarged cardiac chambers, and they die with congestive heart failure at about 1 year old. After 3 months, Sorbs2‐cKO mice begin to show atrial enlargement and P‐wave anomalies, without dysregulation of action potential–associated ion channel and gap junction protein expressions. After 6 months, Sorbs2‐cKO mice exhibit impaired contractility in dobutamine‐treated hearts and skinned myofibers, without dysregulation of contractile protein expressions. From our comprehensive survey of potential mechanisms, we found that within 4 months, Sorbs2‐cKO hearts have defective microtubule polymerization and compensatory upregulation of structural cytoskeletal and adapter proteins, suggesting that this early intracellular structural remodeling is responsible for contractile dysfunction. Finally, we identified genetic variants that associate with decreased Sorbs2 expression and human cardiac phenotypes, including conduction abnormalities, atrial enlargement, and dilated cardiomyopathy, consistent with Sorbs2‐cKO mice phenotypes. Conclusions Our studies show that Sorbs2 is essential for maintaining structural integrity in cardiomyocytes, likely through strengthening the interactions between microtubules and other cytoskeletal proteins at cross‐link sites.
Collapse
Affiliation(s)
- Jared M McLendon
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Xiaoming Zhang
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Daniel S Matasic
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Department of Molecular Physiology and Biophysics University of Iowa Carver College of Medicine Iowa City IA
| | - Mohit Kumar
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Olha M Koval
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Isabella M Grumbach
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Sakthivel Sadayappan
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Barry London
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Ryan L Boudreau
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| |
Collapse
|
13
|
Morris T, Sue E, Geniesse C, Brieher WM, Tang VW. Synaptopodin stress fiber and contractomere at the epithelial junction. J Cell Biol 2022; 221:e202011162. [PMID: 35416930 PMCID: PMC9011326 DOI: 10.1083/jcb.202011162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.
Collapse
Affiliation(s)
- Timothy Morris
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Eva Sue
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Caleb Geniesse
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
14
|
Kovacs-Kasa A, Kovacs L, Cherian-Shaw M, Patel V, Meadows ML, Fulton DJ, Su Y, Verin AD. Inhibition of Class IIa HDACs improves endothelial barrier function in endotoxin-induced acute lung injury. J Cell Physiol 2021; 236:2893-2905. [PMID: 32959895 PMCID: PMC9946131 DOI: 10.1002/jcp.30053] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Acute lung injury (ALI) is an acute inflammatory process arises from a wide range of lung insults. A major cause of ALI is dysfunction of the pulmonary vascular endothelial barrier but the mechanisms involved are incompletely understood. The therapeutic potential of histone deacetylase (HDAC) inhibitors for the treatment of cardiovascular and inflammatory diseases is increasingly apparent, but the mechanisms by which HDACs regulate pulmonary vascular barrier function remain to be resolved. We found that specific Class IIa HDACs inhibitor, TMP269, significantly attenuated the lipopolysaccharide (LPS)-induced human lung microvascular endothelial cells (HLMVEC) barrier compromise in vitro and improved vascular barrier integrity and lung function in murine model of ALI in vivo. TMP269 decreased LPS-induced myosin light chain phosphorylation suggesting the role for Class IIa HDACs in LPS-induced cytoskeleton reorganization. TMP269 did not affect microtubule structure and tubulin acetylation in contrast to the HDAC6-specific inhibitor, Tubastatin A suggesting that Class IIa HDACs and HDAC6 (Class IIb) regulate endothelial cytoskeleton and permeability via different mechanisms. Furthermore, LPS increased the expression of ArgBP2 which has recently been attributed to HDAC-mediated activation of Rho. Depletion of ArgBP2 abolished the ability of LPS to disrupt barrier function in HLMVEC and both TMP269 and Tubastatin A decreased the level of ArgBP2 expression after LPS stimulation suggesting that both Class IIa and IIb HDACs regulate endothelial permeability via ArgBP2-dependent mechanism. Collectively, our data strongly suggest that Class IIa HDACs are involved in LPS-induced ALI in vitro and in vivo via specific mechanism which involved contractile responses, but not microtubule reorganization.
Collapse
Affiliation(s)
- Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Vijay Patel
- Department of Cardiothoracic Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary L. Meadows
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - David J. Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Yunchao Su
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
15
|
Wilkinson B, Coba MP. Molecular architecture of postsynaptic Interactomes. Cell Signal 2020; 76:109782. [PMID: 32941943 DOI: 10.1016/j.cellsig.2020.109782] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 01/02/2023]
Abstract
The postsynaptic density (PSD) plays an essential role in the organization of the synaptic signaling machinery. It contains a set of core scaffolding proteins that provide the backbone to PSD protein-protein interaction networks (PINs). These core scaffolding proteins can be seen as three principal layers classified by protein family, with DLG proteins being at the top, SHANKs along the bottom, and DLGAPs connecting the two layers. Early studies utilizing yeast two hybrid enabled the identification of direct protein-protein interactions (PPIs) within the multiple layers of scaffolding proteins. More recently, mass-spectrometry has allowed the characterization of whole interactomes within the PSD. This expansion of knowledge has further solidified the centrality of core scaffolding family members within synaptic PINs and provided context for their role in neuronal development and synaptic function. Here, we discuss the scaffolding machinery of the PSD, their essential functions in the organization of synaptic PINs, along with their relationship to neuronal processes found to be impaired in complex brain disorders.
Collapse
Affiliation(s)
- Brent Wilkinson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Marcelo P Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
16
|
Inaba Y, Chauhan V, van Loon AP, Choudhury LS, Sagasti A. Keratins and the plakin family cytolinker proteins control the length of epithelial microridge protrusions. eLife 2020; 9:58149. [PMID: 32894222 PMCID: PMC7535935 DOI: 10.7554/elife.58149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Actin filaments and microtubules create diverse cellular protrusions, but intermediate filaments, the strongest and most stable cytoskeletal elements, are not known to directly participate in the formation of protrusions. Here we show that keratin intermediate filaments directly regulate the morphogenesis of microridges, elongated protrusions arranged in elaborate maze-like patterns on the surface of mucosal epithelial cells. We found that microridges on zebrafish skin cells contained both actin and keratin filaments. Keratin filaments stabilized microridges, and overexpressing keratins lengthened them. Envoplakin and periplakin, plakin family cytolinkers that bind F-actin and keratins, localized to microridges, and were required for their morphogenesis. Strikingly, plakin protein levels directly dictate microridge length. An actin-binding domain of periplakin was required to initiate microridge morphogenesis, whereas periplakin-keratin binding was required to elongate microridges. These findings separate microridge morphogenesis into distinct steps, expand our understanding of intermediate filament functions, and identify microridges as protrusions that integrate actin and intermediate filaments.
Collapse
Affiliation(s)
- Yasuko Inaba
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | - Vasudha Chauhan
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | - Aaron Paul van Loon
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | - Lamia Saiyara Choudhury
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | - Alvaro Sagasti
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
17
|
Ding Y, Yang J, Chen P, Lu T, Jiao K, Tester DJ, Giudicessi JR, Jiang K, Ackerman MJ, Li Y, Wang DW, Lee H, Wang DW, Xu X. Knockout of SORBS2 Protein Disrupts the Structural Integrity of Intercalated Disc and Manifests Features of Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2020; 9:e017055. [PMID: 32808564 PMCID: PMC7660791 DOI: 10.1161/jaha.119.017055] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Background Sorbs2b (sorbin and SH3 domain-containing 2b) was recently identified as a cardiomyopathy gene from a zebrafish mutagenesis screen. However, cardiac functions of its mammalian ortholog remain elusive. Methods and Results We conducted a detailed expression and subcellular localization analysis of Sorbs2 ortholog in mice and a phenotypic characterization in Sorbs2 knockout mice. Sorbs2 is highly expressed in the mouse heart and encodes an adhesion junction/desmosome protein that is mainly localized to the intercalated disc. A mutation with near complete depletion of the Sorbs2 protein in mice results in phenotypes characteristic of human arrhythmogenic cardiomyopathy (ACM), including right ventricular dilation, right ventricular dysfunction, spontaneous ventricular tachycardia, and premature death. Sorbs2 is required to maintain the structural integrity of intercalated disc. Its absence resulted in profound cardiac electrical remodeling with impaired impulse conduction and action potential derangements. Targeted sequencing of human patients with ACM identified 2 rare splicing variants classified as likely pathogenic were in 2 unrelated individuals with ACM from a cohort of 59 patients with ACM. Conclusions The Sorbs2 knockout mouse manifests several key features reminiscent of human ACM. Although the candidacy of SORBS2 as a new ACM-susceptibility gene is supported by preliminary human genetics study, future validation in larger cohorts with ACM is needed.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Jingchun Yang
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Peng Chen
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tong Lu
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Kunli Jiao
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | | | | | - Kai Jiang
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Michael J. Ackerman
- Department of Cardiovascular Medicine (Division of Heart Rhythm Services)Mayo ClinicRochesterMN
- Pediatric and Adolescent Medicine (Division of Pediatric Cardiology)Mayo ClinicRochesterMN
- Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory)Mayo ClinicRochesterMN
| | - Yigang Li
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Dao Wu Wang
- State Key Laboratory of Reproductive MedicineClinical Center of Reproductive Medicine and Department of CardiologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - HoN‐chi Lee
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Dao Wen Wang
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaolei Xu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| |
Collapse
|
18
|
Chen J, Loukola A, Gillespie NA, Peterson R, Jia P, Riley B, Maes H, Dick DM, Kendler KS, Damaj MI, Miles MF, Zhao Z, Li MD, Vink JM, Minica CC, Willemsen G, Boomsma DI, Qaiser B, Madden PAF, Korhonen T, Jousilahti P, Hällfors J, Gelernter J, Kranzler HR, Sherva R, Farrer L, Maher B, Vanyukov M, Taylor M, Ware JJ, Munafò MR, Lutz SM, Hokanson JE, Gu F, Landi MT, Caporaso NE, Hancock DB, Gaddis NC, Baker TB, Bierut LJ, Johnson EO, Chenoweth M, Lerman C, Tyndale R, Kaprio J, Chen X. Genome-Wide Meta-Analyses of FTND and TTFC Phenotypes. Nicotine Tob Res 2020; 22:900-909. [PMID: 31294817 PMCID: PMC7249921 DOI: 10.1093/ntr/ntz099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/14/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION FTND (Fagerstrӧm test for nicotine dependence) and TTFC (time to smoke first cigarette in the morning) are common measures of nicotine dependence (ND). However, genome-wide meta-analysis for these phenotypes has not been reported. METHODS Genome-wide meta-analyses for FTND (N = 19,431) and TTFC (N = 18,567) phenotypes were conducted for adult smokers of European ancestry from 14 independent cohorts. RESULTS We found that SORBS2 on 4q35 (p = 4.05 × 10-8), BG182718 on 11q22 (p = 1.02 × 10-8), and AA333164 on 14q21 (p = 4.11 × 10-9) were associated with TTFC phenotype. We attempted replication of leading candidates with independent samples (FTND, N = 7010 and TTFC, N = 10 061), however, due to limited power of the replication samples, the replication of these new loci did not reach significance. In gene-based analyses, COPB2 was found associated with FTND phenotype, and TFCP2L1, RELN, and INO80C were associated with TTFC phenotype. In pathway and network analyses, we found that the interconnected interactions among the endocytosis, regulation of actin cytoskeleton, axon guidance, MAPK signaling, and chemokine signaling pathways were involved in ND. CONCLUSIONS Our analyses identified several promising candidates for both FTND and TTFC phenotypes, and further verification of these candidates was necessary. Candidates supported by both FTND and TTFC (CHRNA4, THSD7B, RBFOX1, and ZNF804A) were associated with addiction to alcohol, cocaine, and heroin, and were associated with autism and schizophrenia. We also identified novel pathways involved in cigarette smoking. The pathway interactions highlighted the importance of receptor recycling and internalization in ND. IMPLICATIONS Understanding the genetic architecture of cigarette smoking and ND is critical to develop effective prevention and treatment. Our study identified novel candidates and biological pathways involved in FTND and TTFC phenotypes, and this will facilitate further investigation of these candidates and pathways.
Collapse
Affiliation(s)
- Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV
| | - Anu Loukola
- Department of Public Health, University of Helsinki, Helsinki, FI, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Nathan A Gillespie
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Roseann Peterson
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Peilin Jia
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX
| | - Brien Riley
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Hermine Maes
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Daniella M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA
| | - Kenneth S Kendler
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| | - Michael F Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| | - Zhongming Zhao
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jacqueline M Vink
- Netherlands Twin Register, Department of Biological Psychology, VU University, the Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, the Netherlands
| | - Camelia C Minica
- Netherlands Twin Register, Department of Biological Psychology, VU University, the Netherlands
- Neuroscience Campus Amsterdam, the Netherlands
- EMGO+ Institute for Health and Care Research, VU Medical Center, Amsterdam, the Netherlands
| | - Gonneke Willemsen
- Netherlands Twin Register, Department of Biological Psychology, VU University, the Netherlands
- Neuroscience Campus Amsterdam, the Netherlands
- EMGO+ Institute for Health and Care Research, VU Medical Center, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Netherlands Twin Register, Department of Biological Psychology, VU University, the Netherlands
- Neuroscience Campus Amsterdam, the Netherlands
- EMGO+ Institute for Health and Care Research, VU Medical Center, Amsterdam, the Netherlands
| | - Beenish Qaiser
- Department of Public Health, University of Helsinki, Helsinki, FI, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | | | - Tellervo Korhonen
- Department of Public Health, University of Helsinki, Helsinki, FI, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Finland
| | | | - Jenni Hällfors
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Joel Gelernter
- Department of Psychiatry, Yale University, New Haven, CT
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Richard Sherva
- Section of Biomedical Genetics, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Lindsay Farrer
- Section of Biomedical Genetics, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Brion Maher
- Department of Mental Health, Johns Hopkins University, Baltimore, MD
| | - Michael Vanyukov
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Michelle Taylor
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, Bristol, BS, UK
| | - Jenifer J Ware
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, Bristol, BS, UK
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, Bristol, BS, UK
| | - Sharon M Lutz
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - John E Hokanson
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Fangyi Gu
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD
| | - Maria T Landi
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD
| | - Neil E Caporaso
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD
| | - Dana B Hancock
- Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC
| | - Nathan C Gaddis
- Research Computing Division, RTI International, Research Triangle Park, NC
| | - Timothy B Baker
- Center for Tobacco Research and Intervention, University of Wisconsin, Madison, WI
| | - Laura J Bierut
- Department of Psychiatry, Washington University, St. Louis, MO
| | - Eric O Johnson
- Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC
- Fellow Program, RTI International, Research Triangle Park, NC
| | - Meghan Chenoweth
- Centre for Addiction and Mental Health, and Departments of Pharmacology and Toxicology, and Psychiatry, University of Toronto, Toronto, Canada
| | - Caryn Lerman
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Rachel Tyndale
- Centre for Addiction and Mental Health, and Departments of Pharmacology and Toxicology, and Psychiatry, University of Toronto, Toronto, Canada
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki, FI, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Xiangning Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV
| |
Collapse
|
19
|
Cho WC, Jang JE, Kim KH, Yoo BC, Ku JL. SORBS1 serves a metastatic role via suppression of AHNAK in colorectal cancer cell lines. Int J Oncol 2020; 56:1140-1151. [PMID: 32319594 PMCID: PMC7115741 DOI: 10.3892/ijo.2020.5006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
Cbl-associated protein (CAP) is encoded by the sorbin and SH3 domain-containing 1 (SORBS1) gene. CAP has been reported to be associated with the actin cytoskeleton, receptor tyrosine kinase signaling and cell adhesion through interactions with various proteins. It may be hypothesized that SORBS1 has numerous unknown functions, which may include providing a favorable condition for metastasis. Although CAP has been demonstrated to possess a number of functions, the role of this protein has only been reported in metabolic signaling pathways and its function in cancer remains to be elucidated. In the present study, SORBS1 expression was detected in colorectal cancer cell lines divided into the primary group and the metastatic group by reverse transcription-quantitative PCR and western blot analysis. In addition, SORBS1 expression was manipulated by vector transfection and lentivirus transduction. The metastatic role of SORBS1, as determined by assessing its effects on cell proliferation and migration, was determined by colony formation assay, cell cycle analysis and Boyden chamber assay. To elucidate the SORBS1-binding protein, immunoprecipitation was performed. Co-localization of SORBS1 and AHNAK nucleoprotein (AHNAK) was identified by confocal microscopy. Notably, the protein expression levels of CAP were higher in SNU-769A and SW480 cells than in SNU-769B and SW620 cells. In addition, the number of colonies in the SORBS1-overexpressing group was significantly increased compared with that of the control group, as determined using the colony formation assay; the SORBS1 overexpression group formed >8-fold more colonies than the control group. The proliferative ability of the SORBS1 overexpression group was also significantly increased compared with the control group over the entire incubation period. Cell migration assays revealed that the number of migrated SORBS1-knockdown cells was reduced compared with the control in both HCT-116 and SNU-C4 cell lines; migration area was decreased to 31 and 26% in HCT-116 and SNU-C4 cell lines, respectively. Consequently, it was confirmed that SORBS1 could form a complex with AHNAK, which functions as a tumor suppressor through inhibition of phosphorylated-ERK and Rho-associated coiled-coil containing protein kinase 1. In conclusion, SORBS1 may serve a crucial role in cancer growth and migration via inhibition of AHNAK expression.
Collapse
Affiliation(s)
- Woo-Cheol Cho
- Department of Biomedical Sciences, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jee-Eun Jang
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyung-Hee Kim
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi 10408, Republic of Korea
| | - Byong-Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi 10408, Republic of Korea
| | - Ja-Lok Ku
- Department of Biomedical Sciences, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
20
|
Kabrawala S, Zimmer MD, Campellone KG. WHIMP links the actin nucleation machinery to Src-family kinase signaling during protrusion and motility. PLoS Genet 2020; 16:e1008694. [PMID: 32196488 PMCID: PMC7112243 DOI: 10.1371/journal.pgen.1008694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/01/2020] [Accepted: 02/22/2020] [Indexed: 12/27/2022] Open
Abstract
Cell motility is governed by cooperation between the Arp2/3 complex and nucleation-promoting factors from the Wiskott-Aldrich Syndrome Protein (WASP) family, which together assemble actin filament networks to drive membrane protrusion. Here we identify WHIMP (WAVE Homology In Membrane Protrusions) as a new member of the WASP family. The Whimp gene is encoded on the X chromosome of a subset of mammals, including mice. Murine WHIMP promotes Arp2/3-dependent actin assembly, but is less potent than other nucleation factors. Nevertheless, WHIMP-mediated Arp2/3 activation enhances both plasma membrane ruffling and wound healing migration, whereas WHIMP depletion impairs protrusion and slows motility. WHIMP expression also increases Src-family kinase activity, and WHIMP-induced ruffles contain the additional nucleation-promoting factors WAVE1, WAVE2, and N-WASP, but not JMY or WASH. Perturbing the function of Src-family kinases, WAVE proteins, or Arp2/3 complex inhibits WHIMP-driven ruffling. These results suggest that WHIMP-associated actin assembly plays a direct role in membrane protrusion, but also results in feedback control of tyrosine kinase signaling to modulate the activation of multiple WASP-family members. The actin cytoskeleton is a collection of protein polymers that assemble and disassemble within cells at specific times and locations. Sophisticated cytoskeletal regulators called nucleation-promoting factors ensure that actin polymerizes when and where it is needed, and many of these factors are members of the Wiskott-Aldrich Syndrome Protein (WASP) family. Several of the 8 known WASP-family proteins function in cell motility, but how the different factors collaborate with one another is not well understood. In this study, we identified WHIMP, a new WASP-family member that is encoded on the X chromosome of a variety of mammals. In mouse cells, WHIMP enhances cell motility by assembling actin filaments that push the plasma membrane forward. Unexpectedly, WHIMP also activates tyrosine kinases, enzymes that stimulate multiple WASP-family members during motility. Our results open new avenues of research into how nucleation factors cooperate during movement and how the molecular activities that underlie motility differ in distinct cell types and organisms.
Collapse
Affiliation(s)
- Shail Kabrawala
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Margaret D. Zimmer
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
21
|
Jie R, Zhu P, Zhong J, Zhang Y, Wu H. LncRNA KCNQ1OT1 affects cell proliferation, apoptosis and fibrosis through regulating miR-18b-5p/SORBS2 axis and NF-ĸB pathway in diabetic nephropathy. Diabetol Metab Syndr 2020; 12:77. [PMID: 32905431 PMCID: PMC7469295 DOI: 10.1186/s13098-020-00585-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It has been reported that long non-coding RNAs (lncRNAs) play vital roles in diabetic nephropathy (DN). Our study aims to research the function of lncRNA KCNQ1OT1 in DN cells and the molecular mechanism. METHODS Human glomerular mesangial cells (HGMCs) and human renal glomerular endothelial cells (HRGECs) were cultured in high glucose (30 mM) condition as models of DN cells. KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) and miR-18b-5p levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mRNA and protein levels of Sorbin and SH3 domain-containing protein 2 (SORBS2), Type IV collagen (Col-4), fibronectin (FN), transcriptional regulatory factor-beta 1 (TGF-β1), Twist, NF-κB and STAT3 were measured by qRT-PCR and western blot. Cell viability was detected by cell counting kit-8 (CCK-8) assay for selecting the proper concentration of glucose treatment. Additionally, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and flow cytometry assay were employed to determine cell proliferation and apoptosis, respectively. The targets of KCNQ1OT1 was predicted by online software and confirmed by dual-luciferase reporter assay. RESULTS KCNQ1OT1 and SORBS2 were elevated in DN. Both knockdown of KCNQ1OT1 and silencing of SORBS2 restrained proliferation and fibrosis and induced apoptosis in DN cells. Besides, Overexpression of SORBS2 restored the KCNQ1OT1 knockdown-mediate effects on proliferation, apoptosis and fibrosis in DN cells. In addition, miR-18b-5p served as a target of KCNQ1OT1 as well as targeted SORBS2. KCNQ1OT1 knockdown repressed NF-ĸB pathway. CONCLUSION KCNQ1OT1 regulated DN cells proliferation, apoptosis and fibrosis via KCNQ1OT1/miR-18b-5p/SORBS2 axis and NF-ĸB pathway.
Collapse
Affiliation(s)
- Ran Jie
- Department of Endocrinology, First People’s Hospital of Jingzhou, Shashi District, No. 8 Hangkong Road, Jingzhou, 434000 Hubei China
| | - Pengpeng Zhu
- Department of Anesthesiology, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Jiao Zhong
- Health Management Center, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Yan Zhang
- Department of Gastroenterology, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Hongyan Wu
- Department of Endocrinology, First People’s Hospital of Jingzhou, Shashi District, No. 8 Hangkong Road, Jingzhou, 434000 Hubei China
| |
Collapse
|
22
|
Schlaweck AE, Tazi-Ahnini R, Ü. Basmanav FB, Mohungoo J, Pasternack-Ziach SM, Mattheisen M, Oprisoreanu AM, Humbatova A, Wolf S, Messenger A, Betz RC. Autosomal-dominant hypotrichosis with woolly hair: Novel gene locus on chromosome 4q35.1-q35.2. PLoS One 2019; 14:e0225943. [PMID: 31790498 PMCID: PMC6886801 DOI: 10.1371/journal.pone.0225943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 11/15/2019] [Indexed: 11/18/2022] Open
Abstract
Hypotrichosis simplex (HS) with and without woolly hair (WH) comprises a group of rare, monogenic disorders of hair loss. Patients present with a diffuse loss of scalp and/or body hair, which usually begins in early childhood and progresses into adulthood. Some of the patients also show hair that is tightly curled. Approximately 10 genes for autosomal recessive and autosomal dominant forms of HS have been identified in the last decade, among them five genes for the dominant form. We collected blood and buccal samples from 17 individuals of a large British family with HS and WH. After having sequenced all known dominant genes for HS in this family without the identification of any disease causing mutation, we performed a genome-wide scan, using the HumanLinkage-24 BeadChip, followed by a classical linkage analysis; and whole exome-sequencing (WES). Evidence for linkage was found for a region on chromosome 4q35.1-q35.2 with a maximum LOD score of 3.61. WES led to the identification of a mutation in the gene SORBS2, encoding sorbin and SH3 domain containing 2. Unfortunately, we could not find an additional mutation in any other patient/family with HS; and in cell culture, we could not observe any difference between cloned wildtype and mutant SORBS2 using western blotting and immunofluorescence analyses. Therefore, at present, SORBS2 cannot be considered a definite disease gene for this phenotype. However, the locus on chromosome 4q is a robust and novel finding for hypotrichosis with woolly hair. Further fine mapping and sequencing efforts are therefore warranted in order to confirm SORBS2 as a plausible HS disease gene.
Collapse
Affiliation(s)
- Annika E. Schlaweck
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Rachid Tazi-Ahnini
- Department of Infection, Immunity, and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - F. Buket Ü. Basmanav
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Javed Mohungoo
- Department of Dermatology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Sandra M. Pasternack-Ziach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Manuel Mattheisen
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Ana-Maria Oprisoreanu
- Department of Neuropathology and Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Aytaj Humbatova
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Sabrina Wolf
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Andrew Messenger
- Department of Dermatology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Regina C. Betz
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
23
|
Zanni G, Nardella M, Barresi S, Bellacchio E, Niceta M, Ciolfi A, Pro S, D'Arrigo S, Tartaglia M, Bertini E. De novo p.T362R mutation in MORC2 causes early onset cerebellar ataxia, axonal polyneuropathy and nocturnal hypoventilation. Brain 2019; 140:e34. [PMID: 28402445 DOI: 10.1093/brain/awx083] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Ginevra Zanni
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marta Nardella
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabina Barresi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Marcello Niceta
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefano Pro
- Department of Neurosciences, Unit of Neurology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefano D'Arrigo
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Enrico Bertini
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
24
|
Lima Caldeira G, Peça J, Carvalho AL. New insights on synaptic dysfunction in neuropsychiatric disorders. Curr Opin Neurobiol 2019; 57:62-70. [PMID: 30743178 DOI: 10.1016/j.conb.2019.01.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 01/01/2023]
Abstract
Growing evidence implicates synaptic proteins in the pathogenesis of neuropsychiatric disorders such as autism spectrum disorder (ASD), intellectual disability (ID) and schizophrenia. In fact, mutations in genes encoding synaptic proteins are enriched and overlap among different conditions highlighting the complex and pleiotropic nature of these disorders. In this review, we discuss recently described candidate genes that affect excitatory synapse function and result in changes in spine number and morphology. Spine pathology has been observed in several animal models of disease and in human brain post-mortem samples from ID, ASD, and schizophrenia patients. Recent data point to convergent mechanisms, such as dysregulation of the actin cytoskeleton and dysfunction of microglia synaptic remodeling, underlying dendritic spine dysgenesis. Interestingly, the reversion of important pathologic features, including spine abnormalities, has been observed in adult animal models of neuropsychiatric disorders, suggesting that therapies may not be restricted to a specific developmental window. Shedding light on the specific mechanisms impacted in neuropsychiatric disorders will undeniably contribute to the development of more directed and personalized therapies.
Collapse
Affiliation(s)
- Gladys Lima Caldeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIUC-Interdisciplinary Research Institute, University of Coimbra, 3030-789 Coimbra, Portugal
| | - João Peça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIUC-Interdisciplinary Research Institute, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
25
|
Pan Z, Ding Q, Guo Q, Guo Y, Wu L, Wu L, Tang M, Yu H, Zhou F. MORC2, a novel oncogene, is upregulated in liver cancer and contributes to proliferation, metastasis and chemoresistance. Int J Oncol 2018; 53:59-72. [PMID: 29620211 PMCID: PMC5958890 DOI: 10.3892/ijo.2018.4333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
Microrchidia 2 (MORC2) is important in DNA damage repair and lipogenesis, however, the clinical and functional role of MORC2 in liver cancer remains to be fully elucidated. The aim the present study was to clarify the role of MORC2 in liver cancer. Expression profile analysis, immunohistochemical staining, reverse transcription-quantitative polymerase chain reaction analysis and western blot analysis were performed to evaluate the levels of MORC2 in liver cancer patient specimens and cell lines; subsequently the expression of MORC2 was suppressed or increased in liver cancer cells and the effects of MORC2 on the cancerous transformation of liver cancer cells were examined in vitro and in vivo. MORC2 was upregulated in liver cancer tissues, and the upregulation was associated with certain clinicopathologic features of patients with liver cancer. MORC2 knockdown caused marked inhibition of liver cancer cell proliferation and clonogenicity, whereas the overexpression of MORC2 substantially promoted liver cancer cell proliferation. In addition, the knockdown of MORC2 inhibited the migratory and invasive ability of liver cancer cells, whereas increased migration and invasion rates were observed in cells with ectopic expression of MORC2. In a model of nude mice, the overexpression of MORC2 promoted tumorigenicity and markedly enhanced pulmonary metastasis of liver cancer. Furthermore, MORC2 regulated apoptosis and its expression level had an effect on the sensitivity of liver cancer cells to doxorubicin, 5-fluorouracil and cisplatin. Mechanically, MORC2 modulated the mitochondrial apoptotic pathway, possibly in a p53-dependent manner, and its dysregulation also resulted in the abnormal activation of the Hippo pathway. For the first time, to the best of our knowledge, the present study confirmed that MORC2 was a novel oncogene in liver cancer. These results provide useful insight into the mechanism underlying the tumorigenesis and progression of liver cancer, and offers clues into potential novel liver cancer therapies.
Collapse
Affiliation(s)
- Zhihong Pan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Qianshan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Qian Guo
- Hepatic Disease Institute, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061
| | - Yong Guo
- College of Biotechnology, Guilin Medical University, Guilin, Guanxi 541004
| | - Lianlian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Lu Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Meng Tang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
26
|
nArgBP2-SAPAP-SHANK, the core postsynaptic triad associated with psychiatric disorders. Exp Mol Med 2018; 50:1-9. [PMID: 29628500 PMCID: PMC5938024 DOI: 10.1038/s12276-017-0018-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Despite the complex genetic architecture, a broad spectrum of psychiatric disorders can still be caused by mutation(s) in the same gene. These disorders are interrelated with overlapping causative mechanisms including variations in the interaction among the risk-associated proteins that may give rise to the specific spectrum of each disorder. Additionally, multiple lines of evidence implicate an imbalance between excitatory and inhibitory neuronal activity (E/I imbalance) as the shared key etiology. Thus, understanding the molecular mechanisms underlying E/I imbalance provides essential insight into the etiology of these disorders. One important class of candidate risk genes is the postsynaptic scaffolding proteins, such as nArgBP2, SAPAP, and SHANK that regulate the actin cytoskeleton in dendritic spines of excitatory synapses. This review will cover and discuss recent studies that examined how these proteins, especially nArgBP2, are associated with psychiatric disorders. Next, we propose a possibility that variations in the interaction among these proteins in a specific brain region might contribute to the onset of diverse phenotypes of psychiatric disorders. The assembly of scaffolding proteins, key regulators of many signaling pathways, found in the brain’s synapses underpin a diverse range of neuropsychiatric disorders. Sunghoe Chang and colleagues from Seoul National University, South Korea, review how these postsynaptic proteins regulate the cellular cytoskeleton in nerve cell protrusions to maintain the balance between excitatory and inhibitory inputs in the brain. They discuss how perturbations in three particular proteins can cause an imbalance in synaptic signals that leads to conditions such as bipolar disorder, schizophrenia and autism. The authors propose that these proteins form a “core scaffolding triad” and interact in different ways to cause different mental illnesses. Dysregulation of these proteins could explain how mutations in the same genes, depending on whether they boost or decrease gene expression, contribute to the onset of diverse psychiatric disorders.
Collapse
|
27
|
Loveless T, Qadota H, Benian GM, Hardin J. Caenorhabditis elegans SORB-1 localizes to integrin adhesion sites and is required for organization of sarcomeres and mitochondria in myocytes. Mol Biol Cell 2017; 28:3621-3633. [PMID: 28978740 PMCID: PMC5706990 DOI: 10.1091/mbc.e16-06-0455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 01/07/2023] Open
Abstract
We have identified and characterized sorb-1, the only sorbin and SH3 domain-containing protein family member in Caenorhabditis elegans SORB-1 is strongly localized to integrin adhesion complexes in larvae and adults, including adhesion plaques and dense bodies (Z-disks) of striated muscles and attachment plaques of smooth muscles. SORB-1 is recruited to the actin-binding, membrane-distal regions of dense bodies via its C-terminal SH3 domains in an ATN-1(α-actinin)- and ALP-1(ALP/Enigma)-dependent manner, where it contributes to the organization of sarcomeres. SORB-1 is also found in other tissues known to be under mechanical stress, including stress fibers in migratory distal tip cells and the proximal gonad sheath, where it becomes enriched in response to tissue distention. We provide evidence for a novel role for sorbin family proteins: SORB-1 is required for normal positioning of the mitochondrial network in muscle cells. Finally, we demonstrate that SORB-1 interacts directly with two other dense body components, DEB-1(vinculin) and ZYX-1(zyxin). This work establishes SORB-1 as a bona fide sorbin family protein-one of the late additions to the dense body complex and a conserved regulator of body wall muscle sarcomere organization and organelle positioning.
Collapse
Affiliation(s)
- Timothy Loveless
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Hiroshi Qadota
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Guy M Benian
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Jeff Hardin
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
28
|
Fredriksson-Lidman K, Van Itallie CM, Tietgens AJ, Anderson JM. Sorbin and SH3 domain-containing protein 2 (SORBS2) is a component of the acto-myosin ring at the apical junctional complex in epithelial cells. PLoS One 2017; 12:e0185448. [PMID: 28961272 PMCID: PMC5621683 DOI: 10.1371/journal.pone.0185448] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
SORBS2 is a scaffolding protein associated with Abl/Arg non-receptor tyrosine kinase pathways and is known to interact with actin and several other cytoskeletal proteins in various cell types. Previous BioID proximity labeling of tight and adherens junction proteins suggested that SORBS2 is a component of the apical junction complex of epithelial cells. We asked whether SORBS2 plays a previously unappreciated role in controlling perijunctional actin and tight junction barrier function. Using super resolution imaging we confirmed that SORBS2 is localized at the apical junction complex but farther from the membrane than ZO-1 and located partially overlapping both the tight- and adherens junctions with a periodic concentration that alternates with myosin IIB in polarized epithelial cells. Overexpression of GFP-SORBS2 recruited alpha-actinin, vinculin and N-WASP, and possibly CIP4 to cellular junctions. However, CRISPR-Cas9 knock-out of SORBS2 did not alter the localization- or immunofluorescent staining intensity of these or several other junctional- and cytoskeletal proteins. SORBS2 knock-out also did not affect the barrier function as measured by TER and dextran flux; nor did it change actin-dependent junction re-assembly as measured by Ca2+-switch and Latrunculin-B wash-out assays. The kinetics of HGF-induced cell scattering and wound healing, and dextran flux increase induced by PDGF also were unaffected by SORBS2 knock-out. SORBS2 concentrates with apical junctional actin that accumulates in response to knock-down of ZO-1 and ZO-2. In spite of our finding that SORBS2 is clearly a component of the apical junction complex, it does not appear to be required for either normal tight- or adherens junction assembly, structure or function or for growth factor-mediated changes in tight junction dynamics.
Collapse
Affiliation(s)
- Karin Fredriksson-Lidman
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Christina M. Van Itallie
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amber J. Tietgens
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James M. Anderson
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
29
|
Ichikawa T, Kita M, Matsui TS, Nagasato AI, Araki T, Chiang SH, Sezaki T, Kimura Y, Ueda K, Deguchi S, Saltiel AR, Kioka N. Vinexin family (SORBS) proteins play different roles in stiffness-sensing and contractile force generation. J Cell Sci 2017; 130:3517-3531. [PMID: 28864765 DOI: 10.1242/jcs.200691] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 08/10/2017] [Indexed: 12/17/2022] Open
Abstract
Vinexin, c-Cbl associated protein (CAP) and Arg-binding protein 2 (ArgBP2) constitute an adaptor protein family called the vinexin (SORBS) family that is targeted to focal adhesions (FAs). Although numerous studies have focused on each of the SORBS proteins and partially elucidated their involvement in mechanotransduction, a comparative analysis of their function has not been well addressed. Here, we established mouse embryonic fibroblasts that individually expressed SORBS proteins and analysed their functions in an identical cell context. Both vinexin-α and CAP co-localized with vinculin at FAs and promoted the appearance of vinculin-rich FAs, whereas ArgBP2 co-localized with α-actinin at the proximal end of FAs and punctate structures on actin stress fibers (SFs), and induced paxillin-rich FAs. Furthermore, both vinexin-α and CAP contributed to extracellular matrix stiffness-dependent vinculin behaviors, while ArgBP2 stabilized α-actinin on SFs and enhanced intracellular contractile forces. These results demonstrate the differential roles of SORBS proteins in mechanotransduction.
Collapse
Affiliation(s)
- Takafumi Ichikawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Masahiro Kita
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tsubasa S Matsui
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tomohiko Araki
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan
| | - Shian-Huey Chiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takuhito Sezaki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Yasuhisa Kimura
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Shinji Deguchi
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Alan R Saltiel
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan .,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| |
Collapse
|
30
|
Kleinert M, Parker BL, Fritzen AM, Knudsen JR, Jensen TE, Kjøbsted R, Sylow L, Ruegg M, James DE, Richter EA. Mammalian target of rapamycin complex 2 regulates muscle glucose uptake during exercise in mice. J Physiol 2017; 595:4845-4855. [PMID: 28464351 DOI: 10.1113/jp274203] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/26/2017] [Indexed: 01/04/2023] Open
Abstract
KEY POINTS Exercise is a potent physiological stimulus to clear blood glucose from the circulation into skeletal muscle. The mammalian target of rapamycin complex 2 (mTORC2) is an important regulator of muscle glucose uptake in response to insulin stimulation. Here we report for the first time that the activity of mTORC2 in mouse muscle increases during exercise. We further show that glucose uptake during exercise is decreased in mouse muscle that lacks mTORC2 activity. We also provide novel identifications of new mTORC2 substrates during exercise in mouse muscle. ABSTRACT Exercise increases glucose uptake into insulin-resistant muscle. Thus, elucidating the exercise signalling network in muscle may uncover new therapeutic targets. The mammalian target of rapamycin complex 2 (mTORC2), a regulator of insulin-controlled glucose uptake, has been reported to interact with ras-related C3 botulinum toxin substrate 1 (Rac1), which plays a role in exercise-induced glucose uptake in muscle. Therefore, we tested the hypothesis that mTORC2 activity is necessary for muscle glucose uptake during treadmill exercise. We used mice that specifically lack mTORC2 signalling in muscle by deletion of the obligatory mTORC2 component Rictor (Ric mKO). Running capacity and running-induced changes in blood glucose, plasma lactate and muscle glycogen levels were similar in wild-type (Ric WT) and Ric mKO mice. At rest, muscle glucose uptake was normal, but during running muscle glucose uptake was reduced by 40% in Ric mKO mice compared to Ric WT mice. Running increased muscle phosphorylated 5' AMP-activated protein kinase (AMPK) similarly in Ric WT and Ric mKO mice, and glucose transporter type 4 (GLUT4) and hexokinase II (HKII) protein expressions were also normal in Ric mKO muscle. The mTORC2 substrate, phosphorylated protein kinase C α (PKCα), and the mTORC2 activity readout, phosphorylated N-myc downstream regulated 1 (NDRG1) protein increased with running in Ric WT mice, but were not altered by running in Ric mKO muscle. Quantitative phosphoproteomics uncovered several additional potential exercise-dependent mTORC2 substrates, including contractile proteins, kinases, transcriptional regulators, actin cytoskeleton regulators and ion-transport proteins. Our study suggests that mTORC2 is a component of the exercise signalling network that regulates muscle glucose uptake and we provide a resource of new potential members of the mTORC2 signalling network.
Collapse
Affiliation(s)
- Maximilian Kleinert
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Germany
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, Australia
| | - Andreas M Fritzen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Lykke Sylow
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Markus Ruegg
- Biozentrum, University of Basel, Basel, Switzerland
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Marcogliese PC, Abuaish S, Kabbach G, Abdel-Messih E, Seang S, Li G, Slack RS, Haque ME, Venderova K, Park DS. LRRK2(I2020T) functional genetic interactors that modify eye degeneration and dopaminergic cell loss in Drosophila. Hum Mol Genet 2017; 26:1247-1257. [PMID: 28158614 DOI: 10.1093/hmg/ddx030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/19/2017] [Indexed: 11/14/2022] Open
Abstract
Progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta is the primary cause for motor symptoms observed in Parkinson's disease (PD). Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most commonly linked contributor to familial PD. LRRK2 is suggested to be involved in a wide variety of cellular processes, but deciphering its role in the pathogenesis of PD has been difficult. Modelling PD in rodents has been a persistent challenge for the field. However, the fruit fly has been exploited to recapitulate PD gene related dopaminergic cell loss. Using the GAL4-UAS system and established models of hLRRK2 induced eye degeneration in Drosophila, we conducted an unbiased suppressor/enhancer screen to uncover genetic modifiers of LRRK2. We have identified 36 candidate interactors that modify LRRK2 induced toxicity in the Drosophila eye. Importantly, we determined that a subset of these interactors also modified hLRRK2(I2020T) induced dopaminergic neuronal loss in the fly brain and uncovered 16 candidates that modify dopaminergic cell loss. Our results suggest LRRK2 may be involved in a wide variety of cellular processes and the results from this screen provide an important genetic resource for further evaluation of LRRK2 function.
Collapse
Affiliation(s)
- Paul C Marcogliese
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Sameera Abuaish
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Ghassan Kabbach
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Elizabeth Abdel-Messih
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Sarah Seang
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Gang Li
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - M Emdadul Haque
- Department of Biochemistry, College of Medicine and Health Science, United Arab Emirates University, Al Ain, UAE
| | - Katerina Venderova
- Department of Biopharmaceutical Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - David S Park
- Department of Cellular and Molecular Medicine.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
32
|
nArgBP2 regulates excitatory synapse formation by controlling dendritic spine morphology. Proc Natl Acad Sci U S A 2016; 113:6749-54. [PMID: 27226294 DOI: 10.1073/pnas.1600944113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neural Abelson-related gene-binding protein 2 (nArgBP2) was originally identified as a protein that directly interacts with synapse-associated protein 90/postsynaptic density protein 95-associated protein 3 (SAPAP3), a postsynaptic scaffolding protein critical for the assembly of glutamatergic synapses. Although genetic deletion of nArgBP2 in mice leads to manic/bipolar-like behaviors resembling many aspects of symptoms in patients with bipolar disorder, the actual function of nArgBP2 at the synapse is completely unknown. Here, we found that the knockdown (KD) of nArgBP2 by specific small hairpin RNAs (shRNAs) resulted in a dramatic change in dendritic spine morphology. Reintroducing shRNA-resistant nArgBP2 reversed these defects. In particular, nArgBP2 KD impaired spine-synapse formation such that excitatory synapses terminated mostly at dendritic shafts instead of spine heads in spiny neurons, although inhibitory synapse formation was not affected. nArgBP2 KD further caused a marked increase of actin cytoskeleton dynamics in spines, which was associated with increased Wiskott-Aldrich syndrome protein-family verprolin homologous protein 1 (WAVE1)/p21-activated kinase (PAK) phosphorylation and reduced activity of cofilin. These effects of nArgBP2 KD in spines were rescued by inhibiting PAK or activating cofilin combined with sequestration of WAVE. Together, our results suggest that nArgBP2 functions to regulate spine morphogenesis and subsequent spine-synapse formation at glutamatergic synapses. They also raise the possibility that the aberrant regulation of synaptic actin filaments caused by reduced nArgBP2 expression may contribute to the manifestation of the synaptic dysfunction observed in manic/bipolar disorder.
Collapse
|
33
|
Zhang Q, Gao X, Li C, Feliciano C, Wang D, Zhou D, Mei Y, Monteiro P, Anand M, Itohara S, Dong X, Fu Z, Feng G. Impaired Dendritic Development and Memory in Sorbs2 Knock-Out Mice. J Neurosci 2016; 36:2247-60. [PMID: 26888934 PMCID: PMC4756157 DOI: 10.1523/jneurosci.2528-15.2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/27/2022] Open
Abstract
Intellectual disability is a common neurodevelopmental disorder characterized by impaired intellectual and adaptive functioning. Both environmental insults and genetic defects contribute to the etiology of intellectual disability. Copy number variations of SORBS2 have been linked to intellectual disability. However, the neurobiological function of SORBS2 in the brain is unknown. The SORBS2 gene encodes ArgBP2 (Arg/c-Abl kinase binding protein 2) protein in non-neuronal tissues and is alternatively spliced in the brain to encode nArgBP2 protein. We found nArgBP2 colocalized with F-actin at dendritic spines and growth cones in cultured hippocampal neurons. In the mouse brain, nArgBP2 was highly expressed in the cortex, amygdala, and hippocampus, and enriched in the outer one-third of the molecular layer in dentate gyrus. Genetic deletion of Sorbs2 in mice led to reduced dendritic complexity and decreased frequency of AMPAR-miniature spontaneous EPSCs in dentate gyrus granule cells. Behavioral characterization revealed that Sorbs2 deletion led to a reduced acoustic startle response, and defective long-term object recognition memory and contextual fear memory. Together, our findings demonstrate, for the first time, an important role for nArgBP2 in neuronal dendritic development and excitatory synaptic transmission, which may thus inform exploration of neurobiological basis of SORBS2 deficiency in intellectual disability. SIGNIFICANCE STATEMENT Copy number variations of the SORBS2 gene are linked to intellectual disability, but the neurobiological mechanisms are unknown. We found that nArgBP2, the only neuronal isoform encoded by SORBS2, colocalizes with F-actin at neuronal dendritic growth cones and spines. nArgBP2 is highly expressed in the cortex, amygdala, and dentate gyrus in the mouse brain. Genetic deletion of Sorbs2 in mice leads to impaired dendritic complexity and reduced excitatory synaptic transmission in dentate gyrus granule cells, accompanied by behavioral deficits in acoustic startle response and long-term memory. This is the first study of Sorbs2 function in the brain, and our findings may facilitate the study of neurobiological mechanisms underlying SORBS2 deficiency in the development of intellectual disability.
Collapse
Affiliation(s)
- Qiangge Zhang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Xian Gao
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Chenchen Li
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Catia Feliciano
- Champalimaud Neuroscience Programme, Champalimaud Center for the Unknown, Lisbon 1400-038, Portugal
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Dingxi Zhou
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, School of Life Sciences, Peking University, Beijing 100871, China, and
| | - Yuan Mei
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Patricia Monteiro
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Michelle Anand
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Shigeyoshi Itohara
- Laboratory of Behavioral Genetics, RIKEN Brain Science Institute, Wako 351-0198, Japan
| | - Xiaowei Dong
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Zhanyan Fu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142,
| |
Collapse
|
34
|
Brüser L, Bogdan S. Molecular Control of Actin Dynamics In Vivo: Insights from Drosophila. Handb Exp Pharmacol 2016; 235:285-310. [PMID: 27757759 DOI: 10.1007/164_2016_33] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The actin cytoskeleton provides mechanical support for cells and generates forces to drive cell shape changes and cell migration in morphogenesis. Molecular understanding of actin dynamics requires a genetically traceable model system that allows interdisciplinary experimental approaches to elucidate the regulatory network of cytoskeletal proteins in vivo. Here, we will discuss some examples of how advances in Drosophila genetics and high-resolution imaging techniques contribute to the discovery of new actin functions, signaling pathways, and mechanisms of actin regulation in vivo.
Collapse
Affiliation(s)
- Lena Brüser
- Institute for Neurobiology, University of Muenster, Badestrasse 9, 48149, Muenster, Germany
| | - Sven Bogdan
- Institute for Neurobiology, University of Muenster, Badestrasse 9, 48149, Muenster, Germany.
| |
Collapse
|
35
|
Tong Y, Li Y, Gu H, Wang C, Liu F, Shao Y, Li J, Cao L, Li F. Microchidia protein 2, MORC2, downregulates the cytoskeleton adapter protein, ArgBP2, via histone methylation in gastric cancer cells. Biochem Biophys Res Commun 2015; 467:821-7. [PMID: 26476214 DOI: 10.1016/j.bbrc.2015.10.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 10/10/2015] [Indexed: 11/18/2022]
Abstract
ArgBP2 is an adapter protein that plays an important role in actin-dependent processes such as cell adhesion and migration. However, its function and regulation mechanisms in gastric cancer have not yet been investigated. Here, we showed the low expression of ArgBP2 mRNA level in gastric tumor samples and its repressive function in the proliferation, migration, and invasion of gastric cancer cells. Then, we cloned and identified ArgBP2 promoter and verified that MORC2 bound to the promoter. Moreover, we demonstrated that MORC2 enhanced the recruitment of EZH2, which promoted the tri-methylation of H3K27, leading to the transcriptional repression of ArgBP2. Our results might thus contribute to understanding the molecular mechanisms of ArgBP2 regulation and suggesting ArgBP2 as a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yuxin Tong
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hui Gu
- Department of Key Laboratory of Health Ministry for Congenital Malformation Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Chunyu Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Funan Liu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yangguang Shao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Jiabin Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Liu Cao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
36
|
Pur-alpha functionally interacts with FUS carrying ALS-associated mutations. Cell Death Dis 2015; 6:e1943. [PMID: 26492376 PMCID: PMC4632316 DOI: 10.1038/cddis.2015.295] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder due to motor neuron loss. Fused in sarcoma (FUS) protein carrying ALS-associated mutations localizes to stress granules and causes their coalescence into larger aggregates. Here we show that Pur-alpha physically interacts with mutated FUS in an RNA-dependent manner. Pur-alpha colocalizes with FUS carrying mutations in stress granules of motoneuronal cells differentiated from induced pluripotent stem cells and that are derived from ALS patients. We observe that both Pur-alpha and mutated FUS upregulate phosphorylation of the translation initiation factor eukaryotic translation initiation factor 2 alpha and consistently inhibit global protein synthesis. In vivo expression of Pur-alpha in different Drosophila tissues significatively exacerbates the neurodegeneration caused by mutated FUS. Conversely, the downregulation of Pur-alpha in neurons expressing mutated FUS significatively improves fly climbing activity. All these findings suggest that Pur-alpha, through the control of mRNA translation, might be involved in the pathogenesis of ALS associated with the mutation of FUS, and that an alteration of protein synthesis may be directly implicated in the disease. Finally, in vivo RNAi-mediated ablation of Pur-alpha produced locomotion defects in Drosophila, indicating a pivotal role for this protein in the motoneuronal function.
Collapse
|
37
|
Choi SY, Han K. Emerging role of synaptic actin-regulatory pathway in the pathophysiology of mood disorders. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2015.1086435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
38
|
Lee JH, Cheng R, Vardarajan B, Lantigua R, Reyes-Dumeyer D, Ortmann W, Graham RR, Bhangale T, Behrens TW, Medrano M, Jiménez-Velázquez IZ, Mayeux R. Genetic Modifiers of Age at Onset in Carriers of the G206A Mutation in PSEN1 With Familial Alzheimer Disease Among Caribbean Hispanics. JAMA Neurol 2015; 72:1043-51. [PMID: 26214276 PMCID: PMC5010776 DOI: 10.1001/jamaneurol.2015.1424] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE The present study identified potential genetic modifiers that may delay or accelerate age at onset of familial Alzheimer disease (AD) by examining age at onset in PSEN1 mutation carrier families, and further investigation of these modifiers may provide insight into the pathobiology of AD and potential therapeutic measures. OBJECTIVE To identify genetic variants that modify age at onset of AD. DESIGN, SETTING, AND PARTICIPANTS Using a subset of Caribbean Hispanic families that carry the PSEN1 p.G206A mutation, we performed a 2-stage genome study. The mutation carrier families from an ongoing genetic study served as a discovery set, and the cohort of those with LOAD served as a confirmation set. To identify candidate loci, we performed linkage analysis using 5 p.G206A carrier families (n = 56), and we also performed whole-exome association analysis using 31 p.G206A carriers from 26 families. To confirm the genetic modifiers identified from the p.G206A carrier families, we analyzed the GWAS data for 2888 elderly individuals with LOAD. All study participants were Caribbean Hispanics. MAIN OUTCOMES AND MEASURES Age at onset of AD. RESULTS Linkage analysis of AD identified the strongest linkage support at 4q35 (LOD [logarithm of odds] score, 3.69), and the GWAS of age at onset identified variants on 1p13.1, 2q13, 4q25, and 17p11. In the confirmation stage, genewise analysis identified SNX25, PDLIM3, and 3 SH3 domain genes (SORBS2, SH3RF3, and NPHP1) to be significantly associated with LOAD. Subsequent allelic association analysis confirmed SNX25, PDLIM3, and SORBS2 as genetic modifiers of age at onset of EOAD and LOAD and provided modest support for SH3RF3 and NPHP1. CONCLUSIONS AND RELEVANCE Our 2-stage analysis revealed that SNX25, PDLIM3, and SORBS2 may serve as genetic modifiers of age at onset in both EOAD and LOAD.
Collapse
Affiliation(s)
- Joseph H. Lee
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York2Gertrude H. Sergievsky Center, Columbia University, New York, New York3Department of Epidemiology, School of Public Health, Columbia Univer
| | - Rong Cheng
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York2Gertrude H. Sergievsky Center, Columbia University, New York, New York
| | - Badri Vardarajan
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York2Gertrude H. Sergievsky Center, Columbia University, New York, New York
| | - Rafael Lantigua
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York4Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Dolly Reyes-Dumeyer
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York
| | | | | | | | - Timothy W. Behrens
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Martin Medrano
- School of Medicine, Pontificia Universidad Catolica Madre y Maestra, Santiago, Dominican Republic
| | | | - Richard Mayeux
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York2Gertrude H. Sergievsky Center, Columbia University, New York, New York3Department of Epidemiology, School of Public Health, Columbia Univer
| |
Collapse
|
39
|
Wang C, Qu B, Wang Z, Ju J, Wang Y, Wang Z, Cao P, Wang D. Proteomic identification of differentially expressed proteins in vascular wall of patients with ruptured intracranial aneurysms. Atherosclerosis 2015; 238:201-6. [DOI: 10.1016/j.atherosclerosis.2014.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 01/23/2023]
|
40
|
Rowther FB, Wei W, Dawson TP, Ashton K, Singh A, Madiesse-Timchou MP, Thomas DGT, Darling JL, Warr T. Cyclic nucleotide phosphodiesterase-1C (PDE1C) drives cell proliferation, migration and invasion in glioblastoma multiforme cells in vitro. Mol Carcinog 2015; 55:268-79. [PMID: 25620587 DOI: 10.1002/mc.22276] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/17/2022]
Abstract
Cyclic nucleotides (cAMP & cGMP) are critical intracellular second messengers involved in the transduction of a diverse array of stimuli and their catabolism is mediated by phosphodiesterases (PDEs). We previously detected focal genomic amplification of PDE1C in >90 glioblastoma multiforme (GBM) cells suggesting a potential as a novel therapeutic target in these cells. In this report, we show that genomic gain of PDE1C was associated with increased expression in low passage GBM-derived cell cultures. We demonstrate that PDE1C is essential in driving cell proliferation, migration and invasion in GBM cultures since silencing of this gene significantly mitigates these functions. We also define the mechanistic basis of this functional effect through whole genome expression analysis by identifying down-stream gene effectors of PDE1C which are involved in cell cycle and cell adhesion regulation. In addition, we also demonstrate that Vinpocetine, a general PDE1 inhibitor, can also attenuate proliferation with no effect on invasion/migration. Up-regulation of at least one of this gene set (IL8, CXCL2, FOSB, NFE2L3, SUB1, SORBS2, WNT5A, and MMP1) in TCGA GBM cohorts is associated with worse outcome and PDE1C silencing down-regulated their expression, thus also indicating potential to influence patient survival. Therefore we conclude that proliferation, migration, and invasion of GBM cells could also be regulated downstream of PDE1C.
Collapse
Affiliation(s)
- Farjana B Rowther
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Weinbin Wei
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Timothy P Dawson
- Lancashire Teaching Hospitals, Royal Preston Hospital, Preston, UK
| | - Katherine Ashton
- Lancashire Teaching Hospitals, Royal Preston Hospital, Preston, UK
| | - Anushree Singh
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | | | - D G T Thomas
- National Hospital for Neurology and Neurosurgery, London
| | - John L Darling
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Tracy Warr
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| |
Collapse
|
41
|
Anekal PV, Yong J, Manser E. Arg kinase-binding protein 2 (ArgBP2) interaction with α-actinin and actin stress fibers inhibits cell migration. J Biol Chem 2014; 290:2112-25. [PMID: 25429109 DOI: 10.1074/jbc.m114.610725] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cell migration requires dynamic remodeling of the actomyosin network. We report here that an adapter protein, ArgBP2, is a component of α-actinin containing stress fibers and inhibits migration. ArgBP2 is undetectable in many commonly studied cancer-derived cell lines. COS-7 and HeLa cells express ArgBP2 (by Western analysis), but expression was detectable only in approximately half the cells by immunofluorescence. Short term clonal analysis demonstrated 0.2-0.3% of cells switch ArgBP2 expression (on or off) per cell division. ArgBP2 can have a fundamental impact on the actomyosin network: ArgBP2 positive COS-7 cells, for example, are clearly distinguishable by their denser actomyosin (stress fiber) network. ArgBP2γ binding to α-actinin appears to underlie its ability to localize to stress fibers and decrease cell migration. We map a small α-actinin binding region in ArgBP2 (residues 192-228) that is essential for these effects. Protein kinase A phosphorylation of ArgBP2γ at neighboring Ser-259 and consequent 14-3-3 binding blocks its interaction with α-actinin. ArgBP2 is known to be down-regulated in some aggressively metastatic cancers. Our work provides a biochemical explanation for the anti-migratory effect of ArgBP2.
Collapse
Affiliation(s)
- Praju Vikas Anekal
- From the sGSK Group Institute of Molecular and Cell Biology (IMCB), Proteos Building, 61 Biopolis Drive, 138673 Singapore
| | - Jeffery Yong
- From the sGSK Group Institute of Molecular and Cell Biology (IMCB), Proteos Building, 61 Biopolis Drive, 138673 Singapore
| | - Ed Manser
- From the sGSK Group Institute of Molecular and Cell Biology (IMCB), Proteos Building, 61 Biopolis Drive, 138673 Singapore, the Institute of Medical Biology (IMB), 8A Biomedical Grove, 06-06 Immunos Building, 138648 Singapore, and the Department of Pharmacology, National University of Singapore, 119077 Singapore
| |
Collapse
|
42
|
Santos MS, Foss SM, Park CK, Voglmaier SM. Protein interactions of the vesicular glutamate transporter VGLUT1. PLoS One 2014; 9:e109824. [PMID: 25334008 PMCID: PMC4198130 DOI: 10.1371/journal.pone.0109824] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/08/2014] [Indexed: 11/18/2022] Open
Abstract
Exocytotic release of glutamate depends upon loading of the neurotransmitter into synaptic vesicles by vesicular glutamate transporters, VGLUTs. The major isoforms, VGLUT1 and 2, exhibit a complementary pattern of expression in synapses of the adult rodent brain that correlates with the probability of release and potential for plasticity. Indeed, expression of different VGLUT protein isoforms confers different properties of release probability. Expression of VGLUT1 or 2 protein also determines the kinetics of synaptic vesicle recycling. To identify molecular determinants that may be related to reported differences in VGLUT trafficking and glutamate release properties, we investigated some of the intrinsic differences between the two isoforms. VGLUT1 and 2 exhibit a high degree of sequence homology, but differ in their N- and C-termini. While the C-termini of VGLUT1 and 2 share a dileucine-like trafficking motif and a proline-, glutamate-, serine-, and threonine-rich PEST domain, only VGLUT1 contains two polyproline domains and a phosphorylation consensus sequence in a region of acidic amino acids. The interaction of a VGLUT1 polyproline domain with the endocytic protein endophilin recruits VGLUT1 to a fast recycling pathway. To identify trans-acting cellular proteins that interact with the distinct motifs found in the C-terminus of VGLUT1, we performed a series of in vitro biochemical screening assays using the region encompassing the polyproline motifs, phosphorylation consensus sites, and PEST domain. We identify interactors that belong to several classes of proteins that modulate cellular function, including actin cytoskeletal adaptors, ubiquitin ligases, and tyrosine kinases. The nature of these interactions suggests novel avenues to investigate the modulation of synaptic vesicle protein recycling.
Collapse
Affiliation(s)
- Magda S. Santos
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - Sarah M. Foss
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
- Graduate Program in Cell Biology, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - C. Kevin Park
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - Susan M. Voglmaier
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| |
Collapse
|
43
|
Buvall L, Rashmi P, Lopez-Rivera E, Andreeva S, Weins A, Wallentin H, Greka A, Mundel P. Proteasomal degradation of Nck1 but not Nck2 regulates RhoA activation and actin dynamics. Nat Commun 2014; 4:2863. [PMID: 24287595 DOI: 10.1038/ncomms3863] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/04/2013] [Indexed: 11/09/2022] Open
Abstract
The ubiquitously expressed adapter proteins Nck1/2 interact with a multitude of effector molecules to regulate diverse cellular functions including cytoskeletal dynamics. Here we show that Nck1, but not Nck2, is a substrate of c-Cbl-mediated ubiquitination. We uncover lysine 178 in Nck1 as the evolutionarily conserved ubiquitin acceptor site. We previously reported that synaptopodin, a proline-rich actin-binding protein, induces stress fibres by blocking the Smurf1-mediated ubiquitination of RhoA. We now find that synaptopodin competes with c-Cbl for binding to Nck1, which prevents the ubiquitination of Nck1 by c-Cbl. Gene silencing of c-Cbl restores Nck1 protein abundance and stress fibres in synaptopodin knockdown cells. Similarly, expression of c-Cbl-resistant Nck1(K178R) or Nck2 containing the SH3 domain 2 of Nck1 restores stress fibres in synaptopodin-depleted podocytes through activation of RhoA signalling. These findings reveal proteasomal regulation as a key factor in the distinct and non-redundant effects of Nck on RhoA-mediated actin dynamics.
Collapse
Affiliation(s)
- Lisa Buvall
- 1] Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts 02129, USA [2]
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Dynamin is a large GTPase that mediates plasma membrane fission during clathrin-mediated endocytosis. Dynamin assembles into polymers on the necks of budding membranes in cells and has been shown to undergo GTP-dependent conformational changes that lead to membrane fission in vitro. Recent efforts have shed new light on the mechanisms of dynamin-mediated fission, yet exactly how dynamin performs this function in vivo is still not fully understood. Dynamin interacts with a number of proteins during the endocytic process. These interactions are mediated by the C-terminal proline-rich domain (PRD) of dynamin binding to SH3 domain-containing proteins. Three of these dynamin-binding partners (intersectin, amphiphysin and endophilin) have been shown to play important roles in the clathrin-mediated endocytosis process. They promote dynamin-mediated plasma membrane fission by regulating three important sequential steps in the process: recruitment of dynamin to sites of endocytosis; assembly of dynamin into a functional fission complex at the necks of clathrin-coated pits (CCPs); and regulation of dynamin-stimulated GTPase activity, a key requirement for fission.
Collapse
|
45
|
Roignot J, Bonacci T, Ghigo E, Iovanna JL, Soubeyran P. Oligomerization and phosphorylation dependent regulation of ArgBP2 adaptive capabilities and associated functions. PLoS One 2014; 9:e87130. [PMID: 24475245 PMCID: PMC3903627 DOI: 10.1371/journal.pone.0087130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/11/2013] [Indexed: 11/22/2022] Open
Abstract
ArgBP2 (Arg-Binding Protein 2/SORBS2) is an adaptor protein involved in cytoskeleton associated signal transduction, thereby regulating cell migration and adhesion. These features are associated with its antitumoral role in pancreatic cancer cells. Tyrosine phosphorylation of ArgBP2, mediated by c-Abl kinase and counterbalanced by PTP-PEST phosphatase, regulates many of its interactions. However, the exact mechanisms of action and of regulation of ArgBP2 remain largely unknown. We found that ArgBP2 has the capacity to form oligomers which are destabilized by tyrosine phosphorylation. We could show that ArgBP2 oligomerization involves the binding of one of its SH3 domains to a specific proline rich cluster. ArgBP2 self-association increases its binding to some of its molecular partners and decreased its affinity for others. Hence, the phosphorylation/oligomerization state of ArgBP2 directly regulates its functions by modulating its adaptive capabilities. Importantly, using a human pancreatic cancer cell model (MiaPaCa-2 cells), we could validate that this property of ArgBP2 is critical for its cytoskeleton associated functions. In conclusions, we describe a new mechanism of regulation of ArgBP2 where tyrosine phosphorylation of the protein interfere with a SH3 mediated self-interaction, thereby controlling its panel of interacting partners and related functions.
Collapse
Affiliation(s)
- Julie Roignot
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Marseille, France
| | - Thomas Bonacci
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Marseille, France
| | - Eric Ghigo
- URMITE-IRD198, CNRS UMR7278, INSERM U1095, Aix-Marseille Univ, Marseille, France
| | - Juan L. Iovanna
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Marseille, France
- * E-mail:
| |
Collapse
|
46
|
Lee H, Tsygankov AY. Cbl-family proteins as regulators of cytoskeleton-dependent phenomena. J Cell Physiol 2013; 228:2285-93. [DOI: 10.1002/jcp.24412] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/29/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Hojin Lee
- Department of Microbiology and Immunology; Sol Sherry Thrombosis Research Center and Fels Institute for Cancer Research; Temple University School of Medicine; Philadelphia Pennsylvania
| | - Alexander Y. Tsygankov
- Department of Microbiology and Immunology; Sol Sherry Thrombosis Research Center and Fels Institute for Cancer Research; Temple University School of Medicine; Philadelphia Pennsylvania
| |
Collapse
|
47
|
Martin M, Geudens I, Bruyr J, Potente M, Bleuart A, Lebrun M, Simonis N, Deroanne C, Twizere JC, Soubeyran P, Peixoto P, Mottet D, Janssens V, Hofmann WK, Claes F, Carmeliet P, Kettmann R, Gerhardt H, Dequiedt F. PP2A regulatory subunit Bα controls endothelial contractility and vessel lumen integrity via regulation of HDAC7. EMBO J 2013; 32:2491-503. [PMID: 23955003 DOI: 10.1038/emboj.2013.187] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/19/2013] [Indexed: 01/04/2023] Open
Abstract
To supply tissues with nutrients and oxygen, the cardiovascular system forms a seamless, hierarchically branched, network of lumenized tubes. Here, we show that maintenance of patent vessel lumens requires the Bα regulatory subunit of protein phosphatase 2A (PP2A). Deficiency of Bα in zebrafish precludes vascular lumen stabilization resulting in perfusion defects. Similarly, inactivation of PP2A-Bα in cultured ECs induces tubulogenesis failure due to alteration of cytoskeleton dynamics, actomyosin contractility and maturation of cell-extracellular matrix (ECM) contacts. Mechanistically, we show that PP2A-Bα controls the activity of HDAC7, an essential transcriptional regulator of vascular stability. In the absence of PP2A-Bα, transcriptional repression by HDAC7 is abrogated leading to enhanced expression of the cytoskeleton adaptor protein ArgBP2. ArgBP2 hyperactivates RhoA causing inadequate rearrangements of the EC actomyosin cytoskeleton. This study unravels the first specific role for a PP2A holoenzyme in development: the PP2A-Bα/HDAC7/ArgBP2 axis maintains vascular lumens by balancing endothelial cytoskeletal dynamics and cell-matrix adhesion.
Collapse
Affiliation(s)
- Maud Martin
- Laboratory of Protein Signaling and Interactions, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Sart-Tilman, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Signal transduction in cerebral arteries after subarachnoid hemorrhage-a phosphoproteomic approach. J Cereb Blood Flow Metab 2013; 33:1259-69. [PMID: 23715060 PMCID: PMC3734778 DOI: 10.1038/jcbfm.2013.78] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/17/2013] [Accepted: 04/21/2013] [Indexed: 12/25/2022]
Abstract
After subarachnoid hemorrhage (SAH), pathologic changes in cerebral arteries contribute to delayed cerebral ischemia and poor outcome. We hypothesize such changes are triggered by early intracellular signals, targeting of which may prevent SAH-induced vasculopathy. We performed an unbiased quantitative analysis of early SAH-induced phosphorylations in cerebral arteries and evaluated identified signaling components as targets for prevention of delayed vasculopathy and ischemia. Labeled phosphopeptides from rat cerebral arteries were quantified by high-resolution tandem mass spectrometry. Selected SAH-induced phosphorylations were validated by immunoblotting and monitored over a 24-hour time course post SAH. Moreover, inhibition of key phosphoproteins was performed. Major SAH-induced phosphorylations were observed on focal adhesion complexes, extracellular regulated kinase 1/2 (ERK1/2), calcium calmodulin-dependent kinase II, signal transducer and activator of transcription (STAT3) and c-Jun, the latter two downstream of ERK1/2. Inhibition of ERK1/2 6-hour post SAH prevented increases in cerebrovascular constrictor receptors, matrix metalloprotease-9, wall thickness, and improved neurologic outcome. STAT3 inhibition partially mimicked these effects. The study shows that quantitative mass spectrometry is a strong approach to study in vivo vascular signaling. Moreover, it shows that targeting of ERK1/2 prevents delayed pathologic changes in cerebral arteries and improves outcome, and identifies SAH-induced signaling components downstream and upstream of ERK1/2.
Collapse
|
49
|
Briñas L, Vassilopoulos S, Bonne G, Guicheney P, Bitoun M. Role of dynamin 2 in the disassembly of focal adhesions. J Mol Med (Berl) 2013; 91:803-9. [DOI: 10.1007/s00109-013-1040-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 11/29/2022]
|
50
|
Bharadwaj R, Roy M, Ohyama T, Sivan-Loukianova E, Delannoy M, Lloyd TE, Zlatic M, Eberl DF, Kolodkin AL. Cbl-associated protein regulates assembly and function of two tension-sensing structures in Drosophila. Development 2013; 140:627-38. [PMID: 23293294 DOI: 10.1242/dev.085100] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cbl-associated protein (CAP) localizes to focal adhesions and associates with numerous cytoskeletal proteins; however, its physiological roles remain unknown. Here, we demonstrate that Drosophila CAP regulates the organization of two actin-rich structures in Drosophila: muscle attachment sites (MASs), which connect somatic muscles to the body wall; and scolopale cells, which form an integral component of the fly chordotonal organs and mediate mechanosensation. Drosophila CAP mutants exhibit aberrant junctional invaginations and perturbation of the cytoskeletal organization at the MAS. CAP depletion also results in collapse of scolopale cells within chordotonal organs, leading to deficits in larval vibration sensation and adult hearing. We investigate the roles of different CAP protein domains in its recruitment to, and function at, various muscle subcellular compartments. Depletion of the CAP-interacting protein Vinculin results in a marked reduction in CAP levels at MASs, and vinculin mutants partially phenocopy Drosophila CAP mutants. These results show that CAP regulates junctional membrane and cytoskeletal organization at the membrane-cytoskeletal interface of stretch-sensitive structures, and they implicate integrin signaling through a CAP/Vinculin protein complex in stretch-sensitive organ assembly and function.
Collapse
Affiliation(s)
- Rajnish Bharadwaj
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|