1
|
Gerninghaus J, Zhubi R, Krämer A, Karim M, Tran DHN, Joerger AC, Schreiber C, Berger LM, Berger BT, Ehret TAL, Elson L, Lenz C, Saxena K, Müller S, Einav S, Knapp S, Hanke T. Back-Pocket Optimization of 2-Aminopyrimidine-Based Macrocycles Leads to Potent EPHA2/GAK Kinase Inhibitors. J Med Chem 2024; 67:12534-12552. [PMID: 39028937 DOI: 10.1021/acs.jmedchem.4c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Macrocyclization of acyclic compounds is a powerful strategy for improving inhibitor potency and selectivity. Here we have optimized 2-aminopyrimidine-based macrocycles to use these compounds as chemical tools for the ephrin kinase family. Starting with a promiscuous macrocyclic inhibitor, 6, we performed a structure-guided activity relationship and selectivity study using a panel of over 100 kinases. The crystal structure of EPHA2 in complex with the developed macrocycle 23 provided a basis for further optimization by specifically targeting the back pocket, resulting in compound 55, a potent inhibitor of EPHA2/A4 and GAK. Subsequent front-pocket derivatization resulted in an interesting in cellulo selectivity profile, favoring EPHA4 over the other ephrin receptor kinase family members. The dual EPHA2/A4 and GAK inhibitor 55 prevented dengue virus infection of Huh7 liver cells. However, further investigations are needed to determine whether this was a compound-specific effect or target-related.
Collapse
Affiliation(s)
- Joshua Gerninghaus
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Rezart Zhubi
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Marwah Karim
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Do Hoang Nhu Tran
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Christian Schreiber
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Lena M Berger
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Benedict-Tilman Berger
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Theresa A L Ehret
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Christopher Lenz
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Krishna Saxena
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Shirit Einav
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
- Chan Zuckerberg Biohub, 499 Illinois St, San Francisco, California 94158, United States
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Thomas Hanke
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| |
Collapse
|
2
|
Somu P, Mohanty S, Basavegowda N, Yadav AK, Paul S, Baek KH. The Interplay between Heat Shock Proteins and Cancer Pathogenesis: A Novel Strategy for Cancer Therapeutics. Cancers (Basel) 2024; 16:638. [PMID: 38339390 PMCID: PMC10854888 DOI: 10.3390/cancers16030638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Heat shock proteins (HSPs) are developmentally conserved families of protein found in both prokaryotic and eukaryotic organisms. HSPs are engaged in a diverse range of physiological processes, including molecular chaperone activity to assist the initial protein folding or promote the unfolding and refolding of misfolded intermediates to acquire the normal or native conformation and its translocation and prevent protein aggregation as well as in immunity, apoptosis, and autophagy. These molecular chaperonins are classified into various families according to their molecular size or weight, encompassing small HSPs (e.g., HSP10 and HSP27), HSP40, HSP60, HSP70, HSP90, and the category of large HSPs that include HSP100 and ClpB proteins. The overexpression of HSPs is induced to counteract cell stress at elevated levels in a variety of solid tumors, including anticancer chemotherapy, and is closely related to a worse prognosis and therapeutic resistance to cancer cells. HSPs are also involved in anti-apoptotic properties and are associated with processes of cancer progression and development, such as metastasis, invasion, and cell proliferation. This review outlines the previously mentioned HSPs and their significant involvement in diverse mechanisms of tumor advancement and metastasis, as well as their contribution to identifying potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Prathap Somu
- Department of Biotechnology and Chemical Engineering, School of Civil & Chemical Engineering, Manipal University Jaipur, Dehmi Kalan, Jaipur 303007, India;
| | - Sonali Mohanty
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, India;
| | - Nagaraj Basavegowda
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| | - Akhilesh Kumar Yadav
- Department of Environmental Engineering and Management, Chaoyang University of Technology, Taichung 413310, Taiwan;
- Department of Bioengineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, India
| | - Subhankar Paul
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, India;
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| |
Collapse
|
3
|
Zhang Y, Xiong X, Sun R, Zhu X, Wang C, Jiang B, Yang X, Li D, Fan G. Development of the non-receptor tyrosine kinase FER-targeting PROTACs as a potential strategy for antagonizing ovarian cancer cell motility and invasiveness. J Biol Chem 2023:104825. [PMID: 37196766 DOI: 10.1016/j.jbc.2023.104825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/23/2023] [Accepted: 05/07/2023] [Indexed: 05/19/2023] Open
Abstract
Aberrant overexpression of non-receptor tyrosine kinase FER has been reported in various ovarian carcinoma-derived tumor cells and is a poor prognosis factor for patient survival. It plays an essential role in tumor cell migration and invasion, acting concurrently in both kinase-dependent and -independent manners, which is not easily suppressed by conventional enzymatic inhibitors. Nevertheless, the proteolysis-targeting chimeras (PROTACs) technology offers superior efficacy over traditional activity-based inhibitors by simultaneously targeting enzymatic and scaffold functions. Hence in this study, we report the development of two PROTAC compounds that promote robust FER degradation in a cereblon-dependent manner. Both PROTAC degraders outperform an FDA-approved drug, Brigatinib, in ovarian cancer cell motility suppression. Importantly, these PROTAC compounds also degrade multiple oncogenic FER fusion proteins identified in human tumor samples. These results lay an experimental foundation to apply the PROTAC strategy to antagonize cell motility and invasiveness in ovarian and other types of cancers with aberrant expression of FER kinase and highlight PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions.
Collapse
Affiliation(s)
- Yanchun Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xuexue Xiong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Renhong Sun
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China
| | - Xiaotong Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China.
| | - Dake Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
4
|
Huang C, Ji C, Wang J. Current thoughts on cellular functions of numb-associated kinases. Mol Biol Rep 2023; 50:4645-4652. [PMID: 37014568 PMCID: PMC10072014 DOI: 10.1007/s11033-023-08372-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/02/2023] [Indexed: 04/05/2023]
Abstract
Members of the Numb-associated kinase family of serine/threonine kinases play an essential role in many cellular processes, such as endocytosis, autophagy, dendrite morphogenesis, osteoblast differentiation, and the regulation of the Notch pathway. Numb-associated kinases have been relevant to diverse diseases, including neuropathic pain, Parkinson's disease, and prostate cancer. Therefore, they are considered potential therapeutic targets. In addition, it is reported that Numb-associated kinases have been involved in the life cycle of multiple viruses such as hepatitis C virus (HCV), Ebola virus (EBOV), and dengue virus (DENV). Recently, Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to threaten global health. Studies show that Numb-associated kinases are implicated in the infection of SARS-CoV-2 which can be suppressed by Numb-associated kinases inhibitors. Thus, Numb-associated kinases are proposed as potential host targets for broad-spectrum antiviral strategies. We will focus on the recent advances in Numb-associated kinases-related cellular functions and their potential as host targets for viral infections in this review. Questions that remained unknown on the cellular functions of Numb-associated kinases will also be discussed.
Collapse
Affiliation(s)
- Chenxi Huang
- Department of Biology, Faculty of Environment and Life, Beijing University of Technology, 100124, Beijing, China
| | - Cuicui Ji
- Department of Biology, Faculty of Environment and Life, Beijing University of Technology, 100124, Beijing, China.
| | - Juan Wang
- Department of Biology, Faculty of Environment and Life, Beijing University of Technology, 100124, Beijing, China.
| |
Collapse
|
5
|
Millard RS, Bickley LK, Bateman KS, Verbruggen B, Farbos A, Lange A, Moore KA, Stentiford GD, Tyler CR, van Aerle R, Santos EM. Resistance to white spot syndrome virus in the European shore crab is associated with suppressed virion trafficking and heightened immune responses. Front Immunol 2022; 13:1057421. [PMID: 36636327 PMCID: PMC9831657 DOI: 10.3389/fimmu.2022.1057421] [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: 09/29/2022] [Accepted: 12/01/2022] [Indexed: 12/28/2022] Open
Abstract
Introduction All decapod crustaceans are considered potentially susceptible to White Spot Syndrome Virus (WSSV) infection, but the degree of White Spot Disease (WSD) susceptibility varies widely between species. The European shore crab Carcinus maenas can be infected with the virus for long periods of time without signs of disease. Given the high mortality rate of susceptible species, the differential susceptibility of these resistant hosts offers an opportunity to investigate mechanisms of disease resistance. Methods Here, the temporal transcriptional responses (mRNA and miRNA) of C. maenas following WSSV injection were analysed and compared to a previously published dataset for the highly WSSV susceptible Penaeus vannamei to identify key genes, processes and pathways contributing to increased WSD resistance. Results We show that, in contrast to P. vannamei, the transcriptional response during the first 2 days following WSSV injection in C. maenas is limited. During the later time points (7 days onwards), two groups of crabs were identified, a recalcitrant group where no replication of the virus occurred, and a group where significant viral replication occurred, with the transcriptional profiles of the latter group resembling those of WSSV-susceptible species. We identify key differences in the molecular responses of these groups to WSSV injection. Discussion We propose that increased WSD resistance in C. maenas may result from impaired WSSV endocytosis due to the inhibition of internal vesicle budding by dynamin-1, and a delay in movement to the nucleus caused by the downregulation of cytoskeletal transcripts required for WSSV cytoskeleton docking, during early stages of the infection. This response allows resistant hosts greater time to fine-tune immune responses associated with miRNA expression, apoptosis and the melanisation cascade to defend against, and clear, invading WSSV. These findings suggest that the initial stages of infection are key to resistance to WSSV in the crab and highlight possible pathways that could be targeted in farmed crustacean to enhance resistance to WSD.
Collapse
Affiliation(s)
- Rebecca S. Millard
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Weymouth, United Kingdom,Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom,*Correspondence: Rebecca S. Millard, ; Eduarda M. Santos,
| | - Lisa K. Bickley
- Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom,Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Kelly S. Bateman
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Weymouth, United Kingdom,Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom
| | - Bas Verbruggen
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Audrey Farbos
- University of Exeter Sequencing Facility, Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Anke Lange
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Karen A. Moore
- University of Exeter Sequencing Facility, Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Grant D. Stentiford
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Weymouth, United Kingdom,Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom
| | - Charles R. Tyler
- Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom,Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Ronny van Aerle
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Weymouth, United Kingdom,Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom
| | - Eduarda M. Santos
- Sustainable Aquaculture Futures, University of Exeter, Exeter, United Kingdom,Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom,*Correspondence: Rebecca S. Millard, ; Eduarda M. Santos,
| |
Collapse
|
6
|
Mächtel R, Boros FA, Dobert JP, Arnold P, Zunke F. From Lysosomal Storage Disorders to Parkinson's Disease - Challenges and Opportunities. J Mol Biol 2022:167932. [PMID: 36572237 DOI: 10.1016/j.jmb.2022.167932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Lysosomes are specialized organelles with an acidic pH that act as recycling hubs for intracellular and extracellular components. They harbour numerous different hydrolytic enzymes to degrade substrates like proteins, peptides, and glycolipids. Reduced catalytic activity of lysosomal enzymes can cause the accumulation of these substrates and loss of lysosomal integrity, resulting in lysosomal dysfunction and lysosomal storage disorders (LSDs). Post-mitotic cells, such as neurons, seem to be highly sensitive to damages induced by lysosomal dysfunction, thus LSDs often manifest with neurological symptoms. Interestingly, some LSDs and Parkinson's disease (PD) share common cellular pathomechanisms, suggesting convergence of aetiology of the two disease types. This is further underlined by genetic associations of several lysosomal genes involved in LSDs with PD. The increasing number of lysosome-associated genetic risk factors for PD makes it necessary to understand functions and interactions of lysosomal proteins/enzymes both in health and disease, thereby holding the potential to identify new therapeutic targets. In this review, we highlight genetic and mechanistic interactions between the complex lysosomal network, LSDs and PD, and elaborate on methodical challenges in lysosomal research.
Collapse
Affiliation(s)
- Rebecca Mächtel
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friederike Zunke
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany.
| |
Collapse
|
7
|
Sagarika P, Yadav K, Sahi C. Volleying plasma membrane proteins from birth to death: Role of J-domain proteins. Front Mol Biosci 2022; 9:1072242. [PMID: 36589230 PMCID: PMC9798423 DOI: 10.3389/fmolb.2022.1072242] [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: 10/17/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
The function, stability, and turnover of plasma membrane (PM) proteins are crucial for cellular homeostasis. Compared to soluble proteins, quality control of plasma membrane proteins is extremely challenging. Failure to meet the high quality control standards is detrimental to cellular and organismal health. J-domain proteins (JDPs) are among the most diverse group of chaperones that collaborate with other chaperones and protein degradation machinery to oversee cellular protein quality control (PQC). Although fragmented, the available literature from different models, including yeast, mammals, and plants, suggests that JDPs assist PM proteins with their synthesis, folding, and trafficking to their destination as well as their degradation, either through endocytic or proteasomal degradation pathways. Moreover, some JDPs interact directly with the membrane to regulate the stability and/or functionality of proteins at the PM. The deconvoluted picture emerging is that PM proteins are relayed from one JDP to another throughout their life cycle, further underscoring the versatility of the Hsp70:JDP machinery in the cell.
Collapse
|
8
|
Cui L, Li H, Xi Y, Hu Q, Liu H, Fan J, Xiang Y, Zhang X, Shui W, Lai Y. Vesicle trafficking and vesicle fusion: mechanisms, biological functions, and their implications for potential disease therapy. MOLECULAR BIOMEDICINE 2022; 3:29. [PMID: 36129576 PMCID: PMC9492833 DOI: 10.1186/s43556-022-00090-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
Intracellular vesicle trafficking is the fundamental process to maintain the homeostasis of membrane-enclosed organelles in eukaryotic cells. These organelles transport cargo from the donor membrane to the target membrane through the cargo containing vesicles. Vesicle trafficking pathway includes vesicle formation from the donor membrane, vesicle transport, and vesicle fusion with the target membrane. Coat protein mediated vesicle formation is a delicate membrane budding process for cargo molecules selection and package into vesicle carriers. Vesicle transport is a dynamic and specific process for the cargo containing vesicles translocation from the donor membrane to the target membrane. This process requires a group of conserved proteins such as Rab GTPases, motor adaptors, and motor proteins to ensure vesicle transport along cytoskeletal track. Soluble N-ethyl-maleimide-sensitive factor (NSF) attachment protein receptors (SNARE)-mediated vesicle fusion is the final process for vesicle unloading the cargo molecules at the target membrane. To ensure vesicle fusion occurring at a defined position and time pattern in eukaryotic cell, multiple fusogenic proteins, such as synaptotagmin (Syt), complexin (Cpx), Munc13, Munc18 and other tethering factors, cooperate together to precisely regulate the process of vesicle fusion. Dysfunctions of the fusogenic proteins in SNARE-mediated vesicle fusion are closely related to many diseases. Recent studies have suggested that stimulated membrane fusion can be manipulated pharmacologically via disruption the interface between the SNARE complex and Ca2+ sensor protein. Here, we summarize recent insights into the molecular mechanisms of vesicle trafficking, and implications for the development of new therapeutics based on the manipulation of vesicle fusion.
Collapse
|
9
|
Meisgen S, Hedlund M, Ambrosi A, Folkersen L, Ottosson V, Forsberg D, Thorlacius GE, Biavati L, Strandberg L, Mofors J, Ramskold D, Ruhrmann S, Meneghel L, Nyberg W, Espinosa A, Hamilton RM, Franco-Cereceda A, Hamsten A, Olsson T, Greene L, Eriksson P, Gemzell-Danielsson K, Salomonsson S, Kuchroo VK, Herlenius E, Kockum I, Sonesson SE, Wahren-Herlenius M. Auxilin is a novel susceptibility gene for congenital heart block which directly impacts fetal heart function. Ann Rheum Dis 2022; 81:1151-1161. [PMID: 35470161 PMCID: PMC9279836 DOI: 10.1136/annrheumdis-2021-221714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/11/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Neonatal lupus erythematosus (NLE) may develop after transplacental transfer of maternal autoantibodies with cardiac manifestations (congenital heart block, CHB) including atrioventricular block, atrial and ventricular arrhythmias, and cardiomyopathies. The association with anti-Ro/SSA antibodies is well established, but a recurrence rate of only 12%-16% despite persisting maternal autoantibodies suggests that additional factors are required for CHB development. Here, we identify fetal genetic variants conferring risk of CHB and elucidate their effects on cardiac function. METHODS A genome-wide association study was performed in families with at least one case of CHB. Gene expression was analysed by microarrays, RNA sequencing and PCR and protein expression by western blot, immunohistochemistry, immunofluorescence and flow cytometry. Calcium regulation and connectivity were analysed in primary cardiomyocytes and cells induced from pleuripotent stem cells. Fetal heart performance was analysed by Doppler/echocardiography. RESULTS We identified DNAJC6 as a novel fetal susceptibility gene, with decreased cardiac expression of DNAJC6 associated with the disease risk genotype. We further demonstrate that fetal cardiomyocytes deficient in auxilin, the protein encoded by DNAJC6, have abnormal connectivity and Ca2+ homoeostasis in culture, as well as decreased cell surface expression of the Cav1.3 calcium channel. Doppler echocardiography of auxilin-deficient fetal mice revealed cardiac NLE abnormalities in utero, including abnormal heart rhythm with atrial and ventricular ectopias, as well as a prolonged atrioventricular time intervals. CONCLUSIONS Our study identifies auxilin as the first genetic susceptibility factor in NLE modulating cardiac function, opening new avenues for the development of screening and therapeutic strategies in CHB.
Collapse
Affiliation(s)
- Sabrina Meisgen
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Hedlund
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Aurelie Ambrosi
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lasse Folkersen
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Technical University of Denmark, Lyngby, Denmark
| | - Vijole Ottosson
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Gudny Ella Thorlacius
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Luca Biavati
- Department of Physiology and Experimental Medicine, Hospital for Sick Children, Washington, DC, USA
| | - Linn Strandberg
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Johannes Mofors
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel Ramskold
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sabrina Ruhrmann
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lauro Meneghel
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William Nyberg
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Alexander Espinosa
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Anders Hamsten
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lois Greene
- National Institutes of Health, Bethesda, Maryland, USA
| | - Per Eriksson
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Stina Salomonsson
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Sonesson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
10
|
Tian X, Bunda P, Ishibe S. Podocyte Endocytosis in Regulating the Glomerular Filtration Barrier. Front Med (Lausanne) 2022; 9:801837. [PMID: 35223901 PMCID: PMC8866310 DOI: 10.3389/fmed.2022.801837] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/06/2022] [Indexed: 12/26/2022] Open
Abstract
Endocytosis is a mechanism that internalizes and recycles plasma membrane components and transmembrane receptors via vesicle formation, which is mediated by clathrin-dependent and clathrin-independent signaling pathways. Podocytes are specialized, terminally differentiated epithelial cells in the kidney, located on the outermost layer of the glomerulus. These cells play an important role in maintaining the integrity of the glomerular filtration barrier in conjunction with the adjacent basement membrane and endothelial cell layers within the glomerulus. An intact podocyte endocytic machinery appears to be necessary for maintaining podocyte function. De novo pathologic human genetic mutations and loss-of-function studies of critical podocyte endocytosis genes in genetically engineered mouse models suggest that this pathway contributes to the pathophysiology of development and progression of proteinuria in chronic kidney disease. Here, we review the mechanism of cellular endocytosis and its regulation in podocyte injury in the context of glomerular diseases. A thorough understanding of podocyte endocytosis may shed novel insights into its biological function in maintaining a functioning filter and offer potential targeted therapeutic strategies for proteinuric glomerular diseases.
Collapse
Affiliation(s)
| | | | - Shuta Ishibe
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
11
|
Lewis PA. Vesicular dysfunction and pathways to neurodegeneration. Essays Biochem 2021; 65:941-948. [PMID: 34897416 PMCID: PMC8709888 DOI: 10.1042/ebc20210034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022]
Abstract
Cellular control of vesicle biology and trafficking is critical for cell viability, with disruption of these pathways within the cells of the central nervous system resulting in neurodegeneration and disease. The past two decades have provided important insights into both the genetic and biological links between vesicle trafficking and neurodegeneration. In this essay, the pathways that have emerged as being critical for neuronal survival in the human brain will be discussed - illustrating the diversity of proteins and cellular events with three molecular case studies drawn from different neurological diseases.
Collapse
Affiliation(s)
- Patrick A Lewis
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States of America
| |
Collapse
|
12
|
Jaimon E, Tripathi A, Khurana A, Ghosh D, Sugatha J, Datta S. Binding with heat shock cognate protein HSC70 fine-tunes the Golgi association of the small GTPase ARL5B. J Biol Chem 2021; 297:101422. [PMID: 34798070 PMCID: PMC8661063 DOI: 10.1016/j.jbc.2021.101422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/30/2022] Open
Abstract
ARL5B, an ARF-like small GTPase localized to the trans-Golgi, is known for regulating endosome-Golgi trafficking and promoting the migration and invasion of breast cancer cells. Although a few interacting partners have been identified, the mechanism of the shuttling of ARL5B between the Golgi membrane and the cytosol is still obscure. Here, using GFP-binding protein (GBP) pull-down followed by mass spectrometry, we identified heat shock cognate protein (HSC70) as an additional interacting partner of ARL5B. Our pull-down and isothermal titration calorimetry (ITC)-based studies suggested that HSC70 binds to ARL5B in an ADP-dependent manner. Additionally, we showed that the N-terminal helix and the nucleotide status of ARL5B contribute to its recognition by HSC70. The confocal microscopy and cell fractionation studies in MDA-MB-231 breast cancer cells revealed that the depletion of HSC70 reduces the localization of ARL5B to the Golgi. Using in vitro reconstitution approach, we provide evidence that HSC70 fine-tunes the association of ARL5B with Golgi membrane. Finally, we demonstrated that the interaction between ARL5B and HSC70 is important for the localization of cation independent mannose-6-phosphate receptor (CIMPR) at Golgi. Collectively, we propose a mechanism by which HSC70, a constitutively expressed chaperone, modulates the Golgi association of ARL5B, which in turn has implications for the Golgi-associated functions of this GTPase.
Collapse
Affiliation(s)
- Ebsy Jaimon
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Aashutosh Tripathi
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Arohi Khurana
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Dipanjana Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Jini Sugatha
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Sunando Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India.
| |
Collapse
|
13
|
Ramesh ST, Navyasree KV, Sah S, Ashok AB, Qathoon N, Mohanty S, Swain RK, Umasankar PK. BMP2K phosphorylates AP-2 and regulates clathrin-mediated endocytosis. Traffic 2021; 22:377-396. [PMID: 34480404 DOI: 10.1111/tra.12814] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 01/29/2023]
Abstract
Phosphorylation of the central adaptor protein complex, AP-2 is pivotal for clathrin-mediated endocytosis (CME). Here, we uncover the role of an uncharacterized kinase (BMP-2 inducible kinase-BMP2K) in AP-2 phosphorylation. We demonstrate that BMP2K can phosphorylate AP-2 in vitro and in vivo. Functional impairment of BMP2K impedes AP-2 phosphorylation leading to defects in clathrin-coated pit (CCP) morphology and cargo internalization. BMP2K engages AP-2 via its extended C-terminus and this interaction is important for its CCP localization and function. Notably, endogenous BMP2K levels decline upon functional impairment of AP-2 indicating AP-2 dependent BMP2K stabilization in cells. Further, functional inactivation of BMP2K in zebrafish embryos yields gastrulation phenotypes which mirror AP-2 loss-of-function suggesting physiological relevance of BMP2K in vertebrates. Together, our findings propose involvement of a novel kinase in AP-2 phosphorylation and in the operation of CME.
Collapse
Affiliation(s)
- Shikha T Ramesh
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kolaparamba V Navyasree
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sneha Sah
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Anjitha B Ashok
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Nishada Qathoon
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | | | - Perunthottathu K Umasankar
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
14
|
Miyazaki M, Hiramoto M, Takano N, Kokuba H, Takemura J, Tokuhisa M, Hino H, Kazama H, Miyazawa K. Targeted disruption of GAK stagnates autophagic flux by disturbing lysosomal dynamics. Int J Mol Med 2021; 48:195. [PMID: 34468012 PMCID: PMC8416139 DOI: 10.3892/ijmm.2021.5028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/05/2021] [Indexed: 01/13/2023] Open
Abstract
The autophagy-lysosome system allows cells to adapt to environmental changes by regulating the degradation and recycling of cellular components, and to maintain homeostasis by removing aggregated proteins and defective organelles. Cyclin G-associated kinase (GAK) is involved in the regulation of clathrin-dependent endocytosis and cell cycle progression. In addition, a single nucleotide polymorphism at the GAK locus has been reported as a risk factor for Parkinson's disease. However, the roles of GAK in the autophagy-lysosome system are not completely understood, thus the present study aimed to clarify this. In the present study, under genetic disruption or chemical inhibition of GAK, analyzing autophagic flux and observing morphological changes of autophagosomes and autolysosomes revealed that GAK controlled lysosomal dynamics via actomyosin regulation, resulting in a steady progression of autophagy. GAK knockout (KO) in A549 cells impaired autophagosome-lysosome fusion and autophagic lysosome reformation, which resulted in the accumulation of enlarged autophagosomes and autolysosomes during prolonged starvation. The stagnation of autophagic flux accompanied by these phenomena was also observed with the addition of a GAK inhibitor. Furthermore, the addition of Rho-associated protein kinase (ROCK) inhibitor or ROCK1 knockdown mitigated GAK KO-mediated effects. The results suggested a vital role of GAK in controlling lysosomal dynamics via maintaining lysosomal homeostasis during autophagy.
Collapse
Affiliation(s)
- Masaya Miyazaki
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Naoharu Takano
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hiroko Kokuba
- Laboratory of Electron Microscopy, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Jun Takemura
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Mayumi Tokuhisa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hirotsugu Hino
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hiromi Kazama
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| |
Collapse
|
15
|
Bhave M, Mino RE, Wang X, Lee J, Grossman HM, Lakoduk AM, Danuser G, Schmid SL, Mettlen M. Functional characterization of 67 endocytic accessory proteins using multiparametric quantitative analysis of CCP dynamics. Proc Natl Acad Sci U S A 2020; 117:31591-31602. [PMID: 33257546 PMCID: PMC7749282 DOI: 10.1073/pnas.2020346117] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) begins with the nucleation of clathrin assembly on the plasma membrane, followed by stabilization and growth/maturation of clathrin-coated pits (CCPs) that eventually pinch off and internalize as clathrin-coated vesicles. This highly regulated process involves a myriad of endocytic accessory proteins (EAPs), many of which are multidomain proteins that encode a wide range of biochemical activities. Although domain-specific activities of EAPs have been extensively studied, their precise stage-specific functions have been identified in only a few cases. Using single-guide RNA (sgRNA)/dCas9 and small interfering RNA (siRNA)-mediated protein knockdown, combined with an image-based analysis pipeline, we have determined the phenotypic signature of 67 EAPs throughout the maturation process of CCPs. Based on these data, we show that EAPs can be partitioned into phenotypic clusters, which differentially affect CCP maturation and dynamics. Importantly, these clusters do not correlate with functional modules based on biochemical activities. Furthermore, we discover a critical role for SNARE proteins and their adaptors during early stages of CCP nucleation and stabilization and highlight the importance of GAK throughout CCP maturation that is consistent with GAK's multifunctional domain architecture. Together, these findings provide systematic, mechanistic insights into the plasticity and robustness of CME.
Collapse
Affiliation(s)
- Madhura Bhave
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rosa E Mino
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Xinxin Wang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jeon Lee
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Heather M Grossman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ashley M Lakoduk
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Gaudenz Danuser
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sandra L Schmid
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390;
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390;
| |
Collapse
|
16
|
Tian X, Inoue K, Zhang Y, Wang Y, Sperati CJ, Pedigo CE, Zhao T, Yan M, Groener M, Moledina DG, Ebenezer K, Li W, Zhang Z, Liebermann DA, Greene L, Greer P, Parikh CR, Ishibe S. Inhibiting calpain 1 and 2 in cyclin G associated kinase-knockout mice mitigates podocyte injury. JCI Insight 2020; 5:142740. [PMID: 33208557 PMCID: PMC7710277 DOI: 10.1172/jci.insight.142740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Evidence for reduced expression of cyclin G associated kinase (GAK) in glomeruli of patients with chronic kidney disease was observed in the Nephroseq human database, and GAK was found to be associated with the decline in kidney function. To examine the role of GAK, a protein that functions to uncoat clathrin during endocytosis, we generated podocyte-specific Gak-knockout mice (Gak-KO), which developed progressive proteinuria and kidney failure with global glomerulosclerosis. We isolated glomeruli from the mice carrying the mutation to perform messenger RNA profiling and unearthed evidence for dysregulated podocyte calpain protease activity as an important contributor to progressive podocyte damage. Treatment with calpain inhibitor III specifically inhibited calpain-1/-2 activities, mitigated the degree of proteinuria and glomerulosclerosis, and led to a striking increase in survival in the Gak-KO mice. Podocyte-specific deletion of Capns1, essential for calpain-1 and calpain-2 activities, also improved proteinuria and glomerulosclerosis in Gak-KO mice. Increased podocyte calpain activity-mediated proteolysis of IκBα resulted in increased NF-κB p65-induced expression of growth arrest and DNA-damage-inducible 45 beta in the Gak-KO mice. Our results suggest that loss of podocyte-associated Gak induces glomerular injury secondary to calcium dysregulation and aberrant calpain activation, which when inhibited, can provide a protective role.
Collapse
MESH Headings
- Animals
- Calpain/antagonists & inhibitors
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/therapy
- Female
- Glomerulosclerosis, Focal Segmental/etiology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/therapy
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Podocytes/metabolism
- Podocytes/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/physiology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/therapy
Collapse
Affiliation(s)
- Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yan Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - C. John Sperati
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher E. Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tingting Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Meihua Yan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marwin Groener
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dennis G. Moledina
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Karen Ebenezer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Wei Li
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dan A. Liebermann
- Fels Institute of Cancer Research and Molecular Biology and Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania USA
| | - Lois Greene
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Peter Greer
- Queen’s Cancer Research Institute, Kingston, Ontario, Canada
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
17
|
He K, Song E, Upadhyayula S, Dang S, Gaudin R, Skillern W, Bu K, Capraro BR, Rapoport I, Kusters I, Ma M, Kirchhausen T. Dynamics of Auxilin 1 and GAK in clathrin-mediated traffic. J Cell Biol 2020; 219:133624. [PMID: 31962345 PMCID: PMC7054993 DOI: 10.1083/jcb.201908142] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/28/2019] [Accepted: 12/15/2019] [Indexed: 01/08/2023] Open
Abstract
Clathrin-coated vesicles lose their clathrin lattice within seconds of pinching off, through the action of the Hsc70 “uncoating ATPase.” The J- and PTEN-like domain–containing proteins, auxilin 1 (Aux1) and auxilin 2 (GAK), recruit Hsc70. The PTEN-like domain has no phosphatase activity, but it can recognize phosphatidylinositol phosphate head groups. Aux1 and GAK appear on coated vesicles in successive transient bursts, immediately after dynamin-mediated membrane scission has released the vesicle from the plasma membrane. These bursts contain a very small number of auxilins, and even four to six molecules are sufficient to mediate uncoating. In contrast, we could not detect auxilins in abortive pits or at any time during coated pit assembly. We previously showed that clathrin-coated vesicles have a dynamic phosphoinositide landscape, and we have proposed that lipid head group recognition might determine the timing of Aux1 and GAK appearance. The differential recruitment of Aux1 and GAK correlates with temporal variations in phosphoinositide composition, consistent with a lipid-switch timing mechanism.
Collapse
Affiliation(s)
- Kangmin He
- Department of Cell Biology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Eli Song
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Srigokul Upadhyayula
- Department of Cell Biology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Song Dang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Raphael Gaudin
- Department of Cell Biology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Wesley Skillern
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Kevin Bu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA
| | | | - Iris Rapoport
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Ilja Kusters
- Department of Cell Biology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Minghe Ma
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Qiu X, He H, Huang Y, Wang J, Xiao Y. Genome-wide identification of m 6A-associated single-nucleotide polymorphisms in Parkinson's disease. Neurosci Lett 2020; 737:135315. [PMID: 32827573 DOI: 10.1016/j.neulet.2020.135315] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 10/23/2022]
Abstract
N6-methyladenosine (m6A)-associated single nucleotide polymorphisms (SNPs) play a vital role in several neurological diseases. However, little is known about the relationship between m6A modification and Parkinson's disease (PD). We investigated potential functional variants of m6A-SNPs from large-scale genome-wide association studies (GWAS) in PD patients. The candidate m6A-SNPs were further assessed by expression quantitative trait loci (eQTL) analysis and differential gene expression analysis. We identified 12 m6A-SNPs that were significantly associated with PD risk. Further, eQTL and expression analyses identified five of these m6A-SNPs (rs75072999 of GAK, rs1378602, rs4924839 and rs8071834 of ALKBH5, and rs1033500 of C6orf10) that were associated with altered gene expression in PD. Our results suggest that m6A-SNPs could play a role in PD risk. Future studies are needed to confirm these PD-associated m6A-SNPs and elucidate their mechanisms.
Collapse
Affiliation(s)
- Xiaohui Qiu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Honghu He
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanning Huang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Yousheng Xiao
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
19
|
Furini G, Burhan I, Huang L, Savoca MP, Atobatele A, Johnson T, Verderio EAM. Insights into the heparan sulphate-dependent externalisation of transglutaminase-2 (TG2) in glucose-stimulated proximal-like tubular epithelial cells. Anal Biochem 2020; 603:113628. [PMID: 32074489 DOI: 10.1016/j.ab.2020.113628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/25/2020] [Accepted: 02/12/2020] [Indexed: 02/09/2023]
Abstract
The extracellular matrix crosslinking enzyme transglutaminase 2 (TG2) is highly implicated in tissue fibrosis that precedes end-stage kidney failure. TG2 is unconventionally secreted through extracellular vesicles in a way that depends on the heparan sulphate (HS) proteoglycan syndecan-4 (Sdc4), the deletion of which reduces experimental kidney fibrosis as a result of lower extracellular TG2 in the tubule-interstitium. Here we establish a model of TG2 externalisation in NRK-52E tubular epithelial cells subjected to glucose stress. HS-binding TG2 mutants had reduced extracellular TG2 in transfected NRK-52E, suggesting that TG2-externalisation depends on an intact TG2 heparin binding site. Inhibition of N-ethylmaleimide sensitive factor (NSF) vesicle-fusing ATPase, which was identified in the recently elucidated TG2 kidney membrane-interactome, led to significantly lower TG2-externalisation, thus validating the involvement of membrane fusion in TG2 secretion. As cyclin-G-associated kinase (GAK) had emerged as a further TG2-partner in the fibrotic kidney, we investigated whether glucose-induced TG2-externalisation was accompanied by TG2 phosphorylation in consensus sequences of cyclin-dependent kinase (CDK). Glucose stress led to intense TG2 phosphorylation in serine/threonine CDK-target. TG2 phosphorylation by tyrosine kinases was also increased by glucose. Although the precise role of glucose-induced TG2 phosphorylation is unknown, these novel data suggest that phosphorylation may be involved in TG2 membrane-trafficking.
Collapse
Affiliation(s)
- Giulia Furini
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Izhar Burhan
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Linghong Huang
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Maria Pia Savoca
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Adeola Atobatele
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Tim Johnson
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Elisabetta A M Verderio
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK; BiGeA, University of Bologna, Bologna, 40126, Italy.
| |
Collapse
|
20
|
Narasimhan M, Johnson A, Prizak R, Kaufmann WA, Tan S, Casillas-Pérez B, Friml J. Evolutionarily unique mechanistic framework of clathrin-mediated endocytosis in plants. eLife 2020; 9:52067. [PMID: 31971511 PMCID: PMC7012609 DOI: 10.7554/elife.52067] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
In plants, clathrin mediated endocytosis (CME) represents the major route for cargo internalisation from the cell surface. It has been assumed to operate in an evolutionary conserved manner as in yeast and animals. Here we report characterisation of ultrastructure, dynamics and mechanisms of plant CME as allowed by our advancement in electron microscopy and quantitative live imaging techniques. Arabidopsis CME appears to follow the constant curvature model and the bona fide CME population generates vesicles of a predominantly hexagonal-basket type; larger and with faster kinetics than in other models. Contrary to the existing paradigm, actin is dispensable for CME events at the plasma membrane but plays a unique role in collecting endocytic vesicles, sorting of internalised cargos and directional endosome movement that itself actively promote CME events. Internalized vesicles display a strongly delayed and sequential uncoating. These unique features highlight the independent evolution of the plant CME mechanism during the autonomous rise of multicellularity in eukaryotes.
Collapse
Affiliation(s)
| | - Alexander Johnson
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Roshan Prizak
- Institute of Science and Technology Austria, Klosterneuburg, Austria.,Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | | | - Shutang Tan
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Jiří Friml
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
21
|
Beacham GM, Partlow EA, Hollopeter G. Conformational regulation of AP1 and AP2 clathrin adaptor complexes. Traffic 2019; 20:741-751. [PMID: 31313456 DOI: 10.1111/tra.12677] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/09/2019] [Accepted: 07/12/2019] [Indexed: 12/15/2022]
Abstract
Heterotetrameric clathrin adaptor protein complexes (APs) orchestrate the formation of coated vesicles for transport among organelles of the cell periphery. AP1 binds membranes enriched for phosphatidylinositol 4-phosphate, such as the trans Golgi network, while AP2 associates with phosphatidylinositol 4,5-bisphosphate of the plasma membrane. At their respective membranes, AP1 and AP2 bind the cytoplasmic tails of transmembrane protein cargo and clathrin triskelions, thereby coupling cargo recruitment to coat polymerization. Structural, biochemical and genetic studies have revealed that APs undergo conformational rearrangements and reversible phosphorylation to cycle between different activity states. While membrane, cargo and clathrin have been demonstrated to promote AP activation, growing evidence supports that membrane-associated proteins such as Arf1 and FCHo also stimulate this transition. APs may be returned to the inactive state via a regulated process involving phosphorylation and a protein called NECAP. Finally, because antiviral mechanisms often rely on appropriate trafficking of membrane proteins, viruses have evolved novel strategies to evade host defenses by influencing the conformation of APs. This review will cover recent advances in our understanding of the molecular inputs that stimulate AP1 and AP2 to adopt structurally and functionally distinct configurations.
Collapse
Affiliation(s)
| | - Edward A Partlow
- Department of Molecular Medicine, Cornell University, Ithaca, New York
| | | |
Collapse
|
22
|
Wolf B, Busso C, Gönczy P. Live imaging screen reveals that TYRO3 and GAK ensure accurate spindle positioning in human cells. Nat Commun 2019; 10:2859. [PMID: 31253758 PMCID: PMC6599018 DOI: 10.1038/s41467-019-10446-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
Proper spindle positioning is crucial for spatial cell division control. Spindle positioning in human cells relies on a ternary complex comprising Gαi1-3, LGN and NuMA, which anchors dynein at the cell cortex, thus enabling pulling forces to be exerted on astral microtubules. We develop a live imaging siRNA-based screen using stereotyped fibronectin micropatterns to uncover components modulating spindle positioning in human cells, testing 1280 genes, including all kinases and phosphatases. We thus discover 16 components whose inactivation dramatically perturbs spindle positioning, including tyrosine receptor kinase 3 (TYRO3) and cyclin G associated kinase (GAK). TYRO3 depletion results in excess NuMA and dynein at the cortex during metaphase, similar to the effect of blocking the TYRO3 downstream target phosphatidylinositol 3-kinase (PI3K). Furthermore, depletion of GAK leads to impaired astral microtubules, similar to the effect of downregulating the GAK-interactor Clathrin. Overall, our work uncovers components and mechanisms governing spindle positioning in human cells.
Collapse
Affiliation(s)
- Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
23
|
Phosphoproteomic analyses of kidneys of Atlantic salmon infected with Aeromonas salmonicida. Sci Rep 2019; 9:2101. [PMID: 30765835 PMCID: PMC6376026 DOI: 10.1038/s41598-019-38890-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 01/09/2019] [Indexed: 11/08/2022] Open
Abstract
Aeromonas salmonicida (A. salmonicida) is a pathogenic bacterium that causes furunculosis and poses a significant global risk, particularly in economic activities such as Atlantic salmon (Salmo salar) farming. In a previous study, we identified proteins that are significantly upregulated in kidneys of Atlantic salmon challenged with A. salmonicida. Phosphoproteomic analyses were conducted to further clarify the dynamic changes in protein phosphorylation patterns triggered by bacterial infection. To our knowledge, this is the first study to characterize phosphorylation events in proteins from A. salmonicida-infected Atlantic salmon. Overall, we identified over 5635 phosphorylation sites in 3112 proteins, and 1502 up-regulated and 77 down-regulated proteins quantified as a 1.5-fold or greater change relative to control levels. Based on the combined data from proteomic and motif analyses, we hypothesize that five prospective novel kinases (VRK3, GAK, HCK, PKCδ and RSK6) with common functions in inflammatory processes and cellular pathways to regulate apoptosis and the cytoskeleton could serve as potential biomarkers against bacterial propagation in fish. Data from STRING-based functional network analyses indicate that fga is the most central protein. Our collective findings provide new insights into protein phosphorylation patterns, which may serve as effective indicators of A. salmonicida infection in Atlantic salmon.
Collapse
|
24
|
A new role of anterograde motor Kif5b in facilitating large clathrin-coated vesicle mediated endocytosis via regulating clathrin uncoating. Cell Discov 2019; 4:65. [PMID: 30603101 PMCID: PMC6305376 DOI: 10.1038/s41421-018-0067-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 11/30/2022] Open
Abstract
Kif5b-driven anterograde transport and clathrin-mediated endocytosis (CME) are responsible for opposite intracellular trafficking, contributing to plasma membrane homeostasis. However, whether and how the two trafficking processes coordinate remain unclear. Here, we show that Kif5b directly interacts with clathrin heavy chain (CHC) at a region close to that for uncoating catalyst (Hsc70) and preferentially localizes on relatively large clathrin-coated vesicles (CCVs). Uncoating in vitro is decreased for CCVs from the cortex of kif5b conditional knockout (mutant) mouse and facilitated by adding Kif5b fragments containing CHC-binding site, while cell peripheral distribution of CHC or Hsc70 keeps unaffected by Kif5b depletion. Furthermore, cellular entry of vesicular stomatitis virus that internalizes into large CCV is inhibited by Kif5b depletion or introducing a dominant-negative Kif5b fragment. These findings showed a new role of Kif5b in regulating large CCV-mediated CME via affecting CCV uncoating, indicating Kif5b as a molecular knot connecting anterograde transport to CME.
Collapse
|
25
|
Lin AW, Gill KK, Castañeda MS, Matucci I, Eder N, Claxton S, Flynn H, Snijders AP, George R, Ultanir SK. Chemical genetic identification of GAK substrates reveals its role in regulating Na +/K +-ATPase. Life Sci Alliance 2018; 1:e201800118. [PMID: 30623173 PMCID: PMC6312924 DOI: 10.26508/lsa.201800118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/15/2022] Open
Abstract
Novel GAK phosphorylation targets are identified using chemical genetic methods. One of the substrates is the α subunit of the Na+/K+-ATPase, phosphorylation of which is necessary for its surface trafficking from endosomes. Conserved functions of NAK family kinases are described. Cyclin G–associated kinase (GAK) is a ubiquitous serine/threonine kinase that facilitates clathrin uncoating during vesicle trafficking. GAK phosphorylates a coat adaptor component, AP2M1, to help achieve this function. GAK is also implicated in Parkinson's disease through genome-wide association studies. However, GAK's role in mammalian neurons remains unclear, and insight may come from identification of further substrates. Employing a chemical genetics method, we show here that the sodium potassium pump (Na+/K+-ATPase) α-subunit Atp1a3 is a GAK target and that GAK regulates Na+/K+-ATPase trafficking to the plasma membrane. Whole-cell patch clamp recordings from CA1 pyramidal neurons in GAK conditional knockout mice show a larger change in resting membrane potential when exposed to the Na+/K+-ATPase blocker ouabain, indicating compromised Na+/K+-ATPase function in GAK knockouts. Our results suggest a modulatory role for GAK via phosphoregulation of substrates such as Atp1a3 during cargo trafficking.
Collapse
Affiliation(s)
- Amy W Lin
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Kalbinder K Gill
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | | | - Irene Matucci
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom.,Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | - Suzanne Claxton
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Helen Flynn
- Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Roger George
- Protein Purification Facility, The Francis Crick Institute, London, United Kingdom
| | - Sila K Ultanir
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
26
|
Li J, Tang D, Ireland SC, Wang Y. DjA1 maintains Golgi integrity via interaction with GRASP65. Mol Biol Cell 2018; 30:478-490. [PMID: 30566031 PMCID: PMC6594443 DOI: 10.1091/mbc.e18-10-0613] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In mammalian cells, the Golgi reassembly stacking protein of 65 kDa (GRASP65) has been implicated in both Golgi stacking and ribbon linking by forming trans-oligomers. To better understand its function and regulation, we used biochemical methods to identify the DnaJ homolog subfamily A member 1 (DjA1) as a novel GRASP65-binding protein. In cells, depletion of DjA1 resulted in Golgi fragmentation, short and improperly aligned cisternae, and delayed Golgi reassembly after nocodazole washout. In vitro, immunodepletion of DjA1 from interphase cytosol reduced its activity to enhance GRASP65 oligomerization and Golgi membrane fusion, while adding purified DjA1 enhanced GRASP65 oligomerization. DjA1 is a cochaperone of Heat shock cognate 71-kDa protein (Hsc70), but the activity of DjA1 in Golgi structure formation is independent of its cochaperone activity or Hsc70, rather, through DjA1-GRASP65 interaction to promote GRASP65 oligomerization. Thus, DjA1 interacts with GRASP65 to enhance Golgi structure formation through the promotion of GRASP65 trans-oligomerization.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Danming Tang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Stephen C Ireland
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085.,Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109-1085
| |
Collapse
|
27
|
Yoshida A, Sakai N, Uekusa Y, Imaoka Y, Itagaki Y, Suzuki Y, Yoshimura SH. Morphological changes of plasma membrane and protein assembly during clathrin-mediated endocytosis. PLoS Biol 2018; 16:e2004786. [PMID: 29723197 PMCID: PMC5953504 DOI: 10.1371/journal.pbio.2004786] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/15/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) proceeds through a series of morphological changes of the plasma membrane induced by a number of protein components. Although the spatiotemporal assembly of these proteins has been elucidated by fluorescence-based techniques, the protein-induced morphological changes of the plasma membrane have not been fully clarified in living cells. Here, we visualize membrane morphology together with protein localizations during CME by utilizing high-speed atomic force microscopy (HS-AFM) combined with a confocal laser scanning unit. The plasma membrane starts to invaginate approximately 30 s after clathrin starts to assemble, and the aperture diameter increases as clathrin accumulates. Actin rapidly accumulates around the pit and induces a small membrane swelling, which, within 30 s, rapidly covers the pit irreversibly. Inhibition of actin turnover abolishes the swelling and induces a reversible open–close motion of the pit, indicating that actin dynamics are necessary for efficient and irreversible pit closure at the end of CME. Cells communicate with their environments via the plasma membrane and various membrane proteins. Clathrin-mediated endocytosis (CME) plays a central role in such communication and proceeds with a series of multiprotein assembly, deformation of the plasma membrane, and production of a membrane vesicle that delivers extracellular signaling molecules into the cytoplasm. In this study, we utilized our home-built correlative imaging system comprising high-speed atomic force microscopy (HS-AFM) and confocal fluorescence microscopy to simultaneously image morphological changes of the plasma membrane and protein localization during CME in a living cell. The results revealed a tight correlation between the size of the pit and the amount of clathrin assembled. Actin dynamics play multiple roles in the assembly, maturation, and closing phases of the process, and affects membrane morphology, suggesting a close relationship between endocytosis and dynamic events at the cell cortex. Knock down of dynamin also affected the closing motion of the pit and showed functional correlation with actin.
Collapse
Affiliation(s)
- Aiko Yoshida
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | | | - Yuka Imaoka
- R&D Group, Olympus Corporation, Hachioji, Japan
| | | | - Yuki Suzuki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | | |
Collapse
|
28
|
Beacham GM, Partlow EA, Lange JJ, Hollopeter G. NECAPs are negative regulators of the AP2 clathrin adaptor complex. eLife 2018; 7:32242. [PMID: 29345618 PMCID: PMC5785209 DOI: 10.7554/elife.32242] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/17/2018] [Indexed: 12/27/2022] Open
Abstract
Eukaryotic cells internalize transmembrane receptors via clathrin-mediated endocytosis, but it remains unclear how the machinery underpinning this process is regulated. We recently discovered that membrane-associated muniscin proteins such as FCHo and SGIP initiate endocytosis by converting the AP2 clathrin adaptor complex to an open, active conformation that is then phosphorylated (Hollopeter et al., 2014). Here we report that loss of ncap-1, the sole C. elegans gene encoding an adaptiN Ear-binding Coat-Associated Protein (NECAP), bypasses the requirement for FCHO-1. Biochemical analyses reveal AP2 accumulates in an open, phosphorylated state in ncap-1 mutant worms, suggesting NECAPs promote the closed, inactive conformation of AP2. Consistent with this model, NECAPs preferentially bind open and phosphorylated forms of AP2 in vitro and localize with constitutively open AP2 mutants in vivo. NECAPs do not associate with phosphorylation-defective AP2 mutants, implying that phosphorylation precedes NECAP recruitment. We propose NECAPs function late in endocytosis to inactivate AP2.
Collapse
Affiliation(s)
| | - Edward A Partlow
- Department of Molecular Medicine, Cornell University, Ithaca, United States
| | - Jeffrey J Lange
- Stowers Institute for Medical Research, Kansas City, United States
| | - Gunther Hollopeter
- Department of Molecular Medicine, Cornell University, Ithaca, United States
| |
Collapse
|
29
|
Asquith CRM, Laitinen T, Bennett JM, Godoi PH, East MP, Tizzard GJ, Graves LM, Johnson GL, Dornsife RE, Wells CI, Elkins JM, Willson TM, Zuercher WJ. Identification and Optimization of 4-Anilinoquinolines as Inhibitors of Cyclin G Associated Kinase. ChemMedChem 2018; 13:48-66. [PMID: 29072804 PMCID: PMC5914168 DOI: 10.1002/cmdc.201700663] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Indexed: 11/09/2022]
Abstract
4-Anilinoquinolines were identified as potent and narrow-spectrum inhibitors of the cyclin G associated kinase (GAK), an important regulator of viral and bacterial entry into host cells. Optimization of the 4-anilino group and the 6,7-quinoline substituents produced GAK inhibitors with nanomolar activity, over 50 000-fold selectivity relative to other members of the numb-associated kinase (NAK) subfamily, and a compound (6,7-dimethoxy-N-(3,4,5-trimethoxyphenyl)quinolin-4-amine; 49) with a narrow-spectrum kinome profile. These compounds may be useful tools to explore the therapeutic potential of GAK in prevention of a broad range of infectious and systemic diseases.
Collapse
Affiliation(s)
- Christopher R. M. Asquith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tuomo Laitinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - James M. Bennett
- Structural Genomics Consortium and Target Discovery Institute, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Paulo H. Godoi
- Structural Genomics Consortium, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, 13083-886, Brazil
| | - Michael P. East
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Graham J. Tizzard
- UK National Crystallography Service, School of Chemistry, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Lee M. Graves
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Gary L. Johnson
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Ronna E. Dornsife
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carrow I. Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jonathan M. Elkins
- Structural Genomics Consortium and Target Discovery Institute, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- Structural Genomics Consortium, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, 13083-886, Brazil
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William J. Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
miR-206 inhibits renal cell cancer growth by targeting GAK. ACTA ACUST UNITED AC 2016; 36:852-858. [PMID: 27924503 DOI: 10.1007/s11596-016-1674-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 10/18/2016] [Indexed: 12/17/2022]
Abstract
Renal cell cancer (RCC) remains one of the most lethal types of cancer in adults. MicroRNAs (miRNAs) play key roles in the pathogenesis of RCC. The role of miR-206 in RCC has not been fully understood. The purpose of this study was to examine the role of miR-206 in the regulation of proliferation and metastasis of RCC and the possible mechanism. miR-206 expression was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in RCC cell lines (786-O and OS-RC-2 cells) and clinical samples. MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] method, colony formation and transwell assay were used to detect the tumor-suppressing ability of miR-206 in RCC. Luciferase assay was performed to verify the precise target of miR-206. The results showed that the expression of miR-206 was significantly down-regulated in RCC tissues and cells. The expression level of cyclin G-associated kinase (GAK), a master regulator of tumor proliferation and metastasis, was up-regulated with the decrease in miR-206 in RCC tissues as well as RCC cell lines. In addition, the miR-206 inhibitor promoted the proliferation, migration and invasion of 786-O and OS-RC-2 cells. Bioinformatics combined with luciferase and Western blot assays revealed that miR-206 inhibited the expression of GAK. Moreover, miR-206 regulates RCC cell growth partly through targeting GAK. Our study indicated that miR-206 functions as a tumor suppressor in regulating the proliferation, migration and invasion of RCC by directly targeting GAK, and it holds promises as a potential therapeutic target for RCC.
Collapse
|
31
|
The 4p16.3 Parkinson Disease Risk Locus Is Associated with GAK Expression and Genes Involved with the Synaptic Vesicle Membrane. PLoS One 2016; 11:e0160925. [PMID: 27508417 PMCID: PMC4980018 DOI: 10.1371/journal.pone.0160925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 07/27/2016] [Indexed: 11/29/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified the GAK/DGKQ/IDUA region on 4p16.3 among the top three risk loci for Parkinson’s disease (PD), but the specific gene and risk mechanism are unclear. Here, we report transcripts containing the 3’ clathrin-binding domain of GAK identified by RNA deep-sequencing in post-mortem human brain tissue as having increased expression in PD. Furthermore, carriers of 4p16.3 PD GWAS risk SNPs show decreased expression of one of these transcripts, GAK25 (Gencode Transcript 009), which correlates with the expression of genes functioning in the synaptic vesicle membrane. Together, these findings provide strong evidence for GAK clathrin-binding- and J-domain transcripts’ influence on PD pathogenicity, and for a role for GAK in regulating synaptic function in PD.
Collapse
|
32
|
Schroeter S, Beckmann S, Schmitt HD. Coat/Tether Interactions-Exception or Rule? Front Cell Dev Biol 2016; 4:44. [PMID: 27243008 PMCID: PMC4868844 DOI: 10.3389/fcell.2016.00044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/25/2016] [Indexed: 12/02/2022] Open
Abstract
Coat complexes are important for cargo selection and vesicle formation. Recent evidence suggests that they may also be involved in vesicle targeting. Tethering factors, which form an initial bridge between vesicles and the target membrane, may bind to coat complexes. In this review, we ask whether these coat/tether interactions share some common mechanisms, or whether they are special adaptations to the needs of very specific transport steps. We compare recent findings in two multisubunit tethering complexes, the Dsl1 complex and the HOPS complex, and put them into context with the TRAPP I complex as a prominent example for coat/tether interactions. We explore where coat/tether interactions are found, compare their function and structure, and comment on a possible evolution from a common ancestor of coats and tethers.
Collapse
Affiliation(s)
- Saskia Schroeter
- Neurobiology, Max Planck Institute for Biophysical Chemistry Göttingen, Germany
| | - Sabrina Beckmann
- Neurobiology, Max Planck Institute for Biophysical Chemistry Göttingen, Germany
| | - Hans Dieter Schmitt
- Neurobiology, Max Planck Institute for Biophysical Chemistry Göttingen, Germany
| |
Collapse
|
33
|
The Potential Mutation of GAK Gene in the Typical Sporadic Parkinson's Disease from the Han Population of Chinese Mainland. Mol Neurobiol 2015; 53:7119-7136. [PMID: 26676575 DOI: 10.1007/s12035-015-9595-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
The genetic factors about the pathogenesis of sporadic Parkinson's disease (sPD) is not completely clear at present; therefore, we performed a genome-wide association study, high-throughput sequencing analysis (HTPSA) of all cyclin G-associated kinase (GAK) exons, loss-of-function assessment, and sorting intolerant from tolerant analysis of HTPSA data in 250 typical sPD and 250 controls, which found 55 candidate single nucleotide polymorphisms (SNPs). To further explore these SNPs, we sequenced the 30 most strongly associated SNPs in the 460 typical sPD cases and the 525 controls. All subjects were from the Han population of Chinese mainland and excluded the toxic exposure, the heavy coffee drinking, and the early- and late-onset sPD. The minor allele frequencies (MAFs) at c.3824T>G, c.3794T>C, and c.3819G>A were higher in the control. The TG of c.3824T>G, the TC of c.3794T>C, and the AG of c.3819G>A were associated with the decreased risk of sPD. The subjects carrying the minor C allele of c.3794T>C or the minor A allele of c.3819G>A exhibited a decreased risk of sPD. c.3824T>G negatively affected the binding affinity of heat shock protein 70 (HSP70). c.3794T>C increased the surface area exposed to substrates. c.3819G>A most likely reduced the expression level of GAK. Our data suggest that the multiple SNPs of GAK synergistically participate in the pathogenesis of sPD through multiple pathways.
Collapse
|
34
|
Park BC, Yim YI, Zhao X, Olszewski MB, Eisenberg E, Greene LE. The clathrin-binding and J-domains of GAK support the uncoating and chaperoning of clathrin by Hsc70 in the brain. J Cell Sci 2015; 128:3811-21. [PMID: 26345367 DOI: 10.1242/jcs.171058] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022] Open
Abstract
Cyclin-G-associated kinase (GAK), the ubiquitously expressed J-domain protein, is essential for the chaperoning and uncoating of clathrin that is mediated by Hsc70 (also known as HSPA8). Adjacent to the C-terminal J-domain that binds to Hsc70, GAK has a clathrin-binding domain that is linked to an N-terminal kinase domain through a PTEN-like domain. Knocking out GAK in fibroblasts caused inhibition of clathrin-dependent trafficking, which was rescued by expressing a 62-kDa fragment of GAK, comprising just the clathrin-binding and J-domains. Expressing this fragment as a transgene in mice rescued the lethality and the histological defects caused by knocking out GAK in the liver or in the brain. Furthermore, when both GAK and auxilin (also known as DNAJC6), the neuronal-specific homolog of GAK, were knocked out in the brain, mice expressing the 62-kDa GAK fragment were viable, lived a normal life-span and had no major behavior abnormalities. However, these mice were about half the size of wild-type mice. Therefore, the PTEN-like domains of GAK and auxilin are not essential for Hsc70-dependent chaperoning and uncoating of clathrin, but depending on the tissue, these domains appear to increase the efficiency of these co-chaperones.
Collapse
Affiliation(s)
- Bum-Chan Park
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yang-In Yim
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaohong Zhao
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maciej B Olszewski
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Evan Eisenberg
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lois E Greene
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Beilina A, Cookson MR. Genes associated with Parkinson's disease: regulation of autophagy and beyond. J Neurochem 2015. [PMID: 26223426 DOI: 10.1111/jnc.13266] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial progress has been made in the genetic basis of Parkinson's disease (PD). In particular, by identifying genes that segregate with inherited PD or show robust association with sporadic disease, and by showing the same genes are found on both lists, we have generated an outline of the cause of this condition. Here, we will discuss what those genes tell us about the underlying biology of PD. We specifically discuss the relationships between protein products of PD genes and show that common links include regulation of the autophagy-lysosome system, an important way by which cells recycle proteins and organelles. We also discuss whether all PD genes should be considered to be in the same pathway and propose that in some cases the relationships are closer, whereas in other cases the interactions are more distant and might be considered separate. Beilina and Cookson review the links between genes for Parkinson's disease (red) and the autophagy-lysosomal system. They propose the hypothesis that many of the known PD genes can be assigned to pathways that affect (I) turnover of mitochondria via mitophagy (II) turnover of several vesicular structures via macroautophagy or chaperone-mediated autophagy or (III) general lysosome function. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Alexandra Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.
| |
Collapse
|
36
|
Inoue K, Ishibe S. Podocyte endocytosis in the regulation of the glomerular filtration barrier. Am J Physiol Renal Physiol 2015; 309:F398-405. [PMID: 26084928 DOI: 10.1152/ajprenal.00136.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Severe defects in the glomerular filtration barrier result in nephrotic syndrome, which is characterized by massive proteinuria. The podocyte, a specialized epithelial cell with interdigitating foot processes separated by a slit diaphragm, plays a vital role in regulating the passage of proteins from the capillary lumen to Bowman's space. Recent findings suggest a critical role for endocytosis in podocyte biology as highlighted by genetic mouse models of disease and human genetic mutations that result in the loss of the integrity of the glomerular filtration barrier. In vitro podocyte studies have also unraveled a plethora of constituents that are differentially internalized to maintain homeostasis. These observations provide a framework and impetus for understanding the precise regulation of podocyte endocytic machinery in both health and disease.
Collapse
Affiliation(s)
- Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
37
|
Sousa R, Lafer EM. The role of molecular chaperones in clathrin mediated vesicular trafficking. Front Mol Biosci 2015; 2:26. [PMID: 26042225 PMCID: PMC4436892 DOI: 10.3389/fmolb.2015.00026] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/05/2015] [Indexed: 12/04/2022] Open
Abstract
The discovery that the 70 kD “uncoating ATPase,” which removes clathrin coats from vesicles after endocytosis, is the constitutively expressed Hsc70 chaperone was a surprise. Subsequent work, however, revealed that uncoating is an archetypal Hsp70 reaction: the cochaperone auxilin, which contains a clathrin binding domain and an Hsc70 binding J domain, recruits Hsc70*ATP to the coat and, concomitant with ATP hydrolysis, transfers it to a hydrophobic Hsc70-binding element found on a flexible tail at the C-terminus of the clathrin heavy chain. Release of clathrin in association with Hsc70*ADP follows, and the subsequent, persistent association of clathrin with Hsc70 is important to prevent aberrant clathrin polymerization. Thus, the two canonical functions of Hsp70—dissociation of existing protein complexes or aggregates, and binding to a protein to inhibit its inappropriate aggregation—are recapitulated in uncoating. Association of clathrin with Hsc70 in vivo is regulated by Hsp110, an Hsp70 NEF that is itself a member of the Hsp70 family. How Hsp110 activity is itself regulated to make Hsc70-free clathrin available for endocytosis is unclear, though at synapses it's possible that the influx of calcium that accompanies depolarization activates the Ca++/calmodulin dependent calcineurin phosphatase which then dephosphorylates and activates Hsp110 to stimulate ADP/ATP exchange and release clathrin from Hsc70*ADP:clathrin complexes.
Collapse
Affiliation(s)
- Rui Sousa
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Eileen M Lafer
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| |
Collapse
|
38
|
Abstract
Endocytosis, the process whereby the plasma membrane invaginates to form vesicles, is essential for bringing many substances into the cell and for membrane turnover. The mechanism driving clathrin-mediated endocytosis (CME) involves > 50 different protein components assembling at a single location on the plasma membrane in a temporally ordered and hierarchal pathway. These proteins perform precisely choreographed steps that promote receptor recognition and clustering, membrane remodeling, and force-generating actin-filament assembly and turnover to drive membrane invagination and vesicle scission. Many critical aspects of the CME mechanism are conserved from yeast to mammals and were first elucidated in yeast, demonstrating that it is a powerful system for studying endocytosis. In this review, we describe our current mechanistic understanding of each step in the process of yeast CME, and the essential roles played by actin polymerization at these sites, while providing a historical perspective of how the landscape has changed since the preceding version of the YeastBook was published 17 years ago (1997). Finally, we discuss the key unresolved issues and where future studies might be headed.
Collapse
Affiliation(s)
- Bruce L Goode
- Brandeis University, Department of Biology, Rosenstiel Center, Waltham, Massachusetts 02454
| | - Julian A Eskin
- Brandeis University, Department of Biology, Rosenstiel Center, Waltham, Massachusetts 02454
| | - Beverly Wendland
- The Johns Hopkins University, Department of Biology, Baltimore, Maryland 21218
| |
Collapse
|
39
|
Merrifield CJ, Kaksonen M. Endocytic accessory factors and regulation of clathrin-mediated endocytosis. Cold Spring Harb Perspect Biol 2014; 6:a016733. [PMID: 25280766 DOI: 10.1101/cshperspect.a016733] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Up to 60 different proteins are recruited to the site of clathrin-mediated endocytosis in an ordered sequence. These accessory proteins have roles during all the different stages of clathrin-mediated endocytosis. First, they participate in the initiation of the endocytic event, thereby determining when and where endocytic vesicles are made; later they are involved in the maturation of the clathrin coat, recruitment of specific cargo molecules, bending of the membrane, and finally in scission and uncoating of the nascent vesicle. In addition, many of the accessory components are involved in regulating and coupling the actin cytoskeleton to the endocytic membrane. We will discuss the different accessory components and their various roles. Most of the data comes from studies performed with cultured mammalian cells or yeast cells. The process of endocytosis is well conserved between these different organisms, but there are also many interesting differences that may shed light on the mechanistic principles of endocytosis.
Collapse
Affiliation(s)
- Christien J Merrifield
- Laboratoire d'Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique UPR3082, 91198 Gif-sur-Yvette, France
| | - Marko Kaksonen
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| |
Collapse
|
40
|
Abstract
GAK (cyclin G-associated kinase) is a key regulator of clathrin-coated vesicle trafficking and plays a central role during development. Additionally, due to the unusually high plasticity of its catalytic domain, it is a frequent ‘off-target’ of clinical kinase inhibitors associated with respiratory side effects of these drugs. In the present paper, we determined the crystal structure of the GAK catalytic domain alone and in complex with specific single-chain antibodies (nanobodies). GAK is constitutively active and weakly associates in solution. The GAK apo structure revealed a dimeric inactive state of the catalytic domain mediated by an unusual activation segment interaction. Co-crystallization with the nanobody NbGAK_4 trapped GAK in a dimeric arrangement similar to the one observed in the apo structure, whereas NbGAK_1 captured the activation segment of monomeric GAK in a well-ordered conformation, representing features of the active kinase. The presented structural and biochemical data provide insight into the domain plasticity of GAK and demonstrate the utility of nanobodies to gain insight into conformational changes of dynamic molecules. In addition, we present structural data on the binding mode of ATP mimetic inhibitors and enzyme kinetic data, which will support rational inhibitor design of inhibitors to reduce the off-target effect on GAK. Cyclin G-associated kinase (GAK) is a regulator of clathrin-coated vesicle trafficking. The determined crystal structures of GAK in complex with specific single chain antibodies (nanobodies) revealed the domain plasticity of this kinase and unusual activation segment architecture.
Collapse
|
41
|
Protein aggregation can inhibit clathrin-mediated endocytosis by chaperone competition. Proc Natl Acad Sci U S A 2014; 111:E1481-90. [PMID: 24706768 DOI: 10.1073/pnas.1321811111] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Protein conformational diseases exhibit complex pathologies linked to numerous molecular defects. Aggregation of a disease-associated protein causes the misfolding and aggregation of other proteins, but how this interferes with diverse cellular pathways is unclear. Here, we show that aggregation of neurodegenerative disease-related proteins (polyglutamine, huntingtin, ataxin-1, and superoxide dismutase-1) inhibits clathrin-mediated endocytosis (CME) in mammalian cells by aggregate-driven sequestration of the major molecular chaperone heat shock cognate protein 70 (HSC70), which is required to drive multiple steps of CME. CME suppression was also phenocopied by HSC70 RNAi depletion and could be restored by conditionally increasing HSC70 abundance. Aggregation caused dysregulated AMPA receptor internalization and also inhibited CME in primary neurons expressing mutant huntingtin, showing direct relevance of our findings to the pathology in neurodegenerative diseases. We propose that aggregate-associated chaperone competition leads to both gain-of-function and loss-of-function phenotypes as chaperones become functionally depleted from multiple clients, leading to the decline of multiple cellular processes. The inherent properties of chaperones place them at risk, contributing to the complex pathologies of protein conformational diseases.
Collapse
|
42
|
Freeman CL, Hesketh G, Seaman MNJ. RME-8 coordinates the activity of the WASH complex with the function of the retromer SNX dimer to control endosomal tubulation. J Cell Sci 2014; 127:2053-70. [PMID: 24643499 DOI: 10.1242/jcs.144659] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Retromer is a vital element of the endosomal protein sorting machinery and comprises two subcomplexes that operate together to sort membrane proteins (cargo) and tubulate membranes. Tubules are formed by a dimer of sorting nexins, a key component of which is SNX1. Cargo selection is mediated by the VPS35-VPS29-VPS26 trimer, which additionally recruits the WASH complex through VPS35 binding to the WASH complex subunit FAM21. Loss of function of the WASH complex leads to dysregulation of endosome tubulation, although it is unclear how this occurs. Here, we show that FAM21 also binds to the SNX1-interacting DNAJ protein RME-8. Loss of RME-8 causes altered kinetics of SNX1 membrane association and a pronounced increase in highly branched endosomal tubules. Building on previous observations from other laboratories, we show that these tubules contain membrane proteins that are dependent upon WASH complex activity for their localization to the plasma membrane. Therefore, we propose that the interaction between RME-8 and the WASH complex provides a means to coordinate the activity of the WASH complex with the membrane-tubulating function of the sorting nexins at sites where retromer-mediated endosomal protein sorting occurs.
Collapse
Affiliation(s)
- Caroline L Freeman
- University of Cambridge, Cambridge Institute for Medical Research/Department of Clinical Biochemistry, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| | | | | |
Collapse
|
43
|
Bellei B, Pitisci A, Migliano E, Cardinali G, Picardo M. Pyridinyl imidazole compounds interfere with melanosomes sorting through the inhibition of cyclin G-associated Kinase, a regulator of cathepsins maturation. Cell Signal 2014; 26:716-23. [PMID: 24412755 DOI: 10.1016/j.cellsig.2013.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 12/31/2013] [Indexed: 01/07/2023]
Abstract
Transfer of melanin-containing melanosomes from melanocytes to neighboring keratinocytes results in skin pigmentation. Pharmacological modulation of melanosomal transfer has recently gained much attention as a strategy for modifying normal or abnormal pigmentation. In this study, while investigating the impact of pyridinyl imidazole (PI) compounds, a class of p38 MAPK inhibitors, on melanocyte differentiation we observed that some, but not all PIs interfere with the physiological melanosome sorting producing a strong retention of melanin in the intracellular compartment associated with a general reduction of melanin synthesis. Electron microscopy studies illustrated an accumulation of melanosomes inside melanocytes with enrichment in immature melanosome at stages II and III at the end of dendrites. We identified cyclin G-associated kinase GAK, a protein expressed ubiquitously in various tissues, as the off-target responsible of intracellular melanin accumulation and we report evidence that reduced GAK-dependent cathepsin maturation is implicated in melanosome sorting deficiency. The co-regulation of GAK and cathepsin B and L expression with the melanogenic biosynthetic pathway in normal human melanocytes as well as in B16-F0 melanoma cells strengthen the idea that these proteins represent new possible targets for prevention and treatment of irregular pigmentation.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatologic Institute, IRCCS, Rome 00144, Italy.
| | - Angela Pitisci
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatologic Institute, IRCCS, Rome 00144, Italy
| | - Emilia Migliano
- Department of Plastic and Reconstructive Surgery, San Gallicano Dermatologic Institute, IRCCS, Rome 00144, Italy
| | - Giorgia Cardinali
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatologic Institute, IRCCS, Rome 00144, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatologic Institute, IRCCS, Rome 00144, Italy
| |
Collapse
|
44
|
Olszewski MB, Chandris P, Park BC, Eisenberg E, Greene LE. Disruption of clathrin-mediated trafficking causes centrosome overduplication and senescence. Traffic 2013; 15:60-77. [PMID: 24138026 DOI: 10.1111/tra.12132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 10/13/2013] [Accepted: 10/18/2013] [Indexed: 12/24/2022]
Abstract
The Hsc70 cochaperone, G cyclin-associated kinase (GAK), has been shown to be essential for the chaperoning of clathrin by Hsc70 in the cell. In this study, we used conditional GAK knockout mouse embryonic fibroblasts (MEFs) to determine the effect of completely inhibiting clathrin-dependent trafficking on the cell cycle. After GAK was knocked out, the cells developed the unusual phenotype of having multiple centrosomes, but at the same time failed to divide and ultimately became senescent. To explain this phenotype, we examined the signaling profile and found that mitogenic stimulation of the GAK KO cells and the control cells were similar except for increased phosphorylation of Akt. In addition, the disruption of intracellular trafficking caused by knocking out GAK destabilized the lysosomal membranes, resulting in DNA damage due to iron leakage. Knocking down clathrin heavy chain or inhibiting dynamin largely reproduced the GAK KO phenotype, but inhibiting only clathrin-mediated endocytosis by knocking down adaptor protein (AP2) caused growth arrest and centrosome overduplication, but no DNA damage or senescence. We conclude that disruption of clathrin-dependent trafficking induces senescence accompanied by centrosome overduplication because of a combination of DNA damage and changes in mitogenic signaling that uncouples centrosomal duplication from DNA replication.
Collapse
Affiliation(s)
- Maciej B Olszewski
- Laboratory of Cell Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
45
|
Stricher F, Macri C, Ruff M, Muller S. HSPA8/HSC70 chaperone protein: structure, function, and chemical targeting. Autophagy 2013; 9:1937-54. [PMID: 24121476 DOI: 10.4161/auto.26448] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
HSPA8/HSC70 protein is a fascinating chaperone protein. It represents a constitutively expressed, cognate protein of the HSP70 family, which is central in many cellular processes. In particular, its regulatory role in autophagy is decisive. We focused this review on HSC70 structure-function considerations and based on this, we put a particular emphasis on HSC70 targeting by small molecules and peptides in order to develop intervention strategies that deviate some of HSC70 properties for therapeutic purposes. Generating active biomolecules regulating autophagy via its effect on HSC70 can effectively be designed only if we understand the fine relationships between HSC70 structure and functions.
Collapse
Affiliation(s)
- François Stricher
- CNRS; Institut de Biologie Moléculaire et Cellulaire; Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis; Strasbourg, France
| | | | | | | |
Collapse
|
46
|
Abstract
Membrane trafficking and mitosis are two essential processes in eukaryotic cells. Surprisingly, many proteins best known for their role in membrane trafficking have additional 'moonlighting' functions in mitosis. Despite having proteins in common, there is insufficient evidence for a specific connection between these two processes. Instead, these phenomena demonstrate the adaptability of the membrane trafficking machinery that allows its repurposing for different cellular functions.
Collapse
Affiliation(s)
- Stephen J Royle
- Division of Biomedical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry, CV4 7AL, UK.
| |
Collapse
|
47
|
Krantz KC, Puchalla J, Thapa R, Kobayashi C, Bisher M, Viehweg J, Carr CM, Rye HS. Clathrin coat disassembly by the yeast Hsc70/Ssa1p and auxilin/Swa2p proteins observed by single-particle burst analysis spectroscopy. J Biol Chem 2013; 288:26721-30. [PMID: 23913685 DOI: 10.1074/jbc.m113.491753] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of clathrin-coated vesicles in receptor-mediated endocytosis is conserved among eukaryotes, and many of the proteins required for clathrin coat assembly and disassembly have orthologs in yeast and mammals. In yeast, dozens of proteins have been identified as regulators of the multistep reaction required for endocytosis, including those that regulate disassembly of the clathrin coat. In mammalian systems, clathrin coat disassembly has been reconstituted using neuronal clathrin baskets mixed with the purified chaperone ATPase 70-kDa heat shock cognate (Hsc70), plus a clathrin-specific co-chaperone, such as the synaptic protein auxilin. Yet, despite previous characterization of the yeast Hsc70 ortholog, Ssa1p, and the auxilin-like ortholog, Swa2p, testing mechanistic models for disassembly of nonneuronal clathrin coats has been limited by the absence of a functional reconstitution assay. Here we use single-particle burst analysis spectroscopy, in combination with fluorescence correlation spectroscopy, to follow the population dynamics of fluorescently tagged yeast clathrin baskets in the presence of purified Ssa1p and Swa2p. An advantage of this combined approach for mechanistic studies is the ability to measure, as a function of time, changes in the number and size of objects from a starting population to the reaction products. Our results indicate that Ssa1p and Swa2p cooperatively disassemble yeast clathrin baskets into fragments larger than the individual triskelia, suggesting that disassembly of clathrin-coated vesicles may proceed through a partially uncoated intermediate.
Collapse
Affiliation(s)
- Kelly C Krantz
- From the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Clathrin, a protein best known for its role in membrane trafficking, has been recognised for many years as localising to the spindle apparatus during mitosis, but its function at the spindle remained unclear. Recent work has better defined the role of clathrin in the function of the mitotic spindle and proposed that clathrin crosslinks the microtubules (MTs) comprising the kinetochore fibres (K-fibres) in the mitotic spindle. This mitotic function is unrelated to the role of clathrin in membrane trafficking and occurs in partnership with two other spindle proteins: transforming acidic coiled-coil protein 3 (TACC3) and colonic hepatic tumour overexpressed gene (ch-TOG; also known as cytoskeleton-associated protein 5, CKAP5). This review summarises the role of clathrin in mitotic spindle organisation with an emphasis on the recent discovery of the TACC3-ch-TOG-clathrin complex.
Collapse
Affiliation(s)
- Stephen J Royle
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
49
|
Bandyopadhyay U, Cotney J, Nagy M, Oh S, Leng J, Mahajan M, Mane S, Fenton WA, Noonan JP, Horwich AL. RNA-Seq profiling of spinal cord motor neurons from a presymptomatic SOD1 ALS mouse. PLoS One 2013; 8:e53575. [PMID: 23301088 PMCID: PMC3536741 DOI: 10.1371/journal.pone.0053575] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/03/2012] [Indexed: 12/12/2022] Open
Abstract
Mechanisms involved with degeneration of motor neurons in amyotrophic lateral sclerosis (ALS; Lou Gehrig's Disease) are poorly understood, but genetically inherited forms, comprising ~10% of the cases, are potentially informative. Recent observations that several inherited forms of ALS involve the RNA binding proteins TDP43 and FUS raise the question as to whether RNA metabolism is generally disturbed in ALS. Here we conduct whole transcriptome profiling of motor neurons from a mouse strain, transgenic for a mutant human SOD1 (G85R SOD1-YFP), that develops symptoms of ALS and paralyzes at 5-6 months of age. Motor neuron cell bodies were laser microdissected from spinal cords at 3 months of age, a time when animals were presymptomatic but showed aggregation of the mutant protein in many lower motor neuron cell bodies and manifested extensive neuromuscular junction morphologic disturbance in their lower extremities. We observed only a small number of transcripts with altered expression levels or splicing in the G85R transgenic compared to age-matched animals of a wild-type SOD1 transgenic strain. Our results indicate that a major disturbance of polyadenylated RNA metabolism does not occur in motor neurons of mutant SOD1 mice, suggesting that the toxicity of the mutant protein lies at the level of translational or post-translational effects.
Collapse
Affiliation(s)
- Urmi Bandyopadhyay
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Justin Cotney
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Maria Nagy
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sunghee Oh
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jing Leng
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Milind Mahajan
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Wayne A. Fenton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - James P. Noonan
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Arthur L. Horwich
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
50
|
Comprehensive review on the HSC70 functions, interactions with related molecules and involvement in clinical diseases and therapeutic potential. Pharmacol Ther 2012; 136:354-74. [PMID: 22960394 DOI: 10.1016/j.pharmthera.2012.08.014] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 08/14/2012] [Indexed: 12/28/2022]
Abstract
Heat shock cognate protein 70 (HSC70) is a constitutively expressed molecular chaperone which belongs to the heat shock protein 70 (HSP70) family. HSC70 shares some of the structural and functional similarity with HSP70. HSC70 also has different properties compared with HSP70 and other heat shock family members. HSC70 performs its full functions by the cooperation of co-chaperones. It interacts with many other molecules as well and regulates various cellular functions. It is also involved in various diseases and may become a biomarker for diagnosis and potential therapeutic targets for design, discovery, and development of novel drugs to treat various diseases. In this article, we provide a comprehensive review on HSC70 from the literatures including the basic general information such as classification, structure and cellular location, genetics and function, as well as its protein association and interaction with other proteins. In addition, we also discussed the relationship of HSC70 and related clinical diseases such as cancer, cardiovascular, neurological, hepatic and many other diseases and possible therapeutic potential and highlight the progress and prospects of research in this field. Understanding the functions of HSC70 and its interaction with other molecules will help us to reveal other novel properties of this protein. Scientists may be able to utilize this protein as a biomarker and therapeutic target to make significant advancement in scientific research and clinical setting in the future.
Collapse
|