1
|
Bai YF, Shi XH, Zhang ML, Gu JH, Bai TL, Bai YB. Advances in the study of CCT3 in malignant tumors: A review. Medicine (Baltimore) 2025; 104:e41069. [PMID: 39928781 PMCID: PMC11813047 DOI: 10.1097/md.0000000000041069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 02/12/2025] Open
Abstract
Malignant tumors are among the leading causes of death worldwide, with their underlying mechanisms remaining largely unclear. Tumorigenesis is a complex process involving multiple factors, genes, and pathways. Tumor cells are characterized by abnormal proliferation, infiltration, invasion, and metastasis. Improving tumor diagnosis rates and identifying novel molecular therapeutic targets are of great significance for the advancement of modern medicine. Chaperonin containing TCP-1 subunit 3 (CCT3) is one of the subunits of the chaperonin containing TCP-1 complex, a molecular chaperone involved in protein folding and remodeling. CCT3 plays a crucial role in maintaining protein homeostasis, with key substrates including tubulin and actin. In recent years, CCT3 has been reported to be abnormally expressed in various cancers, correlating with prognosis and therapeutic outcomes. In this review, we summarize the basic structure and function of chaperonin containing TCP-1 complex and CCT3, and discuss the role of CCT3 in tumor development. Additionally, we explore its potential applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yun-Feng Bai
- Baotou Medical College, Baotou, Inner Mongolia, China
| | - Xiao-Hui Shi
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | | | - Jia-hui Gu
- Baotou Medical College, Baotou, Inner Mongolia, China
| | - Ta-La Bai
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yin-Bao Bai
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Zhang L, Huang W, Ma T, Shi X, Chen J, Hu YL, Liu YX, Liu ZX, Lu CH. Targeting CFTR restoring aggrephagy to suppress HSC activation and alleviate liver fibrosis. Int Immunopharmacol 2025; 145:113754. [PMID: 39667045 DOI: 10.1016/j.intimp.2024.113754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND AND AIMS Multiple studies have shown that hepatic fibrosis, a progressive condition that represents the endpoint of various chronic liver diseases, is primarily marked by the extensive activation of hepatic stellate cells (HSCs). However, the exact impact of cystic fibrosis transmembrane conductance regulator (CFTR) on HSCs during the development of hepatic fibrosis remains unclear. METHODS In our study, we measured CFTR levels in tissue samples and in HSCs activated by TGF-β stimulation. We established mouse models of liver fibrosis using carbon tetrachloride (CCl4) and bile duct ligation (BDL). In vitro, we investigated the specific mechanisms of CFTR action in HSCs by exploring aggrephagy. We employed co-immunoprecipitation (co-IP) experiments to identify potential downstream targets of CFTR. Finally, through rescue experiments, we examined the impact of GTPase-activating protein - binding protein 1 (G3BP1) on CFTR-mediated activation of hepatic stellate cells. RESULT In activated HSCs induced by TGF-β, the reduction of CFTR, various liver fibrosis models, and fibrotic tissue samples were identified. In vitro functional experiments confirmed that CFTR promoted the expression of fibrosis-related markers and aggrephagy in HSCs. Mechanistically, we found that CFTR directly interacts with G3BP1, thereby further promoting the TGF-β/Smad2/3 pathway. The inhibition of G3BP1 caused by CFTR knockdown reduced extracellular matrix deposition, contributing to alleviating liver fibrosis. CONCLUSION We emphasize that CFTR activates aggrephagy and promotes HSC activation and hepatic fibrosis by targeting G3BP1, participating in the TGF-β/Smad2/3 signaling pathway. Overall, CFTR has been identified as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Wei Huang
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Tao Ma
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Xiang Shi
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Jing Chen
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Yi-Lin Hu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China
| | - Yong-Xia Liu
- Department of Gastroenterology, Tongzhou District Traditional Chinese Medicine Hospital, Nantong, China
| | - Zhao-Xiu Liu
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China.
| | - Cui-Hua Lu
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001 China.
| |
Collapse
|
3
|
Kraft F, Rodriguez-Aliaga P, Yuan W, Franken L, Zajt K, Hasan D, Lee TT, Flex E, Hentschel A, Innes AM, Zheng B, Julia Suh DS, Knopp C, Lausberg E, Krause J, Zhang X, Trapane P, Carroll R, McClatchey M, Fry AE, Wang L, Giesselmann S, Hoang H, Baldridge D, Silverman GA, Radio FC, Bertini E, Ciolfi A, Blood KA, de Sainte Agathe JM, Charles P, Bergant G, Čuturilo G, Peterlin B, Diderich K, Streff H, Robak L, Oegema R, van Binsbergen E, Herriges J, Saunders CJ, Maier A, Wolking S, Weber Y, Lochmüller H, Meyer S, Aleman A, Polavarapu K, Nicolas G, Goldenberg A, Guyant L, Pope K, Hehmeyer KN, Monaghan KG, Quade A, Smol T, Caumes R, Duerinckx S, Depondt C, Van Paesschen W, Rieubland C, Poloni C, Guipponi M, Arcioni S, Meuwissen M, Jansen AC, Rosenblum J, Haack TB, Bertrand M, Gerstner L, Magg J, Riess O, Schulz JB, Wagner N, Wiesmann M, Weis J, Eggermann T, Begemann M, Roos A, Häusler M, Schedl T, Tartaglia M, Bremer J, Pak SC, Frydman J, Elbracht M, Kurth I. Brain malformations and seizures by impaired chaperonin function of TRiC. Science 2024; 386:516-525. [PMID: 39480921 DOI: 10.1126/science.adp8721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/22/2024] [Indexed: 11/02/2024]
Abstract
Malformations of the brain are common and vary in severity, from negligible to potentially fatal. Their causes have not been fully elucidated. Here, we report pathogenic variants in the core protein-folding machinery TRiC/CCT in individuals with brain malformations, intellectual disability, and seizures. The chaperonin TRiC is an obligate hetero-oligomer, and we identify variants in seven of its eight subunits, all of which impair function or assembly through different mechanisms. Transcriptome and proteome analyses of patient-derived fibroblasts demonstrate the various consequences of TRiC impairment. The results reveal an unexpected and potentially widespread role for protein folding in the development of the central nervous system and define a disease spectrum of "TRiCopathies."
Collapse
Affiliation(s)
- Florian Kraft
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | | | - Weimin Yuan
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Lena Franken
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Kamil Zajt
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Dimah Hasan
- Department for Diagnostic and Interventional Neuroradiology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Andreas Hentschel
- Leibniz- Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund 44139, Germany
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary T2N 1N4, Canada
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Dong Sun Julia Suh
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Cordula Knopp
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Eva Lausberg
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Jeremias Krause
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Xiaomeng Zhang
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Pamela Trapane
- Division of Pediatric Genetics, Department of Pediatrics, University of Florida College of Medicine-Jacksonville, Jacksonville, FL 32209, USA
| | - Riley Carroll
- Division of Pediatric Genetics, Department of Pediatrics, University of Florida College of Medicine-Jacksonville, Jacksonville, FL 32209, USA
| | - Martin McClatchey
- Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Andrew E Fry
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- All Wales Medical Genomics Service, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Lisa Wang
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Sebastian Giesselmann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Hieu Hoang
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Dustin Baldridge
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Gary A Silverman
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | | | - Enrico Bertini
- Neuromuscular Disorders, Ospedale Pediatrico Bambino Gesù IRCCS, Rome 00146, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù IRCCS, Rome 00146, Italy
| | - Katherine A Blood
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 2A1, Canada
| | - Jean-Madeleine de Sainte Agathe
- Department of Medical Genetics, Pitié-Salpêtrière Hospital, AP-HP.Sorbonne University, Paris 75005, France
- Laboratoire de Médecine Génomique Sorbonne Université, LBM SeqOIA, Paris 75014, France
| | - Perrine Charles
- Department of Medical Genetics, Pitié-Salpêtrière Hospital, AP-HP.Sorbonne University, Paris 75005, France
| | - Gaber Bergant
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
| | - Goran Čuturilo
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia, and University Children's Hospital, 11000 Belgrade, Serbia
| | - Borut Peterlin
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
| | - Karin Diderich
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 GD, Netherlands
| | - Haley Streff
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Laurie Robak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Renske Oegema
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht 3584 CX, Netherlands
| | - Ellen van Binsbergen
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht 3584 CX, Netherlands
| | - John Herriges
- Department of Pathology and Laboratory Medicine, Children's Mercy-Kansas City, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Carol J Saunders
- Department of Pathology and Laboratory Medicine, Children's Mercy-Kansas City, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO 64108, USA
| | - Andrea Maier
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen 52074, Germany
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Stefan Wolking
- Department of Epileptology and Neurology, Medical Faculty, RWTH Aachen University, Aachen 52074, Germany
| | - Yvonne Weber
- Department of Epileptology and Neurology, Medical Faculty, RWTH Aachen University, Aachen 52074, Germany
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa K1H 8L1, Canada
| | - Stefanie Meyer
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa K1H 8L1, Canada
| | - Alberto Aleman
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa K1H 8L1, Canada
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa K1H 8L1, Canada
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bangalore 560030, India
| | - Gael Nicolas
- Univ Rouen Normandie, Normandie univ, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Neurogenetics Diorders, F-76000 Rouen, France
| | - Alice Goldenberg
- Univ Rouen Normandie, Normandie univ, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Neurogenetics Diorders, F-76000 Rouen, France
| | - Lucie Guyant
- Univ Rouen Normandie, Normandie univ, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Neurogenetics Diorders, F-76000 Rouen, France
| | - Kathleen Pope
- University of South Florida, College of Public Health, Tampa, FL 33612, USA
- Nemours Children's Health, Department of Pediatrics, Division of Genetics, Orlando, FL 32827, USA
| | - Katherine N Hehmeyer
- Nemours Children's Health, Department of Pediatrics, Division of Genetics, Orlando, FL 32827, USA
| | | | - Annegret Quade
- Division of Pediatric Neurology and Social Pediatrics, Department of Pediatrics, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - Thomas Smol
- Department of Clinical Genetics, Lille University Hospital, CHU Lille, Lille 59000, France
| | - Roseline Caumes
- Department of Clinical Genetics, Lille University Hospital, CHU Lille, Lille 59000, France
| | - Sarah Duerinckx
- Department of Pediatric Neurology, Hôpital Universitaire de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Chantal Depondt
- Department of Neurology, Hôpital Universitaire de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Wim Van Paesschen
- Laboratory for Epilepsy Research, KU Leuven, Leuven 3000, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven 3000, Belgium
| | - Claudine Rieubland
- Department of Medical Genetics, Central Institute of the Hospitals, Hospital of the Valais, Sion 1951, Switzerland
| | - Claudia Poloni
- Department of Medical Genetics, Central Institute of the Hospitals, Hospital of the Valais, Sion 1951, Switzerland
| | - Michel Guipponi
- Department of Genetic Medicine, University Hospitals of Geneva and University of Geneva Medical Faculty, Geneva 1205, Switzerland
| | - Severine Arcioni
- Department of Medical Genetics, Central Institute of the Hospitals, Hospital of the Valais, Sion 1951, Switzerland
- Division of Medical Genetics, Central Institute of Hospitals, Valais Hospital, Sion 1951, Switzerland
| | - Marije Meuwissen
- Center of Medical Genetics, Antwerp University Hospital/ University of Antwerp, Edegem 2650, Belgium
| | - Anna C Jansen
- Department of Pediatrics, Division of Child Neurology, Antwerp University Hospital, University of Antwerp, Edegem 2650, Belgium
| | - Jessica Rosenblum
- Center of Medical Genetics, Antwerp University Hospital/ University of Antwerp, Edegem 2650, Belgium
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
| | - Miriam Bertrand
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
| | - Lea Gerstner
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
| | - Janine Magg
- Department of Neuropediatrics, Developmental Neurology, Social Pediatrics, University Children's Hospital, University of Tübingen, Tübingen 72076, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
| | - Jörg B Schulz
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen 52074, Germany
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Norbert Wagner
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
- Department of Pediatrics, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - Martin Wiesmann
- Department for Diagnostic and Interventional Neuroradiology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Thomas Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Matthias Begemann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
| | - Andreas Roos
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa K1H 8L1, Canada
- Department for Pediatric Neurology, University Medicine Essen, Duisburg-Essen University, 45147 Essen, Germany
- Institute of Neurology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Martin Häusler
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
- Division of Pediatric Neurology and Social Pediatrics, Department of Pediatrics, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - Tim Schedl
- Department of Genetics, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù IRCCS, Rome 00146, Italy
| | - Juliane Bremer
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Stephen C Pak
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Miriam Elbracht
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, 52074, Germany
- Center for Rare Diseases Aachen (ZSEA), RWTH Aachen University Hospital, Aachen 52074, Germany
| |
Collapse
|
4
|
Sharma M. Some brain disorders are "chaperonopathies". Science 2024; 386:496-497. [PMID: 39480954 DOI: 10.1126/science.adt0039] [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: 11/02/2024]
Abstract
Mutations that impair a protein-folding chaperone can lead to brain malformations.
Collapse
Affiliation(s)
- Manu Sharma
- Appel Alzheimer's Disease Research Institute and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Gvozdenov Z, Peng AYT, Biswas A, Barcutean Z, Gestaut D, Frydman J, Struhl K, Freeman BC. TRiC/CCT Chaperonin Governs RNA Polymerase II Activity in the Nucleus to Support RNA Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615188. [PMID: 39386699 PMCID: PMC11463447 DOI: 10.1101/2024.09.26.615188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The chaperonin TRiC/CCT is a large hetero-oligomeric ringed-structure that is essential in eukaryotes. While present in the nucleus, TRiC/CCT is typically considered to function in the cytosol where it mediates nascent polypeptide folding and the assembly/disassembly of protein complexes. Here, we investigated the nuclear role of TRiC/CCT. Inactivation of TRiC/CCT resulted in a significant increase in the production of nascent RNA leading to the accumulation of noncoding transcripts. The influence on transcription was not due to cytoplasmic TRiC/CCT-activities or other nuclear proteins as the effect was observed when TRiC/CCT was evicted from the nucleus and restricted to the cytoplasm. Rather, our data support a direct role of TRiC/CCT in regulating RNA polymerase II activity, as the chaperonin modulated nascent RNA production both in vivo and in vitro. Overall, our studies reveal a new avenue by which TRiC/CCT contributes to cell homeostasis by regulating the activity of nuclear RNA polymerase II.
Collapse
|
6
|
Selicky T, Jurcik M, Mikolaskova B, Pitelova A, Mayerova N, Kretova M, Osadska M, Jurcik J, Holic R, Kohutova L, Bellova J, Benko Z, Gregan J, Bagelova Polakova S, Barath P, Cipak L, Cipakova I. Defining the Functional Interactome of Spliceosome-Associated G-Patch Protein Gpl1 in the Fission Yeast Schizosaccharomyces pombe. Int J Mol Sci 2022; 23:12800. [PMID: 36361590 PMCID: PMC9658070 DOI: 10.3390/ijms232112800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 08/31/2023] Open
Abstract
Pre-mRNA splicing plays a fundamental role in securing protein diversity by generating multiple transcript isoforms from a single gene. Recently, it has been shown that specific G-patch domain-containing proteins are critical cofactors involved in the regulation of splicing processes. In this study, using the knock-out strategy, affinity purification and the yeast-two-hybrid assay, we demonstrated that the spliceosome-associated G-patch protein Gpl1 of the fission yeast S. pombe mediates interactions between putative RNA helicase Gih35 (SPAC20H4.09) and WD repeat protein Wdr83, and ensures their binding to the spliceosome. Furthermore, RT-qPCR analysis of the splicing efficiency of deletion mutants indicated that the absence of any of the components of the Gpl1-Gih35-Wdr83 complex leads to defective splicing of fet5 and pwi1, the reference genes whose unspliced isoforms harboring premature stop codons are targeted for degradation by the nonsense-mediated decay (NMD) pathway. Together, our results shed more light on the functional interactome of G-patch protein Gpl1 and revealed that the Gpl1-Gih35-Wdr83 complex plays an important role in the regulation of pre-mRNA splicing in S. pombe.
Collapse
Affiliation(s)
- Tomas Selicky
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Matus Jurcik
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Barbora Mikolaskova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Alexandra Pitelova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia
| | - Nina Mayerova
- Department of Genetics, Faculty of Natural Sciences, Comenius University in Bratislava, 841 04 Bratislava, Slovakia
| | - Miroslava Kretova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michaela Osadska
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Jan Jurcik
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Roman Holic
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia
| | - Lenka Kohutova
- Department of Glycobiology, Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
| | - Jana Bellova
- Department of Glycobiology, Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
| | - Zsigmond Benko
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia
- Department of Molecular Biotechnology and Microbiology, University of Debrecen, Egyetem tér 1, H4032 Debrecen, Hungary
| | - Juraj Gregan
- Department of Applied Genetics and Cell Biology, Institute of Microbial Genetics, University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad Lorenz Strasse 24, 3430 Tulln an der Donau, Austria
| | - Silvia Bagelova Polakova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia
| | - Peter Barath
- Department of Glycobiology, Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
- Medirex Group Academy, Novozamocka 67, 949 05 Nitra, Slovakia
| | - Lubos Cipak
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Ingrid Cipakova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
7
|
Shaheen N, Akhtar J, Umer Z, Khan MHF, Bakhtiari MH, Saleem M, Faisal A, Tariq M. Polycomb Requires Chaperonin Containing TCP-1 Subunit 7 for Maintaining Gene Silencing in Drosophila. Front Cell Dev Biol 2021; 9:727972. [PMID: 34660585 PMCID: PMC8517254 DOI: 10.3389/fcell.2021.727972] [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: 06/20/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
In metazoans, heritable states of cell type-specific gene expression patterns linked with specialization of various cell types constitute transcriptional cellular memory. Evolutionarily conserved Polycomb group (PcG) and trithorax group (trxG) proteins contribute to the transcriptional cellular memory by maintaining heritable patterns of repressed and active expression states, respectively. Although chromatin structure and modifications appear to play a fundamental role in maintenance of repression by PcG, the precise targeting mechanism and the specificity factors that bind PcG complexes to defined regions in chromosomes remain elusive. Here, we report a serendipitous discovery that uncovers an interplay between Polycomb (Pc) and chaperonin containing T-complex protein 1 (TCP-1) subunit 7 (CCT7) of TCP-1 ring complex (TRiC) chaperonin in Drosophila. CCT7 interacts with Pc at chromatin to maintain repressed states of homeotic and non-homeotic targets of PcG, which supports a strong genetic interaction observed between Pc and CCT7 mutants. Depletion of CCT7 results in dissociation of Pc from chromatin and redistribution of an abundant amount of Pc in cytoplasm. We propose that CCT7 is an important modulator of Pc, which helps Pc recruitment at chromatin, and compromising CCT7 can directly influence an evolutionary conserved epigenetic network that supervises the appropriate cellular identities during development and homeostasis of an organism.
Collapse
Affiliation(s)
- Najma Shaheen
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Jawad Akhtar
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Zain Umer
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Haider Farooq Khan
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Mahnoor Hussain Bakhtiari
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Murtaza Saleem
- Department of Physics, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Tariq
- Epigenetics and Gene Regulation Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
8
|
Carman PJ, Barrie KR, Dominguez R. Novel human cell expression method reveals the role and prevalence of posttranslational modification in nonmuscle tropomyosins. J Biol Chem 2021; 297:101154. [PMID: 34478714 PMCID: PMC8463859 DOI: 10.1016/j.jbc.2021.101154] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022] Open
Abstract
Biochemical studies require large quantities of proteins, which are typically obtained using bacterial overexpression. However, the folding machinery in bacteria is inadequate for expressing many mammalian proteins, which additionally undergo posttranslational modifications (PTMs) that bacteria, yeast, or insect cells cannot perform. Many proteins also require native N- and C-termini and cannot tolerate extra tag amino acids for proper function. Tropomyosin (Tpm), a coiled coil protein that decorates most actin filaments in cells, requires both native N- and C-termini and PTMs, specifically N-terminal acetylation (Nt-acetylation), to polymerize along actin filaments. Here, we describe a new method that combines native protein expression in human cells with an intein-based purification tag that can be precisely removed after purification. Using this method, we expressed several nonmuscle Tpm isoforms (Tpm1.6, Tpm1.7, Tpm2.1, Tpm3.1, Tpm3.2, and Tpm4.2) and the muscle isoform Tpm1.1. Proteomics analysis revealed that human-cell-expressed Tpms present various PTMs, including Nt-acetylation, Ser/Thr phosphorylation, Tyr phosphorylation, and Lys acetylation. Depending on the Tpm isoform (humans express up to 40 Tpm isoforms), Nt-acetylation occurs on either the initiator methionine or on the second residue after removal of the initiator methionine. Human-cell-expressed Tpms bind F-actin differently than their Escherichia coli-expressed counterparts, with or without N-terminal extensions intended to mimic Nt-acetylation, and they can form heterodimers in cells and in vitro. The expression method described here reveals previously unknown features of nonmuscle Tpms and can be used in future structural and biochemical studies with Tpms and other proteins, as shown here for α-synuclein.
Collapse
Affiliation(s)
- Peter J Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyle R Barrie
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
9
|
Yang P, Tang W, Li H, Hua R, Yuan Y, Zhang Y, Zhu Y, Cui Y, Sha J. T-complex protein 1 subunit zeta-2 (CCT6B) deficiency induces murine teratospermia. PeerJ 2021; 9:e11545. [PMID: 34141486 PMCID: PMC8176918 DOI: 10.7717/peerj.11545] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Background The CCT complex is an important mediator of microtubule assembly and intracellular protein folding. Owing to its high expression in spermatids, CCT knockdown can disrupt spermatogenesis. In the present report, we therefore evaluated the in vivo functionality of the testis-specific CCT complex component CCT6B using a murine knockout model system. Methods A CRISPR/Cas9 approach was used to generate Cct6b−/− mice, after which candidate gene expression in these animals was evaluated via qPCR and Western blotting. Testicular and epididymal phenotypes were assessed through histological and immunofluorescent staining assays, while a computer-assisted sperm analyzer was employed to assess semen quality. Results Cct6b−/− mice were successfully generated, and exhibited no differences in development, fertility, appearance, testis weight, or sperm counts relative to control littermates. In addition, no differences in spermatogenesis were detected when comparingCct6b+/+ and Cct6b−/− testes. However, when progressive motility was analyzed, the ratio of normal sperm was significantly decreased in Cct6b−/− male mice, with nuclear base bending being the primary detected abnormality. In addition, slight decreases in Cct4 and Cct7 expression were detected. Conclusion These data indicated that CCT6B is an important regulator of murine spermatogenesis, with the loss of this protein resulting in CCT complex dysfunction, providing a foundation for further studies.
Collapse
Affiliation(s)
- Peiyin Yang
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjing Tang
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huiling Li
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Hua
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Yuan
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Zhang
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yunfei Zhu
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiqiang Cui
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiahao Sha
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Bertile F, Plumel M, Maes P, Hirschler A, Challet E. Daytime Restricted Feeding Affects Day-Night Variations in Mouse Cerebellar Proteome. Front Mol Neurosci 2021; 14:613161. [PMID: 33912010 PMCID: PMC8072461 DOI: 10.3389/fnmol.2021.613161] [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: 10/01/2020] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
The cerebellum harbors a circadian clock that can be shifted by scheduled mealtime and participates in behavioral anticipation of food access. Large-scale two-dimensional difference gel electrophoresis (2D-DIGE) combined with mass spectrometry was used to identify day–night variations in the cerebellar proteome of mice fed either during daytime or nighttime. Experimental conditions led to modified expression of 89 cerebellar proteins contained in 63 protein spots. Five and 33 spots were changed respectively by time-of-day or feeding conditions. Strikingly, several proteins of the heat-shock protein family (i.e., Hsp90aa1, 90ab1, 90b1, and Hspa2, 4, 5, 8, 9) were down-regulated in the cerebellum of daytime food-restricted mice. This was also the case for brain fatty acid protein (Fabp7) and enzymes involved in oxidative phosphorylation (Ndufs1) or folate metabolism (Aldh1l1). In contrast, aldolase C (Aldoc or zebrin II) and pyruvate carboxylase (Pc), two enzymes involved in carbohydrate metabolism, and vesicle-fusing ATPase (Nsf) were up-regulated during daytime restricted feeding, possibly reflecting increased neuronal activity. Significant feeding × time-of-day interactions were found for changes in the intensity of 20 spots. Guanine nucleotide-binding protein G(o) subunit alpha (Gnao1) was more expressed in the cerebellum before food access. Neuronal calcium-sensor proteins [i.e., parvalbumin (Pvalb) and visinin-like protein 1 (Vsnl1)] were inversely regulated in daytime food-restricted mice, compared to control mice fed at night. Furthermore, expression of three enzymes modulating the circadian clockwork, namely heterogeneous nuclear ribonucleoprotein K (Hnrnpk), serine/threonine-protein phosphatases 1 (Ppp1cc and Ppp1cb subunits) and 5 (Ppp5), was differentially altered by daytime restricted feeding. Besides cerebellar proteins affected only by feeding conditions or daily cues, specific changes in in protein abundance before food access may be related to behavioral anticipation of food access and/or feeding-induced shift of the cerebellar clockwork.
Collapse
Affiliation(s)
- Fabrice Bertile
- Institut Pluridisciplinaire Hubert Curien, LSMBO, Université de Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Marine Plumel
- Institut Pluridisciplinaire Hubert Curien, LSMBO, Université de Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Pauline Maes
- Institut Pluridisciplinaire Hubert Curien, LSMBO, Université de Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Aurélie Hirschler
- Institut Pluridisciplinaire Hubert Curien, LSMBO, Université de Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Etienne Challet
- Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| |
Collapse
|
11
|
Sadat A, Tiwari S, Verma K, Ray A, Ali M, Upadhyay V, Singh A, Chaphalkar A, Ghosh A, Chakraborty R, Chakraborty K, Mapa K. GROEL/ES Buffers Entropic Traps in Folding Pathway during Evolution of a Model Substrate. J Mol Biol 2020; 432:5649-5664. [PMID: 32835659 DOI: 10.1016/j.jmb.2020.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
The folding landscape of proteins can change during evolution with the accumulation of mutations that may introduce entropic or enthalpic barriers in the protein folding pathway, making it a possible substrate of molecular chaperones in vivo. Can the nature of such physical barriers of folding dictate the feasibility of chaperone-assistance? To address this, we have simulated the evolutionary step to chaperone-dependence keeping GroEL/ES as the target chaperone and GFP as a model protein in an unbiased screen. We find that the mutation conferring GroEL/ES dependence in vivo and in vitro encode an entropic trap in the folding pathway rescued by the chaperonin. Additionally, GroEL/ES can edit the formation of non-native contacts similar to DnaK/J/E machinery. However, this capability is not utilized by the substrates in vivo. As a consequence, GroEL/ES caters to buffer mutations that predominantly cause entropic traps, despite possessing the capacity to edit both enthalpic and entropic traps in the folding pathway of the substrate protein.
Collapse
Affiliation(s)
- Anwar Sadat
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Satyam Tiwari
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Kanika Verma
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Arjun Ray
- Indraprastha Institute of Information Technology-Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India
| | - Mudassar Ali
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Vaibhav Upadhyay
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Anupam Singh
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Aseem Chaphalkar
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Asmita Ghosh
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Rahul Chakraborty
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Kausik Chakraborty
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Koyeli Mapa
- Academy of Scientific and Innovative Research, CSIR-HRDG, Ghaziabad, Uttar Pradesh 201002, India; Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, Gautam Buddha Nagar, Uttar Pradesh 201314, India.
| |
Collapse
|
12
|
Sadler E, Ryals MM, May LA, Martin D, Welsh N, Boger ET, Morell RJ, Hertzano R, Cunningham LL. Cell-Specific Transcriptional Responses to Heat Shock in the Mouse Utricle Epithelium. Front Cell Neurosci 2020; 14:123. [PMID: 32528249 PMCID: PMC7247426 DOI: 10.3389/fncel.2020.00123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022] Open
Abstract
Sensory epithelia of the inner ear contain mechanosensory hair cells (HCs) and glia-like supporting cells (SCs), both of which are required for hearing and balance functions. Each of these cell types has unique responses to ototoxic and cytoprotective stimuli. Non-lethal heat stress in the mammalian utricle induces heat shock proteins (HSPs) and protects against ototoxic drug-induced hair cell death. Induction of HSPs in the utricle demonstrates cell-type specificity at the protein level, with HSP70 induction occurring primarily in SCs, while HSP32 (also known as heme oxygenase 1, HMOX1) is induced primarily in resident macrophages. Neither of these HSPs are robustly induced in HCs, suggesting that HCs may have little capacity for induction of stress-induced protective responses. To determine the transcriptional responses to heat shock of these different cell types, we performed cell-type-specific transcriptional profiling using the RiboTag method, which allows for immunoprecipitation (IP) of actively translating mRNAs from specific cell types. RNA-Seq differential gene expression analyses demonstrated that the RiboTag method identified known cell type-specific markers as well as new markers for HCs and SCs. Gene expression differences suggest that HCs and SCs exhibit differential transcriptional heat shock responses. The chaperonin family member Cct8 was significantly enriched only in heat-shocked HCs, while Hspa1l (HSP70 family), and Hspb1 and Cryab (HSP27 and HSP20 families, respectively) were enriched only in SCs. Together our data indicate that HCs exhibit a limited but unique heat shock response, and SCs exhibit a broader and more robust transcriptional response to protective heat stress.
Collapse
Affiliation(s)
- Erica Sadler
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Matthew M Ryals
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States.,Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lindsey A May
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States.,Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Nora Welsh
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Erich T Boger
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Robert J Morell
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.,Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lisa L Cunningham
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
13
|
Gandhi J, Antonelli AC, Afridi A, Vatsia S, Joshi G, Romanov V, Murray IVJ, Khan SA. Protein misfolding and aggregation in neurodegenerative diseases: a review of pathogeneses, novel detection strategies, and potential therapeutics. Rev Neurosci 2019; 30:339-358. [PMID: 30742586 DOI: 10.1515/revneuro-2016-0035] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
Protein folding is a complex, multisystem process characterized by heavy molecular and cellular footprints. Chaperone machinery enables proper protein folding and stable conformation. Other pathways concomitant with the protein folding process include transcription, translation, post-translational modifications, degradation through the ubiquitin-proteasome system, and autophagy. As such, the folding process can go awry in several different ways. The pathogenic basis behind most neurodegenerative diseases is that the disruption of protein homeostasis (i.e. proteostasis) at any level will eventually lead to protein misfolding. Misfolded proteins often aggregate and accumulate to trigger neurotoxicity through cellular stress pathways and consequently cause neurodegenerative diseases. The manifestation of a disease is usually dependent on the specific brain region that the neurotoxicity affects. Neurodegenerative diseases are age-associated, and their incidence is expected to rise as humans continue to live longer and pursue a greater life expectancy. We presently review the sequelae of protein misfolding and aggregation, as well as the role of these phenomena in several neurodegenerative diseases including Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, Parkinson's disease, transmissible spongiform encephalopathies, and spinocerebellar ataxia. Strategies for treatment and therapy are also conferred with respect to impairing, inhibiting, or reversing protein misfolding.
Collapse
Affiliation(s)
- Jason Gandhi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Medical Student Research Institute, St. George's University School of Medicine, Grenada, West Indies
| | - Anthony C Antonelli
- Department of Pathology, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Adil Afridi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Sohrab Vatsia
- Department of Cardiothoracic Surgery, Lenox Hill Hospital, 130 East 77th Street, New York, NY 10075, USA
| | - Gunjan Joshi
- Department of Internal Medicine, Stony Brook Southampton Hospital, 240 Meeting House Lane, Southampton, NY 11968, USA
| | - Victor Romanov
- Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| | - Ian V J Murray
- Department of Physiology and Neuroscience, St. George's University School of Medicine, Grenada, West Indies
| | - Sardar Ali Khan
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| |
Collapse
|
14
|
Chaix A, Manoogian ENC, Melkani GC, Panda S. Time-Restricted Eating to Prevent and Manage Chronic Metabolic Diseases. Annu Rev Nutr 2019; 39:291-315. [PMID: 31180809 DOI: 10.1146/annurev-nutr-082018-124320] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Molecular clocks are present in almost every cell to anticipate daily recurring and predictable changes, such as rhythmic nutrient availability, and to adapt cellular functions accordingly. At the same time, nutrient-sensing pathways can respond to acute nutrient imbalance and modulate and orient metabolism so cells can adapt optimally to a declining or increasing availability of nutrients. Organismal circadian rhythms are coordinated by behavioral rhythms such as activity-rest and feeding-fasting cycles to temporally orchestrate a sequence of physiological processes to optimize metabolism. Basic research in circadian rhythms has largely focused on the functioning of the self-sustaining molecular circadian oscillator, while research in nutrition science has yielded insights into physiological responses to caloric deprivation or to specific macronutrients. Integration of these two fields into actionable new concepts in the timing of food intake has led to the emerging practice of time-restricted eating. In this paradigm, daily caloric intake is restricted to a consistent window of 8-12 h. This paradigm has pervasive benefits on multiple organ systems.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Emily N C Manoogian
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Girish C Melkani
- Molecular Biology Program and Heart Institute, Department of Biology, San Diego State University, San Diego, California 92182, USA
| | - Satchidananda Panda
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| |
Collapse
|
15
|
Yusof NA, Kamaruddin S, Abu Bakar FD, Mahadi NM, Abdul Murad AM. Structural and functional insights into TRiC chaperonin from a psychrophilic yeast, Glaciozyma antarctica. Cell Stress Chaperones 2019; 24:351-368. [PMID: 30649671 PMCID: PMC6439030 DOI: 10.1007/s12192-019-00969-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/29/2022] Open
Abstract
Studies on TCP1-1 ring complex (TRiC) chaperonin have shown its indispensable role in folding cytosolic proteins in eukaryotes. In a psychrophilic organism, extreme cold temperature creates a low-energy environment that potentially causes protein denaturation with loss of activity. We hypothesized that TRiC may undergo evolution in terms of its structural molecular adaptation in order to facilitate protein folding in low-energy environment. To test this hypothesis, we isolated G. antarctica TRiC (GaTRiC) and found that the expression of GaTRiC mRNA in G. antarctica was consistently expressed at all temperatures indicating their importance in cell regulation. Moreover, we showed GaTRiC has the ability of a chaperonin whereby denatured luciferase can be folded to the functional stage in its presence. Structurally, three categories of residue substitutions were found in α, β, and δ subunits: (i) bulky/polar side chains to alanine or valine, (ii) charged residues to alanine, and (iii) isoleucine to valine that would be expected to increase intramolecular flexibility within the GaTRiC. The residue substitutions observed in the built structures possibly affect the hydrophobic, hydrogen bonds, and ionic and aromatic interactions which lead to an increase in structural flexibility. Our structural and functional analysis explains some possible structural features which may contribute to cold adaptation of the psychrophilic TRiC folding chamber.
Collapse
Affiliation(s)
- Nur Athirah Yusof
- Biotechnology Research Institute, Universiti Malaysia Sabah, 88400, Kota Kinabalu, Sabah, Malaysia.
| | - Shazilah Kamaruddin
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Farah Diba Abu Bakar
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Nor Muhammad Mahadi
- Malaysia Genome Institute, Jalan Bangi, 43000, Kajang, Selangor Darul Ehsan, Malaysia
| | - Abdul Munir Abdul Murad
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| |
Collapse
|
16
|
Wang Q, Liu R, Li Q, Wang F, Zhu B, Zheng M, Cui H, Wen J, Zhao G. Host cell interactome of PB1 N40 protein of H5N1 influenza A virus in chicken cells. J Proteomics 2019; 197:34-41. [PMID: 30790688 DOI: 10.1016/j.jprot.2019.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/15/2019] [Accepted: 02/15/2019] [Indexed: 01/06/2023]
Abstract
H5N1 influenza A virus (IAV) causes seasonal epidemics that represent a worldwide threat to public health. IAV relies on host factors for viral replication. PB1 N40 is translated from the fifth starting code (AUG) of PB1 mRNA, which is the product of the ribosomal scan omission. Here, we report the interactome landscape of H5N1 IAV PB1 N40 protein in chicken cells. The interacting complexes were captured by co-immunoprecipitation and analyzed by mass spectrometry. We identified 135 proteins as PB1 N40-interacting proteins. GO and Pathway analysis showed that proteins with biological functions such as protein localization and viral transcription and proteins related to signaling pathways of DNA replication and cell cycle were significantly enriched in virus-host interactions, suggesting the potential roles of them in infection with H5N1 IAV. Comparative analysis among H1N1 and H5N1 revealed conservation of the virus-host protein interaction between different subtypes or strains of influenza virus. ARCN1 was identified as a host interacting factor of H5N1 IAV PB1 N40 protein, which is the component of the coatomer. Knockdown of ARCN1 significantly decreased the titer of H5N1 IAV in chicken cells. BIOLOGICAL SIGNIFICANCE: Influenza A virus (IAV) is a great threat to public health and avian production. However, the manner in which avian IAV recruits the host cellular machinery for replication and how the host antagonizes the IAV infection was previously poorly understood. Here we present the viral-host interactome of the H5N1 IAV PB1 N40 protein and reveal its involvement with dozens of important host genes during the course of IAV infection.
Collapse
Affiliation(s)
- Qiao Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ranran Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Qinghe Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fei Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Bo Zhu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Maiqing Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Huanxian Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Jie Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; State Key Laboratory of Animal Nutrition, Beijing, China
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; School of Life Science and Engineering, Foshan University, Foshan, China.
| |
Collapse
|
17
|
Abstract
The eukaryotic group II chaperonin TRiC/CCT assists the folding of 10% of cytosolic proteins including many key structural and regulatory proteins. TRiC plays an essential role in maintaining protein homeostasis, and dysfunction of TRiC is closely related to human diseases including cancer and neurodegenerative diseases. TRiC consists of eight paralogous subunits, each of which plays a specific role in the assembly, allosteric cooperativity, and substrate recognition and folding of this complex macromolecular machine. TRiC-mediated substrate folding is regulated through its ATP-driven conformational changes. In recent years, progresses have been made on the structure, subunit arrangement, conformational cycle, and substrate folding of TRiC. Additionally, accumulating evidences also demonstrate the linkage between TRiC oligomer or monomer and diseases. In this review, we focus on the TRiC structure itself, TRiC assisted substrate folding, TRiC and disease, and the potential therapeutic application of TRiC in various diseases.
Collapse
Affiliation(s)
- Mingliang Jin
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Caixuan Liu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenyu Han
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yao Cong
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
18
|
Das R, Bhattacharjee S, Patel AA, Harris JM, Bhattacharya S, Letcher JM, Clark SG, Nanda S, Iyer EPR, Ascoli GA, Cox DN. Dendritic Cytoskeletal Architecture Is Modulated by Combinatorial Transcriptional Regulation in Drosophila melanogaster. Genetics 2017; 207:1401-1421. [PMID: 29025914 PMCID: PMC5714456 DOI: 10.1534/genetics.117.300393] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/04/2017] [Indexed: 01/08/2023] Open
Abstract
Transcription factors (TFs) have emerged as essential cell autonomous mediators of subtype specific dendritogenesis; however, the downstream effectors of these TFs remain largely unknown, as are the cellular events that TFs control to direct morphological change. As dendritic morphology is largely dictated by the organization of the actin and microtubule (MT) cytoskeletons, elucidating TF-mediated cytoskeletal regulatory programs is key to understanding molecular control of diverse dendritic morphologies. Previous studies in Drosophila melanogaster have demonstrated that the conserved TFs Cut and Knot exert combinatorial control over aspects of dendritic cytoskeleton development, promoting actin and MT-based arbor morphology, respectively. To investigate transcriptional targets of Cut and/or Knot regulation, we conducted systematic neurogenomic studies, coupled with in vivo genetic screens utilizing multi-fluor cytoskeletal and membrane marker reporters. These analyses identified a host of putative Cut and/or Knot effector molecules, and a subset of these putative TF targets converge on modulating dendritic cytoskeletal architecture, which are grouped into three major phenotypic categories, based upon neuromorphometric analyses: complexity enhancer, complexity shifter, and complexity suppressor. Complexity enhancer genes normally function to promote higher order dendritic growth and branching with variable effects on MT stabilization and F-actin organization, whereas complexity shifter and complexity suppressor genes normally function in regulating proximal-distal branching distribution or in restricting higher order branching complexity, respectively, with spatially restricted impacts on the dendritic cytoskeleton. Collectively, we implicate novel genes and cellular programs by which TFs distinctly and combinatorially govern dendritogenesis via cytoskeletal modulation.
Collapse
Affiliation(s)
- Ravi Das
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | | | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Jenna M Harris
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | | | - Jamin M Letcher
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Sarah G Clark
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Sumit Nanda
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030
| | | | - Giorgio A Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| |
Collapse
|
19
|
Yeo KS, Tan MC, Lim YY, Ea CK. JMJD8 is a novel endoplasmic reticulum protein with a JmjC domain. Sci Rep 2017; 7:15407. [PMID: 29133832 PMCID: PMC5684140 DOI: 10.1038/s41598-017-15676-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/31/2017] [Indexed: 12/22/2022] Open
Abstract
Jumonji C (JmjC) domain-containing proteins have been shown to regulate cellular processes by hydroxylating or demethylating histone and non-histone targets. JMJD8 belongs to the JmjC domain-only family that was recently shown to be involved in angiogenesis and TNF-induced NF-κB signaling. Here, we employed bioinformatic analysis and immunofluorescence microscopy to examine the physiological properties of JMJD8. We demonstrated that JMJD8 localizes to the lumen of endoplasmic reticulum and that JMJD8 forms dimers or oligomers in vivo. Furthermore, we identified potential JMJD8-interacting proteins that are known to regulate protein complex assembly and protein folding. Taken together, this work demonstrates that JMJD8 is the first JmjC domain-containing protein found in the lumen of the endoplasmic reticulum that may function in protein complex assembly and protein folding.
Collapse
Affiliation(s)
- Kok Siong Yeo
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ming Cheang Tan
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Chee-Kwee Ea
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9148, United States.
| |
Collapse
|
20
|
Counts JT, Hester TM, Rouhana L. Genetic expansion of chaperonin-containing TCP-1 (CCT/TRiC) complex subunits yields testis-specific isoforms required for spermatogenesis in planarian flatworms. Mol Reprod Dev 2017; 84:1271-1284. [PMID: 29095551 DOI: 10.1002/mrd.22925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/16/2017] [Indexed: 12/23/2022]
Abstract
Chaperonin-containing Tail-less complex polypeptide 1 (CCT) is a highly conserved, hetero-oligomeric complex that ensures proper folding of actin, tubulin, and regulators of mitosis. Eight subunits (CCT1-8) make up this complex, and every subunit has a homolog expressed in the testes and somatic tissue of the planarian flatworm Schmidtea mediterranea. Gene duplications of four subunits in the genomes of S. mediterranea and other planarian flatworms created paralogs to CCT1, CCT3, CCT4, and CCT8 that are expressed exclusively in the testes. Functional analyses revealed that each CCT subunit expressed in the S. mediterranea soma is essential for homeostatic integrity and survival, whereas sperm elongation defects were observed upon knockdown of each individual testis-specific paralog (Smed-cct1B; Smed-cct3B; Smed-cct4A; and Smed-cct8B), regardless of potential redundancy with paralogs expressed in both testes and soma (Smed-cct1A; Smed-cct3A; Smed-cct4B; and Smed-cct8A). Yet, no detriment was observed in the number of adult somatic stem cells (neoblasts) that maintain differentiated tissue in planarians. Thus, expression of all eight CCT subunits is required to execute the essential functions of the CCT complex. Furthermore, expression of the somatic paralogs in planarian testes is not sufficient to complete spermatogenesis when testis-specific paralogs are knocked down, suggesting that the evolution of chaperonin subunits may drive changes in the development of sperm structure and that correct CCT subunit stoichiometry is crucial for spermiogenesis.
Collapse
Affiliation(s)
- Jenna T Counts
- Department of Biological Sciences, Wright State University, Dayton, Ohio
| | - Tasha M Hester
- Department of Biological Sciences, Wright State University, Dayton, Ohio
| | - Labib Rouhana
- Department of Biological Sciences, Wright State University, Dayton, Ohio
| |
Collapse
|
21
|
Melkani GC, Bhide S, Han A, Vyas J, Livelo C, Bodmer R, Bernstein SI. TRiC/CCT chaperonins are essential for maintaining myofibril organization, cardiac physiological rhythm, and lifespan. FEBS Lett 2017; 591:3447-3458. [PMID: 28963798 PMCID: PMC5683924 DOI: 10.1002/1873-3468.12860] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/08/2017] [Accepted: 09/19/2017] [Indexed: 01/12/2023]
Abstract
We recently reported that CCT chaperonin subunits are upregulated in a cardiac-specific manner under time-restricted feeding (TRF) [Gill S et al. (2015) Science 347, 1265-1269], suggesting that TRiC/CCT has a heart-specific function. To understand the CCT chaperonin function in cardiomyocytes, we performed its cardiac-specific knock-down in the Drosophila melanogaster model. This resulted in disorganization of cardiac actin- and myosin-containing myofibrils and severe physiological dysfunction, including restricted heart diameters, elevated cardiac dysrhythmia and compromised cardiac performance. We also noted that cardiac-specific knock-down of CCT chaperonin significantly shortens lifespans. Additionally, disruption of circadian rhythm yields further deterioration of cardiac function of hypomorphic CCT mutants. Our analysis reveals that both the orchestration of protein folding and circadian rhythms mediated by CCT chaperonin are critical for maintaining heart contractility.
Collapse
Affiliation(s)
- Girish C. Melkani
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Shruti Bhide
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Andrew Han
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Jay Vyas
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Catherine Livelo
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sanford I. Bernstein
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| |
Collapse
|
22
|
Chatzileontiadou DSM, Samiotaki M, Alexopoulou AN, Cotsiki M, Panayotou G, Stamatiadi M, Balatsos NAA, Leonidas DD, Kontou M. Proteomic Analysis of Human Angiogenin Interactions Reveals Cytoplasmic PCNA as a Putative Binding Partner. J Proteome Res 2017; 16:3606-3622. [PMID: 28777577 DOI: 10.1021/acs.jproteome.7b00335] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human Angiogenin (hAng) is a member of the ribonuclease A superfamily and a potent inducer of neovascularization. Protein interactions of hAng in the nucleus and cytoplasm of the human umbilical vein cell line EA.hy926 have been investigated by mass spectroscopy. Data are available via ProteomeXchange with identifiers PXD006583 and PXD006584. The first gel-free analysis of hAng immunoprecipitates revealed many statistically significant potential hAng-interacting proteins involved in crucial biological pathways. Surprisingly, proliferating cell nuclear antigen (PCNA), was found to be immunoprecipitated with hAng only in the cytoplasm. The hAng-PCNA interaction and colocalization in the specific cellular compartment was validated with immunoprecipitation, immunoblotting, and immunocytochemistry. The results revealed that PCNA is predominantly localized in the cytoplasm, while hAng is distributed both in the nucleus and in the cytoplasm. hAng and PCNA colocalize in the cytoplasm, suggesting that they may interact in this compartment.
Collapse
Affiliation(s)
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | | | - Marina Cotsiki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - George Panayotou
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - Melina Stamatiadi
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Nikolaos A A Balatsos
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Demetres D Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Maria Kontou
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| |
Collapse
|
23
|
Álvarez-Satta M, Castro-Sánchez S, Valverde D. Bardet-Biedl Syndrome as a Chaperonopathy: Dissecting the Major Role of Chaperonin-Like BBS Proteins (BBS6-BBS10-BBS12). Front Mol Biosci 2017; 4:55. [PMID: 28824921 PMCID: PMC5534436 DOI: 10.3389/fmolb.2017.00055] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023] Open
Abstract
Bardet-Biedl syndrome (BBS) is a rare genetic disorder that belongs to the group of ciliopathies, defined as diseases caused by defects in cilia structure and/or function. The six diagnostic features considered for this syndrome include retinal dystrophy, obesity, polydactyly, cognitive impairment and renal and urogenital anomalies. Furthermore, three of the 21 genes currently known to be involved in BBS encode chaperonin-like proteins (MKKS/BBS6, BBS10, and BBS12), so BBS can be also considered a member of the growing group of chaperonopathies. Remarkably, up to 50% of clinically-diagnosed BBS families can harbor disease-causing variants in these three genes, which highlights the importance of chaperone defects as pathogenic factors even for genetically heterogeneous syndromes such as BBS. In addition, it is interesting to note that BBS families with deleterious variants in MKKS/BBS6, BBS10 or BBS12 genes generally display more severe phenotypes than families with changes in other BBS genes. The chaperonin-like BBS proteins have structural homology to the CCT family of group II chaperonins, although they are believed to conserve neither the ATP-dependent folding activity of canonical CCT chaperonins nor the ability to form CCT-like oligomeric complexes. Thus, they play an important role in the initial steps of assembly of the BBSome, which is a multiprotein complex essential for mediating the ciliary trafficking activity. In this review, we present a comprehensive review of those genetic, functional and evolutionary aspects concerning chaperonin-like BBS proteins, trying to provide a new perspective that expands the classical conception of BBS only from a ciliary point of view.
Collapse
Affiliation(s)
- María Álvarez-Satta
- Grupo de Biomarcadores Moleculares, Departamento de Bioquímica, Genética e Inmunología, Facultad de Biología, Universidad de VigoVigo, Spain.,Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGOVigo, Spain.,Centro de Investigaciones Biomédicas (Centro Singular de Investigación de Galicia 2016-2019), Universidad de VigoVigo, Spain
| | - Sheila Castro-Sánchez
- Grupo de Biomarcadores Moleculares, Departamento de Bioquímica, Genética e Inmunología, Facultad de Biología, Universidad de VigoVigo, Spain.,Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGOVigo, Spain.,Centro de Investigaciones Biomédicas (Centro Singular de Investigación de Galicia 2016-2019), Universidad de VigoVigo, Spain
| | - Diana Valverde
- Grupo de Biomarcadores Moleculares, Departamento de Bioquímica, Genética e Inmunología, Facultad de Biología, Universidad de VigoVigo, Spain.,Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGOVigo, Spain.,Centro de Investigaciones Biomédicas (Centro Singular de Investigación de Galicia 2016-2019), Universidad de VigoVigo, Spain
| |
Collapse
|
24
|
Pinna S, Pasella S, Deiana M, Baralla A, Mannu A, Masala AGE, Pileri PV, Deiana N, Scognamillo F, Pala C, Zinellu A, Carru C, Deiana L. Proteomic analysis of human plasma and peripheral blood mononuclear cells in Systemic Lupus Erythematosus patients. J Immunol Methods 2017; 446:37-46. [DOI: 10.1016/j.jim.2017.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/09/2017] [Accepted: 03/28/2017] [Indexed: 01/22/2023]
|
25
|
Abstract
In this review we discuss the history and the current state of ideas related to the mechanism of size regulation of the thick (myosin) and thin (actin) filaments in vertebrate striated muscles. Various hypotheses have been considered during of more than half century of research, recently mostly involving titin and nebulin acting as templates or 'molecular rulers', terminating exact assembly. These two giant, single-polypeptide, filamentous proteins are bound in situ along the thick and thin filaments, respectively, with an almost perfect match in the respective lengths and structural periodicities. However, evidence still questions the possibility that the proteins function as templates, or scaffolds, on which the thin and thick filaments could be assembled. In addition, the progress in muscle research during the last decades highlighted a number of other factors that could potentially be involved in the mechanism of length regulation: molecular chaperones that may guide folding and assembly of actin and myosin; capping proteins that can influence the rates of assembly-disassembly of the myofilaments; Ca2+ transients that can activate or deactivate protein interactions, etc. The entire mechanism of sarcomere assembly appears complex and highly dynamic. This mechanism is also capable of producing filaments of about the correct size without titin and nebulin. What then is the role of these proteins? Evidence points to titin and nebulin stabilizing structures of the respective filaments. This stabilizing effect, based on linear proteins of a fixed size, implies that titin and nebulin are indeed molecular rulers of the filaments. Although the proteins may not function as templates in the assembly of the filaments, they measure and stabilize exactly the same size of the functionally important for the muscles segments in each of the respective filaments.
Collapse
|
26
|
Kaufmann T, Kukolj E, Brachner A, Beltzung E, Bruno M, Kostrhon S, Opravil S, Hudecz O, Mechtler K, Warren G, Slade D. SIRT2 regulates nuclear envelope reassembly through ANKLE2 deacetylation. J Cell Sci 2016; 129:4607-4621. [PMID: 27875273 PMCID: PMC5201015 DOI: 10.1242/jcs.192633] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/09/2016] [Indexed: 12/18/2022] Open
Abstract
Sirtuin 2 (SIRT2) is an NAD-dependent deacetylase known to regulate microtubule dynamics and cell cycle progression. SIRT2 has also been implicated in the pathology of cancer, neurodegenerative diseases and progeria. Here, we show that SIRT2 depletion or overexpression causes nuclear envelope reassembly defects. We link this phenotype to the recently identified regulator of nuclear envelope reassembly ANKLE2. ANKLE2 acetylation at K302 and phosphorylation at S662 are dynamically regulated throughout the cell cycle by SIRT2 and are essential for normal nuclear envelope reassembly. The function of SIRT2 therefore extends beyond the regulation of microtubules to include the regulation of nuclear envelope dynamics.
Collapse
Affiliation(s)
- Tanja Kaufmann
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria.,Department of Molecular Biotechnology, University of Applied Sciences FH Campus Wien, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Eva Kukolj
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Andreas Brachner
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Etienne Beltzung
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Melania Bruno
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Sebastian Kostrhon
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Susanne Opravil
- Institute of Molecular Pathology, Dr Bohr-Gasse 7, Vienna 1030, Austria
| | - Otto Hudecz
- Institute of Molecular Pathology, Dr Bohr-Gasse 7, Vienna 1030, Austria
| | - Karl Mechtler
- Institute of Molecular Pathology, Dr Bohr-Gasse 7, Vienna 1030, Austria
| | - Graham Warren
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| | - Dea Slade
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, Vienna 1030, Austria
| |
Collapse
|
27
|
Dean B, Copolov D, Scarr E. Understanding the pathophysiology of schizophrenia: Contributions from the Melbourne Psychiatric Brain Bank. Schizophr Res 2016; 177:108-114. [PMID: 27184458 DOI: 10.1016/j.schres.2016.04.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/20/2016] [Accepted: 04/23/2016] [Indexed: 11/24/2022]
Abstract
The Melbourne Psychiatric Brain Bank came into existence 25years ago. This review focusses on lines of research that have used tissue from the Brain Bank over periods of time. Hence there is a discussion on the significance of changes in levels of serotonin 2A receptors in the cortex of patients with schizophrenia and the relevance of such changes with regards to the pathophysiology of the disorder. The extensive contribution made by studies using tissue from the Melbourne Psychiatric Brain Bank to understanding the role of muscarinic receptors in the pathophysiology and treatment of schizophrenia is summarised. Finally, findings using brain bank tissue and "omics" technologies are reviewed. In each case, findings using tissue from the Melbourne Psychiatric Brain Bank is placed in context with research carried out on human postmortem CNS in schizophrenia and with findings in other lines of research that can help explain the causes or consequences of changes in CNS molecular cytoarchitecture. This timely review of data from the Melbourne Psychiatric Brain Bank reinforces the challenges faced in trying to increase our understanding of the molecular pathophysiology of schizophrenia. Continuing to increase our understanding of the disorder is important as a precursor to identifying new drug targets that can be exploited to improve the treatment of a disorder where treatment resistance remains a significant problem (Millan et al., 2016).
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.
| | - David Copolov
- Office of the Vice-Chancellor and President, Monash University, Clayton, Victoria, Australia
| | - Elizabeth Scarr
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
28
|
TRiC subunits enhance BDNF axonal transport and rescue striatal atrophy in Huntington's disease. Proc Natl Acad Sci U S A 2016; 113:E5655-64. [PMID: 27601642 DOI: 10.1073/pnas.1603020113] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Corticostriatal atrophy is a cardinal manifestation of Huntington's disease (HD). However, the mechanism(s) by which mutant huntingtin (mHTT) protein contributes to the degeneration of the corticostriatal circuit is not well understood. We recreated the corticostriatal circuit in microfluidic chambers, pairing cortical and striatal neurons from the BACHD model of HD and its WT control. There were reduced synaptic connectivity and atrophy of striatal neurons in cultures in which BACHD cortical and striatal neurons were paired. However, these changes were prevented if WT cortical neurons were paired with BACHD striatal neurons; synthesis and release of brain-derived neurotrophic factor (BDNF) from WT cortical axons were responsible. Consistent with these findings, there was a marked reduction in anterograde transport of BDNF in BACHD cortical neurons. Subunits of the cytosolic chaperonin T-complex 1 (TCP-1) ring complex (TRiC or CCT for chaperonin containing TCP-1) have been shown to reduce mHTT levels. Both CCT3 and the apical domain of CCT1 (ApiCCT1) decreased the level of mHTT in BACHD cortical neurons. In cortical axons, they normalized anterograde BDNF transport, restored retrograde BDNF transport, and normalized lysosomal transport. Importantly, treating BACHD cortical neurons with ApiCCT1 prevented BACHD striatal neuronal atrophy by enhancing release of BDNF that subsequently acts through tyrosine receptor kinase B (TrkB) receptor on striatal neurons. Our findings are evidence that TRiC reagent-mediated reductions in mHTT enhanced BDNF delivery to restore the trophic status of BACHD striatal neurons.
Collapse
|
29
|
Wang Q, Li Q, Liu R, Zheng M, Wen J, Zhao G. Host cell interactome of PA protein of H5N1 influenza A virus in chicken cells. J Proteomics 2016; 136:48-54. [DOI: 10.1016/j.jprot.2016.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/18/2016] [Accepted: 01/26/2016] [Indexed: 01/07/2023]
|
30
|
Völker-Albert MC, Pusch MC, Fedisch A, Schilcher P, Schmidt A, Imhof A. A Quantitative Proteomic Analysis of In Vitro Assembled Chromatin. Mol Cell Proteomics 2016; 15:945-59. [PMID: 26811354 PMCID: PMC4813712 DOI: 10.1074/mcp.m115.053553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 01/17/2016] [Indexed: 12/30/2022] Open
Abstract
The structure of chromatin is critical for many aspects of cellular physiology and is considered to be the primary medium to store epigenetic information. It is defined by the histone molecules that constitute the nucleosome, the positioning of the nucleosomes along the DNA and the non-histone proteins that associate with it. These factors help to establish and maintain a largely DNA sequence-independent but surprisingly stable structure. Chromatin is extensively disassembled and reassembled during DNA replication, repair, recombination or transcription in order to allow the necessary factors to gain access to their substrate. Despite such constant interference with chromatin structure, the epigenetic information is generally well maintained. Surprisingly, the mechanisms that coordinate chromatin assembly and ensure proper assembly are not particularly well understood. Here, we use label free quantitative mass spectrometry to describe the kinetics of in vitro assembled chromatin supported by an embryo extract prepared from preblastoderm Drosophila melanogaster embryos. The use of a data independent acquisition method for proteome wide quantitation allows a time resolved comparison of in vitro chromatin assembly. A comparison of our in vitro data with proteomic studies of replicative chromatin assembly in vivo reveals an extensive overlap showing that the in vitro system can be used for investigating the kinetics of chromatin assembly in a proteome-wide manner.
Collapse
Affiliation(s)
- Moritz Carl Völker-Albert
- From the ‡BioMedical Center and Center for Integrated Protein Sciences Munich, Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| | - Miriam Caroline Pusch
- From the ‡BioMedical Center and Center for Integrated Protein Sciences Munich, Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| | - Andreas Fedisch
- From the ‡BioMedical Center and Center for Integrated Protein Sciences Munich, Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| | - Pierre Schilcher
- §Zentrallabor für Proteinanalytik (Protein Analyis Unit), Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| | - Andreas Schmidt
- §Zentrallabor für Proteinanalytik (Protein Analyis Unit), Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| | - Axel Imhof
- From the ‡BioMedical Center and Center for Integrated Protein Sciences Munich, Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany; §Zentrallabor für Proteinanalytik (Protein Analyis Unit), Ludwig-Maximilians-University of Munich, Groβhaderner Straβe 9, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
31
|
Trotter B, Otte KA, Schoppmann K, Hemmersbach R, Fröhlich T, Arnold GJ, Laforsch C. The influence of simulated microgravity on the proteome of Daphnia magna. NPJ Microgravity 2015; 1:15016. [PMID: 28725717 PMCID: PMC5515502 DOI: 10.1038/npjmgrav.2015.16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/02/2015] [Accepted: 08/11/2015] [Indexed: 01/04/2023] Open
Abstract
Background: The waterflea Daphnia is an interesting candidate for bioregenerative life support systems (BLSS). These animals are particularly promising because of their central role in the limnic food web and its mode of reproduction. However, the response of Daphnia to altered gravity conditions has to be investigated, especially on the molecular level, to evaluate the suitability of Daphnia for BLSS in space. Methods: In this study, we applied a proteomic approach to identify key proteins and pathways involved in the response of Daphnia to simulated microgravity generated by a two-dimensional (2D) clinostat. We analyzed five biological replicates using 2D-difference gel electrophoresis proteomic analysis. Results: We identified 109 protein spots differing in intensity (P<0.05). Substantial fractions of these proteins are involved in actin microfilament organization, indicating the disruption of cytoskeletal structures during clinorotation. Furthermore, proteins involved in protein folding were identified, suggesting altered gravity induced breakdown of protein structures in general. In addition, simulated microgravity increased the abundance of energy metabolism-related proteins, indicating an enhanced energy demand of Daphnia. Conclusions: The affected biological processes were also described in other studies using different organisms and systems either aiming to simulate microgravity conditions or providing real microgravity conditions. Moreover, most of the Daphnia protein sequences are well-conserved throughout taxa, indicating that the response to altered gravity conditions in Daphnia follows a general concept. Data are available via ProteomeXchange with identifier PXD002096.
Collapse
Affiliation(s)
- Benjamin Trotter
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Animal Ecology I and BayCEER, Bayreuth University, Bayreuth, Germany
| | - Kathrin A Otte
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Animal Ecology I and BayCEER, Bayreuth University, Bayreuth, Germany
| | | | - Ruth Hemmersbach
- Biomedical Research, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | | |
Collapse
|
32
|
Gill S, Le HD, Melkani GC, Panda S. Time-restricted feeding attenuates age-related cardiac decline in Drosophila. Science 2015; 347:1265-9. [PMID: 25766238 DOI: 10.1126/science.1256682] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Circadian clocks orchestrate periods of rest or activity and feeding or fasting over the course of a 24-hour day and maintain homeostasis. To assess whether a consolidated 24-hour cycle of feeding and fasting can sustain health, we explored the effect of time-restricted feeding (TRF; food access limited to daytime 12 hours every day) on neural, peripheral, and cardiovascular physiology in Drosophila melanogaster. We detected improved sleep, prevention of body weight gain, and deceleration of cardiac aging under TRF, even when caloric intake and activity were unchanged. We used temporal gene expression profiling and validation through classical genetics to identify the TCP-1 ring complex (TRiC) chaperonin, the mitochondrial electron transport chain complexes, and the circadian clock as pathways mediating the benefits of TRF.
Collapse
Affiliation(s)
- Shubhroz Gill
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA. Division of Biological Sciences, University of California San Diego, La Jolla, CA 92037, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Girish C Melkani
- Departments of Biology and Molecular Biology, and Heart Institutes, San Diego State University, San Diego, CA 92182, USA.
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
33
|
Abstract
How chaperonins orchestrate the successful folding of even the most elaborate of proteins is a question of central importance. In two recent studies in Cell by Joachimiak et al. and Freund et al., a new class of TRiC substrate is identified, and how the chaperonin exploits its different subunits to extend its substrate repertoire and direct productive folding is revealed.
Collapse
Affiliation(s)
- Anastasia Zhuravleva
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
34
|
Derry MM, Somasagara RR, Raina K, Kumar S, Gomez J, Patel M, Agarwal R, Agarwal C. Target identification of grape seed extract in colorectal cancer using drug affinity responsive target stability (DARTS) technique: role of endoplasmic reticulum stress response proteins. Curr Cancer Drug Targets 2015; 14:323-36. [PMID: 24724981 DOI: 10.2174/1568009614666140411101942] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 12/21/2022]
Abstract
Various natural agents, including grape seed extract (GSE), have shown considerable chemopreventive and anti-cancer efficacy against different cancers in pre-clinical studies; however, their specific protein targets are largely unknown and thus, their clinical usefulness is marred by limited scientific evidences about their direct cellular targets. Accordingly, herein, employing, for the first time, the recently developed drug affinity responsive target stability (DARTS) technique, we aimed to profile the potential protein targets of GSE in human colorectal cancer (CRC) cells. Unlike other methods, which can cause chemical alteration of the drug components to allow for detection, this approach relies on the fact that a drug bound protein may become less susceptible to proteolysis and hence the enriched proteins can be detected by Mass Spectroscopy methods. Our results, utilizing the DARTS technique followed by examination of the spectral output by LC/MS and the MASCOT data, revealed that GSE targets endoplasmic reticulum (ER) stress response proteins resulting in overall down regulation of proteins involved in translation and that GSE also causes oxidative protein modifications, specifically on methionine amino acids residues on its protein targets. Corroborating these findings, mechanistic studies revealed that GSE indeed caused ER stress and strongly inhibited PI3k-Akt-mTOR pathway for its biological effects in CRC cells. Furthermore, bioenergetics studies indicated that GSE also interferes with glycolysis and mitochondrial metabolism in CRC cells. Together, the present study identifying GSE molecular targets in CRC cells, combined with its efficacy in vast pre-clinical CRC models, further supports its usefulness for CRC prevention and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO 80045, USA.
| |
Collapse
|
35
|
Miyata Y, Shibata T, Aoshima M, Tsubata T, Nishida E. The molecular chaperone TRiC/CCT binds to the Trp-Asp 40 (WD40) repeat protein WDR68 and promotes its folding, protein kinase DYRK1A binding, and nuclear accumulation. J Biol Chem 2014; 289:33320-32. [PMID: 25342745 PMCID: PMC4246089 DOI: 10.1074/jbc.m114.586115] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 10/18/2014] [Indexed: 11/06/2022] Open
Abstract
Trp-Asp (WD) repeat protein 68 (WDR68) is an evolutionarily conserved WD40 repeat protein that binds to several proteins, including dual specificity tyrosine phosphorylation-regulated protein kinase (DYRK1A), MAPK/ERK kinase kinase 1 (MEKK1), and Cullin4-damage-specific DNA-binding protein 1 (CUL4-DDB1). WDR68 affects multiple and diverse physiological functions, such as controlling anthocyanin synthesis in plants, tissue growth in insects, and craniofacial development in vertebrates. However, the biochemical basis and the regulatory mechanism of WDR68 activity remain largely unknown. To better understand the cellular function of WDR68, here we have isolated and identified cellular WDR68 binding partners using a phosphoproteomic approach. More than 200 cellular proteins with wide varieties of biochemical functions were identified as WDR68-binding protein candidates. Eight T-complex protein 1 (TCP1) subunits comprising the molecular chaperone TCP1 ring complex/chaperonin-containing TCP1 (TRiC/CCT) were identified as major WDR68-binding proteins, and phosphorylation sites in both WDR68 and TRiC/CCT were identified. Co-immunoprecipitation experiments confirmed the binding between TRiC/CCT and WDR68. Computer-aided structural analysis suggested that WDR68 forms a seven-bladed β-propeller ring. Experiments with a series of deletion mutants in combination with the structural modeling showed that three of the seven β-propeller blades of WDR68 are essential and sufficient for TRiC/CCT binding. Knockdown of cellular TRiC/CCT by siRNA caused an abnormal WDR68 structure and led to reduction of its DYRK1A-binding activity. Concomitantly, nuclear accumulation of WDR68 was suppressed by the knockdown of TRiC/CCT, and WDR68 formed cellular aggregates when overexpressed in the TRiC/CCT-deficient cells. Altogether, our results demonstrate that the molecular chaperone TRiC/CCT is essential for correct protein folding, DYRK1A binding, and nuclear accumulation of WDR68.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan and
| | | | | | | | - Eisuke Nishida
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan and
| |
Collapse
|
36
|
Bocchini CE, Kasembeli MM, Roh SH, Tweardy DJ. Contribution of chaperones to STAT pathway signaling. JAKSTAT 2014; 3:e970459. [PMID: 26413421 DOI: 10.4161/21623988.2014.970459] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/21/2014] [Accepted: 09/25/2014] [Indexed: 12/27/2022] Open
Abstract
Aberrant STAT signaling is associated with the development and progression of many cancers and immune related diseases. Recent findings demonstrate that proteostasis modulators under clinical investigation for cancer therapy have a significant impact on STAT signaling, which may be critical for mediating their anti-cancer effects. Chaperones are critical for protein folding, stability and function and, thus, play an essential role in the maintenance of proteostasis. In this review we discuss the role of chaperones in STAT and tyrosine kinase (TK) protein folding, modulation of STAT and TK activity, and degradation of TKs. We highlight the important role of chaperones in STAT signaling, and how this knowledge has provided a framework for the development of new therapeutic avenues of targeting STAT signaling related pathologies.
Collapse
Affiliation(s)
- Claire E Bocchini
- Section of Infectious Disease; Department of Pediatrics; Baylor College of Medicine ; Houston, TX USA
| | - Moses M Kasembeli
- Section of Infectious Disease; Department of Medicine; Baylor College of Medicine ; Houston, TX USA
| | - Soung-Hun Roh
- Department of Biochemistry & Molecular Biology; Baylor College of Medicine ; Houston, TX USA
| | - David J Tweardy
- Section of Infectious Disease; Department of Medicine; Baylor College of Medicine ; Houston, TX USA ; Department of Biochemistry & Molecular Biology; Baylor College of Medicine ; Houston, TX USA ; Department of Molecular & Cellular Biology; Baylor College of Medicine ; Houston, TX USA
| |
Collapse
|
37
|
Protein folding, misfolding and quality control: the role of molecular chaperones. Essays Biochem 2014; 56:53-68. [DOI: 10.1042/bse0560053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cells have to cope with stressful conditions and adapt to changing environments. Heat stress, heavy metal ions or UV stress induce damage to cellular proteins and disturb the balanced status of the proteome. The adjusted balance between folded and folding proteins, called protein homoeostasis, is required for every aspect of cellular functionality. Protective proteins called chaperones are expressed under extreme conditions in order to prevent aggregation of cellular proteins and safeguard protein quality. These chaperones co-operate during de novo folding, refolding and disaggregation of damaged proteins and in many cases refold them to their functional state. Even under physiological conditions these machines support protein homoeostasis and maintain the balance between de novo folding and degradation. Mutations generating unstable proteins, which are observed in numerous human diseases such as Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis and cystic fibrosis, also challenge the protein quality control system. A better knowledge of how the protein homoeostasis system is regulated will lead to an improved understanding of these diseases and provide potential targets for therapy.
Collapse
|
38
|
Kasembeli M, Lau WCY, Roh SH, Eckols TK, Frydman J, Chiu W, Tweardy DJ. Modulation of STAT3 folding and function by TRiC/CCT chaperonin. PLoS Biol 2014; 12:e1001844. [PMID: 24756126 PMCID: PMC3995649 DOI: 10.1371/journal.pbio.1001844] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/17/2014] [Indexed: 02/06/2023] Open
Abstract
Levels, folding, and function of the infamous cancer and inflammatory disease-related signaling molecule Stat3 are regulated by interaction with the chaperonin TRiC; manipulation of this interaction is a therapeutic avenue for exploration. Signal transducer and activator of transcription 3 (Stat3) transduces signals of many peptide hormones from the cell surface to the nucleus and functions as an oncoprotein in many types of cancers, yet little is known about how it achieves its native folded state within the cell. Here we show that Stat3 is a novel substrate of the ring-shaped hetero-oligomeric eukaryotic chaperonin, TRiC/CCT, which contributes to its biosynthesis and activity in vitro and in vivo. TRiC binding to Stat3 was mediated, at least in part, by TRiC subunit CCT3. Stat3 binding to TRiC mapped predominantly to the β-strand rich, DNA-binding domain of Stat3. Notably, enhancing Stat3 binding to TRiC by engineering an additional TRiC-binding domain from the von Hippel-Lindau protein (vTBD), at the N-terminus of Stat3, further increased its affinity for TRiC as well as its function, as determined by Stat3's ability to bind to its phosphotyrosyl-peptide ligand, an interaction critical for Stat3 activation. Thus, Stat3 levels and function are regulated by TRiC and can be modulated by manipulating its interaction with TRiC. Stat3 is a multidomain transcription factor that contributes to many cellular functions by transmitting signals for over 40 peptide hormones from the cell surface to the nucleus. Understanding how multidomain proteins achieve their fully folded and functional state is of substantial biological interest. As Stat3 signaling is up-regulated in many pathological conditions, including cancer and inflammatory diseases, insight into what controls its folding may be useful for the identification of vulnerabilities that can be therapeutically exploited. We demonstrate that the major protein-folding machine or chaperonin within eukaryotic cells, TRiC/CCT, is required for Stat3 to fold during its synthesis and for Stat3 to be fully functional within the cell. We also find that TRiC can refold chemically denatured Stat3 and provide evidence that the CCT3 subunit of TRiC binds to the DNA-binding domain of Stat3. We also show that Stat3 activity is decreased by down-modulating levels of TRiC and can be increased by increasing Stat3's interaction with TRiC. TRiC therefore regulates both Stat3 protein levels and its function, making Stat3 modulation by manipulation of its interaction with TRiC a potential approach for the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Moses Kasembeli
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wilson Chun Yu Lau
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Soung-Hun Roh
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - T. Kris Eckols
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Judith Frydman
- Department of Biology and the BioX Program, Stanford University, Stanford, California, United States of America
| | - Wah Chiu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David J. Tweardy
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Kypri E, Christodoulou A, Maimaris G, Lethan M, Markaki M, Lysandrou C, Lederer CW, Tavernarakis N, Geimer S, Pedersen LB, Santama N. The nucleotide-binding proteins Nubp1 and Nubp2 are negative regulators of ciliogenesis. Cell Mol Life Sci 2014; 71:517-38. [PMID: 23807208 PMCID: PMC11113914 DOI: 10.1007/s00018-013-1401-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 12/12/2022]
Abstract
Nucleotide-binding proteins Nubp1 and Nubp2 are MRP/MinD-type P-loop NTPases with sequence similarity to bacterial division site-determining proteins and are conserved, essential proteins throughout the Eukaryotes. They have been implicated, together with their interacting minus-end directed motor protein KIFC5A, in the regulation of centriole duplication in mammalian cells. Here we show that Nubp1 and Nubp2 are integral components of centrioles throughout the cell cycle, recruited independently of KIFC5A. We further demonstrate their localization at the basal body of the primary cilium in quiescent vertebrate cells or invertebrate sensory cilia, as well as in the motile cilia of mouse cells and in the flagella of Chlamydomonas. RNAi-mediated silencing of nubp-1 in C. elegans causes the formation of morphologically aberrant and additional cilia in sensory neurons. Correspondingly, downregulation of Nubp1 or Nubp2 in mouse quiescent NIH 3T3 cells markedly increases the number of ciliated cells, while knockdown of KIFC5A dramatically reduces ciliogenesis. Simultaneous double silencing of Nubp1 + KIFC5A restores the percentage of ciliated cells to control levels. We document the normal ciliary recruitment, during these silencing regimes, of basal body proteins critical for ciliogenesis, namely CP110, CEP290, cenexin, Chibby, AurA, Rab8, and BBS7. Interestingly, we uncover novel interactions of Nubp1 with several members of the CCT/TRiC molecular chaperone complex, which we find enriched at the basal body and recruited independently of the Nubps or KIFC5A. Our combined results for Nubp1, Nubp2, and KIFC5A and their striking effects on cilium formation suggest a central regulatory role for these proteins, likely involving CCT/TRiC chaperone activity, in ciliogenesis.
Collapse
Affiliation(s)
- Elena Kypri
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Andri Christodoulou
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Giannis Maimaris
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Mette Lethan
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Markaki
- Institute of Molecular Biology and Biotechnology, Crete, Greece
| | - Costas Lysandrou
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | | | | | | | - Lotte B. Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niovi Santama
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| |
Collapse
|
40
|
Srinivasan S, Meyer RD, Lugo R, Rahimi N. Identification of PDCL3 as a novel chaperone protein involved in the generation of functional VEGF receptor 2. J Biol Chem 2013; 288:23171-81. [PMID: 23792958 DOI: 10.1074/jbc.m113.473173] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis, a hallmark step in tumor metastasis and ocular neovascularization, is driven primarily by the function of VEGF ligand on one of its receptors, VEGF receptor 2 (VEGFR-2). Central to the proliferation and ensuing angiogenesis of endothelial cells, the abundance of VEGFR-2 on the surface of endothelial cells is essential for VEGF to recognize and activate VEGFR-2. We have identified phosducin-like 3 (PDCL3, also known as PhLP2A), through a yeast two-hybrid system, as a novel protein involved in the stabilization of VEGFR-2 by serving as a chaperone. PDCL3 binds to the juxtamembrane domain of VEGFR-2 and controls the abundance of VEGFR-2 by inhibiting its ubiquitination and degradation. PDCL3 increases VEGF-induced tyrosine phosphorylation and is required for VEGFR-2-dependent endothelial capillary tube formation and proliferation. Taken together, our data provide strong evidence for the role of PDCL3 in angiogenesis and establishes the molecular mechanism by which it regulates VEGFR-2 expression and function.
Collapse
Affiliation(s)
- Srimathi Srinivasan
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
41
|
Guisbert E, Czyz DM, Richter K, McMullen PD, Morimoto RI. Identification of a tissue-selective heat shock response regulatory network. PLoS Genet 2013; 9:e1003466. [PMID: 23637632 PMCID: PMC3630107 DOI: 10.1371/journal.pgen.1003466] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 03/06/2013] [Indexed: 12/27/2022] Open
Abstract
The heat shock response (HSR) is essential to survive acute proteotoxic stress and has been studied extensively in unicellular organisms and tissue culture cells, but to a lesser extent in intact metazoan animals. To identify the regulatory pathways that control the HSR in Caenorhabditis elegans, we performed a genome-wide RNAi screen and identified 59 genes corresponding to 7 positive activators required for the HSR and 52 negative regulators whose knockdown leads to constitutive activation of the HSR. These modifiers function in specific steps of gene expression, protein synthesis, protein folding, trafficking, and protein clearance, and comprise the metazoan heat shock regulatory network (HSN). Whereas the positive regulators function in all tissues of C. elegans, nearly all of the negative regulators exhibited tissue-selective effects. Knockdown of the subunits of the proteasome strongly induces HS reporter expression only in the intestine and spermatheca but not in muscle cells, while knockdown of subunits of the TRiC/CCT chaperonin induces HS reporter expression only in muscle cells. Yet, both the proteasome and TRiC/CCT chaperonin are ubiquitously expressed and are required for clearance and folding in all tissues. We propose that the HSN identifies a key subset of the proteostasis machinery that regulates the HSR according to the unique functional requirements of each tissue. The heat shock response (HSR) is an essential stress response that functions to maintain protein folding homeostasis, or proteostasis, and whose critical role in human diseases is recently becoming apparent. Previously, most of our understanding of the HSR has come from cultured cells and unicellular organisms. Here we present the identification of the heat shock regulatory network (HSN) in Caenorhabditis elegans, an intact, multicellular organism, using genome-wide RNAi screening. We identify 59 positive and negative regulators of the HSR, all of which have a previously established role in proteostasis, linking the function of the HSR to its regulation. Some HSN genes were previously established in other systems, many were indirectly linked to HSR, and others are novel. Unexpectedly, almost all negative regulators of the HSR act in distinct, tissue-selective patterns, despite their broad expression and universal cellular requirements. Therefore, our data indicate that the HSN consists of a specific subset of the proteostasis machinery that functions to link the proteostasis network to HSR regulation in a tissue-selective manner.
Collapse
Affiliation(s)
- Eric Guisbert
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - Daniel M. Czyz
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - Klaus Richter
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - Patrick D. McMullen
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
42
|
Ludueña RF. A Hypothesis on the Origin and Evolution of Tubulin. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 302:41-185. [DOI: 10.1016/b978-0-12-407699-0.00002-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Ménoret A, Drew DA, Miyamoto S, Nakanishi M, Vella AT, Rosenberg DW. Differential proteomics identifies PDIA3 as a novel chemoprevention target in human colon cancer cells. Mol Carcinog 2012; 53 Suppl 1:E11-22. [PMID: 23255428 DOI: 10.1002/mc.21986] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 10/24/2012] [Accepted: 11/05/2012] [Indexed: 11/06/2022]
Abstract
Chemoprevention offers a promising strategy to prevent or delay the development of various cancers. Critical to this approach is the identification of molecular targets that may track with chemopreventive efficacy. To address this issue, we screened a panel of chemoprevention agents, including resveratrol, epigallocatechin-3-gallate, ursodeoxycholic acid, and sulindac sulfide for their effects on human colon cancer cell viability. Resveratrol elicited the most potent effect in HCT116 cells and was selected for further study. Proteomic PF 2D maps were generated from HCT116 cells treated with resveratrol versus vehicle alone. Analysis of proteomic maps using tandem mass spectrometry (MS) identified a panel of differentially modified proteins. Two proteins, actin and Hsp60, were previously shown in other cell culture systems to be affected by resveratrol, validating our approach. PDIA3, RPL19, histone H2B and TCP1β were uniquely identified by our proteomic discovery platform. PDIA3 was of particular interest given its potential role in regulating chemosensitivity of cancer cells. Total levels of PDIA3 in HCT116 cells were unchanged following 24 h of resveratrol treatment, confirmed by Western blot analysis. Immunoprecipitation of PDIA3 revealed a new set of client proteins following resveratrol treatment, including α, β, and δ-catenins, and cellular fractionation identified decreased nuclear localization of α-catenin by resveratrol. These data establish differential proteomic mapping as a powerful tool for identifying novel molecular targets of chemopreventive agents.
Collapse
Affiliation(s)
- Antoine Ménoret
- Department of Immunology, Colorectal Cancer Prevention Program University of Connecticut Health Center, Farmington, Connecticut
| | | | | | | | | | | |
Collapse
|
44
|
Müller M, Mazur AJ, Behrmann E, Diensthuber RP, Radke MB, Qu Z, Littwitz C, Raunser S, Schoenenberger CA, Manstein DJ, Mannherz HG. Functional characterization of the human α-cardiac actin mutations Y166C and M305L involved in hypertrophic cardiomyopathy. Cell Mol Life Sci 2012; 69:3457-79. [PMID: 22643837 PMCID: PMC11115188 DOI: 10.1007/s00018-012-1030-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 04/22/2012] [Accepted: 05/07/2012] [Indexed: 11/25/2022]
Abstract
Inherited cardiomyopathies are caused by point mutations in sarcomeric gene products, including α-cardiac muscle actin (ACTC1). We examined the biochemical and cell biological properties of the α-cardiac actin mutations Y166C and M305L identified in hypertrophic cardiomyopathy (HCM). Untagged wild-type (WT) cardiac actin, and the Y166C and M305L mutants were expressed by the baculovirus/Sf9-cell system and affinity purified by immobilized gelsolin G4-6. Their correct folding was verified by a number of assays. The mutant actins also displayed a disturbed intrinsic ATPase activity and an altered polymerization behavior in the presence of tropomyosin, gelsolin, and Arp2/3 complex. Both mutants stimulated the cardiac β-myosin ATPase to only 50 % of WT cardiac F-actin. Copolymers of WT and increasing amounts of the mutant actins led to a reduced stimulation of the myosin ATPase. Transfection of established cell lines revealed incorporation of EGFP- and hemagglutinin (HA)-tagged WT and both mutant actins into cytoplasmic stress fibers. Adenoviral vectors of HA-tagged WT and Y166C actin were successfully used to infect adult and neonatal rat cardiomyocytes (NRCs). The expressed HA-tagged actins were incorporated into the minus-ends of NRC thin filaments, demonstrating the ability to form hybrid thin filaments with endogenous actin. In NRCs, the Y166C mutant led after 72 h to a shortening of the sarcomere length when compared to NRCs infected with WT actin. Thus our data demonstrate that a mutant actin can be integrated into cardiomyocyte thin filaments and by its reduced mode of myosin interaction might be the basis for the initiation of HCM.
Collapse
Affiliation(s)
- Mirco Müller
- Institute for Biophysical Chemistry, OE 4350, Hannover Medical School, 30625 Hannover, Germany
| | - Antonina Joanna Mazur
- Department of Anatomy and Molecular Embryology, Ruhr-University, Universitätsstrasse 150, 44780 Bochum, Germany
- Present Address: Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 51-148 Wroclaw, Poland
| | - Elmar Behrmann
- Department of Physical Biochemistry, Max-Planck-Institute for Molecular Physiology, 44227 Dortmund, Germany
| | - Ralph P. Diensthuber
- Institute for Biophysical Chemistry, OE 4350, Hannover Medical School, 30625 Hannover, Germany
| | - Michael B. Radke
- Institute for Biophysical Chemistry, OE 4350, Hannover Medical School, 30625 Hannover, Germany
| | - Zheng Qu
- Department of Anatomy and Molecular Embryology, Ruhr-University, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Christoph Littwitz
- Department of Physiology, Stritch School of Medicine, Loyola University Chicago, Chicago, USA
| | - Stefan Raunser
- Department of Physical Biochemistry, Max-Planck-Institute for Molecular Physiology, 44227 Dortmund, Germany
| | - Cora-Ann Schoenenberger
- Maurice E. Müller Institute for Structural Biology, Biozentrum, University of Basel, 4046 Basel, Switzerland
| | - Dietmar J. Manstein
- Institute for Biophysical Chemistry, OE 4350, Hannover Medical School, 30625 Hannover, Germany
| | - Hans Georg Mannherz
- Department of Anatomy and Molecular Embryology, Ruhr-University, Universitätsstrasse 150, 44780 Bochum, Germany
- Department of Physical Biochemistry, Max-Planck-Institute for Molecular Physiology, 44227 Dortmund, Germany
| |
Collapse
|
45
|
Dun MD, Aitken RJ, Nixon B. The role of molecular chaperones in spermatogenesis and the post-testicular maturation of mammalian spermatozoa. Hum Reprod Update 2012; 18:420-35. [DOI: 10.1093/humupd/dms009] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
46
|
Proteomic analysis of Hemoglobin H-Constant Spring (Hb H-CS) erythroblasts. Blood Cells Mol Dis 2012; 48:77-85. [DOI: 10.1016/j.bcmd.2011.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/07/2011] [Indexed: 12/31/2022]
|
47
|
Chaperonin TRiC assists the refolding of sperm-specific glyceraldehyde-3-phosphate dehydrogenase. Arch Biochem Biophys 2011; 516:75-83. [DOI: 10.1016/j.abb.2011.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/12/2011] [Accepted: 09/28/2011] [Indexed: 11/17/2022]
|
48
|
Management of cytoskeleton architecture by molecular chaperones and immunophilins. Cell Signal 2011; 23:1907-20. [PMID: 21864675 DOI: 10.1016/j.cellsig.2011.07.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 11/20/2022]
Abstract
Cytoskeletal structure is continually remodeled to accommodate normal cell growth and to respond to pathophysiological cues. As a consequence, several cytoskeleton-interacting proteins become involved in a variety of cellular processes such as cell growth and division, cell movement, vesicle transportation, cellular organelle location and function, localization and distribution of membrane receptors, and cell-cell communication. Molecular chaperones and immunophilins are counted among the most important proteins that interact closely with the cytoskeleton network, in particular with microtubules and microtubule-associated factors. In several situations, heat-shock proteins and immunophilins work together as a functionally active heterocomplex, although both types of proteins also show independent actions. In circumstances where homeostasis is affected by environmental stresses or due to genetic alterations, chaperone proteins help to stabilize the system. Molecular chaperones facilitate the assembly, disassembly and/or folding/refolding of cytoskeletal proteins, so they prevent aberrant protein aggregation. Nonetheless, the roles of heat-shock proteins and immunophilins are not only limited to solve abnormal situations, but they also have an active participation during the normal differentiation process of the cell and are key factors for many structural and functional rearrangements during this course of action. Cytoskeleton modifications leading to altered localization of nuclear factors may result in loss- or gain-of-function of such factors, which affects the cell cycle and cell development. Therefore, cytoskeletal components are attractive therapeutic targets, particularly microtubules, to prevent pathological situations such as rapidly dividing tumor cells or to favor the process of cell differentiation in other cases. In this review we will address some classical and novel aspects of key regulatory functions of heat-shock proteins and immunophilins as housekeeping factors of the cytoskeletal network.
Collapse
|
49
|
PPARgamma Inhibitors as Novel Tubulin-Targeting Agents. PPAR Res 2011; 2008:785405. [PMID: 18509498 PMCID: PMC2396401 DOI: 10.1155/2008/785405] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 05/01/2008] [Indexed: 12/11/2022] Open
Abstract
The microtubule-targeting agents (MTAs) are a very successful class of cancer drugs with therapeutic benefits in both hematopoietic and solid tumors. However, resistance to these drugs is a significant problem. Current MTAs bind to microtubules, and/or to their constituent tubulin heterodimers, and affect microtubule polymerization and dynamics. The PPARγ inhibitor T0070907 can reduce tubulin levels in colorectal cancer cell lines and suppress tumor growth in a murine xenograft model. T0070907 does not alter microtubule polymerization in vitro, and does not appear to work by triggering modulation of tubulin RNA levels subsequent to decreased polymerization. This observation suggests the possible development of antimicrotubule drugs that work by a novel mechanism, and implies the presence of cancer therapeutic targets that have not yet been exploited. This review summarizes what is known about PPARγ inhibitors and cancer cell death, with emphasis on the tubulin phenotype and PPAR-dependence, and identifies potential mechanisms of action.
Collapse
|
50
|
Knee KM, Goulet DR, Zhang J, Chen B, Chiu W, King JA. The group II chaperonin Mm-Cpn binds and refolds human γD crystallin. Protein Sci 2011; 20:30-41. [PMID: 20981710 DOI: 10.1002/pro.531] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chaperonins assist in the folding of nascent and misfolded proteins, though the mechanism of folding within the lumen of the chaperonin remains poorly understood. The archeal chaperonin from Methanococcus marapaludis, Mm-Cpn, shares the eightfold double barrel structure with other group II chaperonins, including the eukaryotic TRiC/CCT, required for actin and tubulin folding. However, Mm-Cpn is composed of a single species subunit, similar to group I chaperonin GroEL, rather than the eight subunit species needed for TRiC/CCT. Features of the β-sheet fold have been identified as sites of recognition by group II chaperonins. The crystallins, the major components of the vertebrate eye lens, are β-sheet proteins with two homologous Greek key domains. During refolding in vitro a partially folded intermediate is populated, and partitions between productive folding and off-pathway aggregation. We report here that in the presence of physiological concentrations of ATP, Mm-Cpn suppressed the aggregation of HγD-Crys by binding the partially folded intermediate. The complex was sufficiently stable to permit recovery by size exclusion chromatography. In the presence of ATP, Mm-Cpn promoted the refolding of the HγD-Crys intermediates to the native state. The ability of Mm-Cpn to bind and refold a human β-sheet protein suggests that Mm-Cpn may be useful as a simplified model for the substrate recognition mechanism of TRiC/CCT.
Collapse
Affiliation(s)
- Kelly M Knee
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | |
Collapse
|