1
|
Li B, Wen M, Gao F, Wang Y, Wei G, Duan Y. Regulation of HNRNP family by post-translational modifications in cancer. Cell Death Discov 2024; 10:427. [PMID: 39366930 PMCID: PMC11452504 DOI: 10.1038/s41420-024-02198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024] Open
Abstract
Heterogeneous nuclear ribonucleoproteins (HNRNPs) represent a large family of RNA-binding proteins consisting of more than 20 members and have attracted great attention with their distinctive roles in cancer progression by regulating RNA splicing, transcription, and translation. Nevertheless, the cancer-specific modulation of HNRNPs has not been fully elucidated. The research of LC-MS/MS technology has documented that HNRNPs were widely and significantly targeted by different post-translational modifications (PTMs), which have emerged as core regulators in shaping protein functions and are involved in multiple physiological processes. Accumulating studies have highlighted that several PTMs are involved in the mechanisms of HNRNPs regulation in cancer and may be suitable therapeutic targets. In this review, we summarize the existing evidence describing how PTMs modulate HNRNPs functions on gene regulation and the involvement of their dysregulation in cancer, which will help shed insights on their clinical impacts as well as possible therapeutic tools targeting PTMs on HNRNPs.
Collapse
Affiliation(s)
- Bohao Li
- Department of Cell Biology and Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mingxin Wen
- Department of Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fei Gao
- Department of Cell Biology and Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guangwei Wei
- Department of Cell Biology and Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Yangmiao Duan
- Department of Cell Biology and Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Li ML, Ragupathi A, Patel N, Hernandez T, Magsino J, Werlen G, Brewer G, Jacinto E. The RNA-binding protein AUF1 facilitates Akt phosphorylation at the membrane. J Biol Chem 2022; 298:102437. [PMID: 36041631 PMCID: PMC9513781 DOI: 10.1016/j.jbc.2022.102437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Mammalian target of rapamycin (mTOR), which is part of mTOR complex 1 (mTORC1) and mTORC2, controls cellular metabolism in response to levels of nutrients and other growth signals. A hallmark of mTORC2 activation is the phosphorylation of Akt, which becomes upregulated in cancer. How mTORC2 modulates Akt phosphorylation remains poorly understood. Here, we found that the RNA-binding protein, AUF1 (ARE/poly(U)-binding/degradation factor 1), modulates mTORC2/Akt signaling. We determined that AUF1 is required for phosphorylation of Akt at Thr308, Thr450, and Ser473 and that AUF1 also mediates phosphorylation of the mTORC2-modulated metabolic enzyme glutamine fructose-6-phosphate amidotransferase 1 at Ser243. In addition, AUF1 immunoprecipitation followed by quantitative RT–PCR revealed that the mRNAs of Akt, glutamine fructose-6-phosphate amidotransferase 1, and the mTORC2 component SIN1 associate with AUF1. Furthermore, expression of the p40 and p45, but not the p37 or p42, isoforms of AUF1 specifically mediate Akt phosphorylation. In the absence of AUF1, subcellular fractionation indicated that Akt fails to localize to the membrane. However, ectopic expression of a membrane-targeted allele of Akt is sufficient to allow Akt-Ser473 phosphorylation despite AUF1 depletion. Finally, conditions that enhance mTORC2 signaling, such as acute glutamine withdrawal, augment AUF1 phosphorylation, whereas mTOR inhibition abolishes AUF1 phosphorylation. Our findings unravel a role for AUF1 in promoting membrane localization of Akt to facilitate its phosphorylation on this cellular compartment. Targeting AUF1 could have therapeutic benefit for cancers with upregulated mTORC2/Akt signaling.
Collapse
Affiliation(s)
- Mei-Ling Li
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Aparna Ragupathi
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Nikhil Patel
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Tatiana Hernandez
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Jedrick Magsino
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Guy Werlen
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Gary Brewer
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854.
| | - Estela Jacinto
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854.
| |
Collapse
|
3
|
Bi Y, Cui D, Xiong X, Zhao Y. The characteristics and roles of β-TrCP1/2 in carcinogenesis. FEBS J 2020; 288:3351-3374. [PMID: 33021036 DOI: 10.1111/febs.15585] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/02/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
β-transducin repeat-containing protein (β-TrCP), one of the well-characterized F-box proteins, acts as a substrate receptor and constitutes an active SCFβ-TrCP E3 ligase with a scaffold protein CUL1, a RING protein RBX1, and an adaptor protein SKP1. β-TrCP plays a critical role in the regulation of various physiological and pathological processes, including signal transduction, cell cycle progression, cell migration, DNA damage response, and tumorigenesis, by governing burgeoning amounts of key regulators for ubiquitination and proteasomal degradation. Given that a variety of β-TrCP substrates are well-known oncoproteins and tumor suppressors, and dysregulation of β-TrCP is frequently identified in human cancers, β-TrCP plays a vital role in carcinogenesis. In this review, we first briefly introduce the characteristics of β-TrCP1, β-TrCP2, and SCFβ-TrCP ubiquitin ligase, and then discuss SCFβ-TrCP ubiquitin ligase regulated biological processes by targeting its substrates for degradation. Moreover, we summarize the regulation of β-TrCP1 and β-TrCP2 at multiple layers and further discuss the various roles of β-TrCP1 and β-TrCP2 in human cancer, functioning as either an oncoprotein or a tumor suppressor in a manner dependent of cellular context. Finally, we provide novel insights for future perspectives on the potential of targeting β-TrCP1 and β-TrCP2 for cancer therapy.
Collapse
Affiliation(s)
- Yanli Bi
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Danrui Cui
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiufang Xiong
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
mRNA Post-Transcriptional Regulation by AU-Rich Element-Binding Proteins in Liver Inflammation and Cancer. Int J Mol Sci 2020; 21:ijms21186648. [PMID: 32932781 PMCID: PMC7554771 DOI: 10.3390/ijms21186648] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
AU-rich element-binding proteins (AUBPs) represent important post-transcriptional regulators of gene expression. AUBPs can bind to the AU-rich elements present in the 3'-UTR of more than 8% of all mRNAs and are thereby able to control the stability and/or translation of numerous target mRNAs. The regulation of the stability and the translation of mRNA transcripts by AUBPs are highly complex processes that occur through multiple mechanisms depending on the cell type and the cellular context. While AUBPs have been shown to be involved in inflammatory processes and the development of various cancers, their important role and function in the development of chronic metabolic and inflammatory fatty liver diseases (FLDs), as well as in the progression of these disorders toward cancers such as hepatocellular carcinoma (HCC), has recently started to emerge. Alterations of either the expression or activity of AUBPs are indeed significantly associated with FLDs and HCC, and accumulating evidence indicates that several AUBPs are deeply involved in a significant number of cellular processes governing hepatic metabolic disorders, inflammation, fibrosis, and carcinogenesis. Herein, we discuss our current knowledge of the roles and functions of AUBPs in liver diseases and cancer. The relevance of AUBPs as potential biomarkers for different stages of FLD and HCC, or as therapeutic targets for these diseases, are also highlighted.
Collapse
|
5
|
Anerillas C, Abdelmohsen K, Gorospe M. Regulation of senescence traits by MAPKs. GeroScience 2020; 42:397-408. [PMID: 32300964 PMCID: PMC7205942 DOI: 10.1007/s11357-020-00183-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023] Open
Abstract
A phenotype of indefinite growth arrest acquired in response to sublethal damage, cellular senescence affects normal aging and age-related disease. Mitogen-activated protein kinases (MAPKs) are capable of sensing changes in cellular conditions, and in turn elicit adaptive responses including cell senescence. MAPKs modulate the levels and function of many proteins, including proinflammatory factors and factors in the p21/p53 and p16/RB pathways, the main senescence-regulatory axes. Through these actions, MAPKs implement key traits of senescence-growth arrest, cell survival, and the senescence-associated secretory phenotype (SASP). In this review, we summarize and discuss our current knowledge of the impact of MAPKs in senescence. In addition, given that eliminating or suppressing senescent cells can improve health span, we discuss the function and possible exploitation of MAPKs in the elimination (senolysis) or suppression (senostasis) of senescent cells.
Collapse
Affiliation(s)
- Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
6
|
Liao Y, Liu Y, Xia X, Shao Z, Huang C, He J, Jiang L, Tang D, Liu J, Huang H. Targeting GRP78-dependent AR-V7 protein degradation overcomes castration-resistance in prostate cancer therapy. Am J Cancer Res 2020; 10:3366-3381. [PMID: 32206096 PMCID: PMC7069092 DOI: 10.7150/thno.41849] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/26/2020] [Indexed: 01/09/2023] Open
Abstract
Rationale: Androgen receptor splice variant 7 (AR-V7) is a leading cause of the development of castration-resistant prostate cancer (CRPC). However, the regulation and function of AR-V7 at levels of post-translational modifications in prostate cancer therapy remain poorly understood. Here, we conducted a library screen of natural products to identify potential small molecules responsible for AR-V7 protein degradation in human prostate cancer cell lines. Methods: A natural product library was used to screen the inhibitor of AR-V7. Co-IP and biomass spectrum assays were used to identify the AR-V7-interacting proteins, whereas western blot, confocal microscopy, RNA interfering, and gene transfection were used to validate these interactions. Cell viability, EDU staining, and colony formation assays were employed to detect cell growth and proliferation. Flowcytometry assays were used to detect the distribution of cell cycle. Mouse xenograft models were used to study the anti-CRPC effects in vivo. Results: This screen identified rutaecarpine, one of the major components of the Chinese medicine Evodia rutaecarpa, as a novel chemical that selectively induces AR-V7 protein degradation via K48-linked ubiquitination. Mechanically, this effect relies on rutaecarpine inducing the formation of a GRP78-AR-V7 protein complex, which further recruits the E3 ligase SIAH2 to directly promote the ubiquitination of AR-V7. Consequently, the genetic and pharmacological activation of the GRP78-dependent AR-V7 protein degradation restores the sensitivity of castration-resistant prostate cancer to anti-androgen therapy in cell culture and animal models. Conclusions: These findings not only provide a new approach for overcoming castration-resistance in prostate cancer therapy, but also increase our understanding about the interplay between molecular chaperones and ubiquitin ligase in shaping protein stability.
Collapse
|
7
|
Ehlting C, Rex J, Albrecht U, Deenen R, Tiedje C, Köhrer K, Sawodny O, Gaestel M, Häussinger D, Bode JG. Cooperative and distinct functions of MK2 and MK3 in the regulation of the macrophage transcriptional response to lipopolysaccharide. Sci Rep 2019; 9:11021. [PMID: 31363109 PMCID: PMC6667695 DOI: 10.1038/s41598-019-46791-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/29/2019] [Indexed: 12/04/2022] Open
Abstract
The p38MAPK downstream targets MAPKAP kinases (MK) 2 and 3 are critical for the regulation of the macrophage response to LPS. The extents to which these two kinases act cooperatively and distinctly in regulating LPS-induced inflammatory cytokine expression are still unclear. To address this uncertainty, whole transcriptome analyses were performed using bone marrow-derived macrophages (BMDM) generated from MK2−/− or MK2/3−/− animals and their wild-type littermates. The results suggest that in BMDM, MK2 and MK3 not only cooperatively regulate the transcript expression of signaling intermediates, including IL-10, IL-19, CXCL2 and the IL-4 receptor (IL-4R)α subunit, they also exert distinct regulatory effects on the expression of specific transcripts. Based on the differential regulation of gene expression by MK2 and MK3, at least six regulatory patterns were identified. Importantly, we confirmed our previous finding, which showed that in the absence of MK2, MK3 negatively regulates IFN-β. Moreover, this genome-wide analysis identified the regulation of Cr1A, NOD1 and Serpina3f as similar to that of IFN-β. In the absence of MK2, MK3 also delayed the nuclear translocation of NFκB by delaying the ubiquitination and subsequent degradation of IκBβ, reflecting the substantial plasticity of the response of BMDM to LPS.
Collapse
Affiliation(s)
- Christian Ehlting
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Julia Rex
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - René Deenen
- Biological and Medical Research Center (BMFZ), Genomics & Transcriptomics Laboratory, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Christopher Tiedje
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany.,Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Genomics & Transcriptomics Laboratory, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Oliver Sawodny
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
8
|
Soni S, Anand P, Padwad YS. MAPKAPK2: the master regulator of RNA-binding proteins modulates transcript stability and tumor progression. J Exp Clin Cancer Res 2019; 38:121. [PMID: 30850014 PMCID: PMC6408796 DOI: 10.1186/s13046-019-1115-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/21/2019] [Indexed: 01/09/2023] Open
Abstract
The p38 mitogen-activated protein kinase (p38MAPK) pathway has been implicated in a variety of pathological conditions including inflammation and metastasis. Post-transcriptional regulation of genes harboring adenine/uridine-rich elements (AREs) in their 3'-untranslated region (3'-UTR) is controlled by MAPK-activated protein kinase 2 (MAPKAPK2 or MK2), a downstream substrate of the p38MAPK. In response to diverse extracellular stimuli, MK2 influences crucial signaling events, regulates inflammatory cytokines, transcript stability and critical cellular processes. Expression of genes involved in these vital cellular cascades is controlled by subtle interactions in underlying molecular networks and post-transcriptional gene regulation that determines transcript fate in association with RNA-binding proteins (RBPs). Several RBPs associate with the 3'-UTRs of the target transcripts and regulate their expression via modulation of transcript stability. Although MK2 regulates important cellular phenomenon, yet its biological significance in tumor progression has not been well elucidated till date. In this review, we have highlighted in detail the importance of MK2 as the master regulator of RBPs and its role in the regulation of transcript stability, tumor progression, as well as the possibility of use of MK2 as a therapeutic target in tumor management.
Collapse
Affiliation(s)
- Sourabh Soni
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh India
- Academy of Scientific and Innovative Research, Chennai, Tamil Nadu India
| | - Prince Anand
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh India
- Academy of Scientific and Innovative Research, Chennai, Tamil Nadu India
| | - Yogendra S. Padwad
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh India
- Academy of Scientific and Innovative Research, Chennai, Tamil Nadu India
| |
Collapse
|
9
|
Dependence of HSP27 cellular level on protein kinase CK2 discloses novel therapeutic strategies. Biochim Biophys Acta Gen Subj 2018; 1862:2902-2910. [PMID: 30279146 DOI: 10.1016/j.bbagen.2018.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/21/2018] [Accepted: 09/18/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND HSP27 plays a role in various diseases, including neurodegenerative diseases, ischemia, and atherosclerosis. It is particularly important in the regulation of the development, progression and metastasis of cancer as well as cell apoptosis and drug resistance. However, the absence of an ATP binding domain, that is, instead, present in other HSPs such as HSP90 and HSP70, hampers the development of small molecules as inhibitors of HSP27. METHODS Knockout cell lines generated by Crispr/Cas9 gene editing tool, specific kinase inhibitors and siRNA transfections were exploited to demonstrate that the expression of HSP27 is dependent on the integrity/activity of protein kinase CK2 holoenzyme. The interaction between these proteins has been confirmed by co-immunoprecipitation, confocal immunofluorescence microscopy, and by density gradient separation of protein complexes. Finally, using a proliferation assay this study demonstrates the potential efficacy of a combinatory therapy of heath shock and CK2 inhibitors in cancer treatment. RESULTS Our data demonstrate that CK2 is able to regulate HSP27 turnover by affecting the expression of its ubiquitin ligase SMURF2 (Smad ubiquitination regulatory factor 2). Moreover, for the first time we show an increased sensitivity of CK2-inhibited tumour cells to hyperthermia treatment. CONCLUSION Being HSP27 involved in several pathological conditions, including protein conformational diseases (i.e Cystic Fibrosis) and cancer, the need of drugs to modulate its activity is growing and CK2-targeting could represent a new strategy to reduce cellular HSP27 level. GENERAL SIGNIFICANCE This study identifies CK2 as a molecular target to control HSP27 cellular expression.
Collapse
|
10
|
Breed ER, Hilliard CA, Yoseph B, Mittal R, Liang Z, Chen CW, Burd EM, Brewster LP, Hansen LM, Gleason RL, Pandita TK, Ford ML, Hunt CR, Coopersmith CM. The small heat shock protein HSPB1 protects mice from sepsis. Sci Rep 2018; 8:12493. [PMID: 30131526 PMCID: PMC6104051 DOI: 10.1038/s41598-018-30752-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/01/2018] [Indexed: 12/29/2022] Open
Abstract
In vitro studies have implicated the small heat shock protein HSPB1 in a range of physiological functions. However, its in vivo relevance is unclear as the phenotype of unstressed HSPB1−/− mice is unremarkable. To determine the impact of HSPB1 in injury, HSPB1−/− and wild type (WT) mice were subjected to cecal ligation and puncture, a model of polymicrobial sepsis. Ten-day mortality was significantly higher in HSPB1−/− mice following the onset of sepsis (65% vs. 35%). Ex vivo mechanical testing revealed that common carotid arteries from HSPB1−/− mice were more compliant than those in WT mice over pressures of 50–120 mm Hg. Septic HSPB1−/− mice also had increased peritoneal levels of IFN-γ and decreased systemic levels of IL-6 and KC. There were no differences in frequency of either splenic CD4+ or CD8+ T cells, nor were there differences in apoptosis in either cell type. However, splenic CD4+ T cells and CD8+ T cells from HSPB1−/− mice produced significantly less TNF and IL-2 following ex vivo stimulation. Systemic and local bacterial burden was similar in HSPB1−/− and WT mice. Thus while HSPB1−/− mice are uncompromised under basal conditions, HSPB1 has a critical function in vivo in sepsis, potentially mediated through alterations in arterial compliance and the immune response.
Collapse
Affiliation(s)
- Elise R Breed
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Carolyn A Hilliard
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Benyam Yoseph
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Rohit Mittal
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Ching-Wen Chen
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Luke P Brewster
- Department of Surgery, Division of Vascular Surgery, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Laura M Hansen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Rudolph L Gleason
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Tej K Pandita
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Clayton R Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, Georgia.
| |
Collapse
|
11
|
Forouzanfar F, Butler AE, Banach M, Barreto GE, Sahbekar A. Modulation of heat shock proteins by statins. Pharmacol Res 2018; 134:134-144. [DOI: 10.1016/j.phrs.2018.06.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
|
12
|
Involvement of p38-βTrCP-Tristetraprolin-TNFα axis in radiation pneumonitis. Oncotarget 2018; 8:47767-47779. [PMID: 28548957 PMCID: PMC5564603 DOI: 10.18632/oncotarget.17770] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/14/2017] [Indexed: 11/25/2022] Open
Abstract
Early release of tumor necrosis factor-alpha (TNF-α) during radiotherapy of thoracic cancers plays an important role in radiation pneumonitis, whose inhibition may provide lung radioprotection. We previously reported radiation inactivates Tristetraprolin (TTP), a negative regulator of TNF-α synthesis, which correlated with increased TNF-α release. However, the molecular events involved in radiation-induced TTP inactivation remain unclear. To determine if eliminating Ttp in mice resulted in a phenotypic response to radiation, Ttp-null mice lungs were exposed to a single dose of 15 Gy, and TNF-α release and lung inflammation were analyzed at different time points post-irradiation. Ttp−/− mice with elevated (9.5±0.6 fold) basal TNF-α showed further increase (12.2±0.9 fold, p<0.02) in TNF-α release and acute lung inflammation within a week post-irradiation. Further studies using mouse lung macrophage (MH-S), human lung fibroblast (MRC-5), and exogenous human TTP overexpressing U2OS and HEK293 cells upon irradiation (a single dose of 4 Gy) promoted p38-mediated TTP phosphorylation at the serine 186 position, which primed it to be recognized by an ubiquitin ligase (E3), beta transducing repeat containing protein (β-TrCP), to promote polyubiquitination-mediated proteasomal degradation. Consequently, a serine 186 to alanine (SA) mutant of TTP was resistant to radiation-induced degradation. Similarly, either a p38 kinase inhibitor (SB203580), or siRNA-mediated β-TrCP knockdown, or overexpression of dominant negative Cullin1 mutants protected TTP from radiation-induced degradation. Consequently, SB203580 pretreatment blocked radiation-induced TNF-α release and radioprotected macrophages. Together, these data establish the involvement of the p38-βTrCP-TTP-TNFα signaling axis in radiation-induced lung inflammation and identified p38 inhibition as a possible lung radioprotection strategy.
Collapse
|
13
|
Corsini E, Galbiati V, Papale A, Kummer E, Pinto A, Guaita A, Racchi M. The role of HSP27 in RACK1-mediated PKC activation in THP-1 cells. Immunol Res 2017; 64:940-50. [PMID: 27178349 DOI: 10.1007/s12026-016-8802-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Receptor for Activated C Kinase 1 (RACK1) pseudosubstrate is a commercially available peptide that directly activates protein kinase C-β (PKCβ). We have recently shown that RACK1 pseudosubstrate, alone or in combination with classical immune activators, results in increased cytokine production and CD86 upregulation in primary leukocytes. Furthermore, we demonstrated a role of PKCβ and RACK1 in chemical allergen-induced CD86 expression and IL-8 production in both THP-1 cells and primary human dendritic cells. Aim of this study was to shed light on the mechanisms underlying RACK1 pseudosubstrate-induced immune activation and to compare it to lipopolysaccharide (LPS). The human promyelocytic cell line THP-1 was used throughout the study. RACK1 pseudosubstrate induced rapid (5 min) and dose-related PKCβ activation as assessed by its membrane translocation. Among the proteins phosphorylated, we identified Hsp27. Both RACK1 pseudosubstrate and LPS induce its phosphorylation and release in culture medium. The release of Hsp27 induced by RACK1 pseudosubstrate was also confirmed in peripheral blood mononuclear cells. To evaluate the role of Hsp27 in RACK1 pseudosubstrate or LPS-induced cell activation, we conducted Hsp27 silencing and neutralization experiments. Both strategies confirmed the central role of Hsp27 in RACK1 pseudosubstrate or LPS-induced cell activation, as assessed by IL-8 production and upregulation of CD86.
Collapse
Affiliation(s)
- Emanuela Corsini
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Valentina Galbiati
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Angela Papale
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Elena Kummer
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Antonella Pinto
- Department of Drug Sciences - Pharmacology, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy
| | | | - Marco Racchi
- Department of Drug Sciences - Pharmacology, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy.
| |
Collapse
|
14
|
Kennedy D, Mnich K, Oommen D, Chakravarthy R, Almeida-Souza L, Krols M, Saveljeva S, Doyle K, Gupta S, Timmerman V, Janssens S, Gorman AM, Samali A. HSPB1 facilitates ERK-mediated phosphorylation and degradation of BIM to attenuate endoplasmic reticulum stress-induced apoptosis. Cell Death Dis 2017; 8:e3026. [PMID: 29048431 PMCID: PMC5596589 DOI: 10.1038/cddis.2017.408] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/30/2017] [Accepted: 07/06/2017] [Indexed: 01/11/2023]
Abstract
BIM, a pro-apoptotic BH3-only protein, is a key regulator of the intrinsic (or mitochondrial) apoptosis pathway. Here, we show that BIM induction by endoplasmic reticulum (ER) stress is suppressed in rat PC12 cells overexpressing heat shock protein B1 (HSPB1 or HSP27) and that this is due to enhanced proteasomal degradation of BIM. HSPB1 and BIM form a complex that immunoprecipitates with p-ERK1/2. We found that HSPB1-mediated proteasomal degradation of BIM is dependent on MEK-ERK signaling. Other studies have shown that several missense mutations in HSPB1 cause the peripheral neuropathy, Charcot-Marie-Tooth (CMT) disease, which is associated with nerve degeneration. Here we show that cells overexpressing CMT-related HSPB1 mutants exhibited increased susceptibility to ER stress-induced cell death and high levels of BIM. These findings identify a novel function for HSPB1 as a negative regulator of BIM protein stability leading to protection against ER stress-induced apoptosis, a function that is absent in CMT-associated HSPB1 mutants.
Collapse
Affiliation(s)
- Donna Kennedy
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Katarzyna Mnich
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Deepu Oommen
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Reka Chakravarthy
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Leonardo Almeida-Souza
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Michiel Krols
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Svetlana Saveljeva
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Karen Doyle
- Discipline of Physiology, NUI Galway, Galway, Ireland
| | - Sanjeev Gupta
- Discipline of Pathology, School of Medicine, NUI Galway, Galway, Ireland
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Sophie Janssens
- Unit Immunoregulation and Mucosal Immunology, VIB Inflammation Research Centre, Ghent University, Gent, Belgium.,Department of Internal Medicine, Ghent University, Gent, Belgium
| | - Adrienne M Gorman
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| |
Collapse
|
15
|
Kaigorodova EV, Zavyalova MV, Bychkov VA, Perelmuter VM, Choynzonov EL. Functional state of the Hsp27 chaperone as a molecular marker of an unfavorable course of larynx cancer. Cancer Biomark 2017; 17:145-53. [PMID: 27540972 DOI: 10.3233/cbm-160625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The small heat shock protein 27 kDA (Hsp27) acts as an ATP-independent chaperone in protein folding, but is also implicated in architecture of the cytoskeleton, cell migration, metabolism, cell survival, growth/differentiation, mRNA stabilization, and tumor progression. OBJECTIVE To study the intracellular localization of phosphorylated and non-phosphorylated forms of Hsp27 in squamous cell carcinoma of the larynx (SCCL) and to evaluate their relationship with regional lymphatic metastasis and overall five-year survival. METHODS Tumor biopsies of larynx tissue were collected from 50 patients who were between the ages of 30 to 80 years and had a confirmed diagnosis of squamous cell carcinoma of the larynx. Immunohistochemistry was used to determine the intracellular localization of the phosphorylated and non-phosphorylated forms of Hsp27. RESULTS The study revealed that the Hsp27 chaperone was expressed in both the cytoplasm and the nucleus of tumor cells in SCCL. The biopsies of patients with lymph node metastases showed significantly higher expression of the phosphorylated and unphosphorylated forms of Hsp27 in the nucleus compared to those of patients without lymph node metastases. At the same time, the cytoplasmic expression of Hsp27 in these patients did not differ statistically. Analysis of the overall five-year survival rates showed that negative Hsp27 expression in the nucleus of tumor cells is associated with the survival rate of patients with SCCL. CONCLUSION The nuclear expression of phosphorylated and unphosphorylated forms of Hsp27 is a molecular marker of unfavorable squamous cell carcinoma of the larynx associated with lymphogenous metastasis and decreased total five-year survival.
Collapse
Affiliation(s)
- Evgeniya V Kaigorodova
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | - Marina V Zavyalova
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | | | - Vladimir M Perelmuter
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | - Evgenii L Choynzonov
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| |
Collapse
|
16
|
White EJF, Matsangos AE, Wilson GM. AUF1 regulation of coding and noncoding RNA. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27620010 DOI: 10.1002/wrna.1393] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 01/12/2023]
Abstract
AUF1 is a family of four RNA-binding proteins (RBPs) generated by alternative pre-messenger RNA (pre-mRNA) splicing, with canonical roles in controlling the stability and/or translation of mRNA targets based on recognition of AU-rich sequences within mRNA 3' untranslated regions. However, recent studies identifying AUF1 target sites across the transcriptome have revealed that these canonical functions are but a subset of its roles in posttranscriptional regulation of gene expression. In this review, we describe recent developments in our understanding of the RNA-binding properties of AUF1 together with their biochemical implications and roles in directing mRNA decay and translation. This is then followed by a survey of newly discovered activities for AUF1 proteins in control of miRNA synthesis and function, including miRNA assembly into microRNA (miRNA)-loaded RNA-induced silencing complexes (miRISCs), miRISC targeting to mRNA substrates, interplay with an expanding network of other cellular RBPs, and reciprocal regulatory relationships between miRNA and AUF1 synthesis. Finally, we discuss recently reported relationships between AUF1 and long noncoding RNAs and regulatory roles on viral RNA substrates. Cumulatively, these findings have significantly expanded our appreciation of the scope and diversity of AUF1 functions in the cell, and are prompting an exciting array of new questions moving forward. WIREs RNA 2017, 8:e1393. doi: 10.1002/wrna.1393 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Elizabeth J F White
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aerielle E Matsangos
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerald M Wilson
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Li S, Zhang W, Fan J, Lai Y, Che G. Clinicopathological and prognostic significance of heat shock protein 27 (HSP27) expression in non-small cell lung cancer: a systematic review and meta-analysis. SPRINGERPLUS 2016; 5:1165. [PMID: 27512624 PMCID: PMC4960090 DOI: 10.1186/s40064-016-2827-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 07/14/2016] [Indexed: 02/05/2023]
Abstract
Numbers of clinical and experimental investigations have provided increasing evidences to demonstrate that heat shock protein 27 (HSP27) is a qualified predictor for many cancers. However, no consensus has been reached on its clinicopathological and prognostic significance in patients with non-small cell lung cancer (NSCLC). Therefore, we performed this systematic meta-analysis to help addressing this issue. PubMed, EMBASE, the Web of Science and China National Knowledge Infrastructure were searched for full-text literatures met out eligibility criteria. We determined the odds ratio (OR) and hazard ratio (HR) as the appropriate summarized statistics for assessments of clinicopathological and prognostic roles of HSP27, respectively. Q-test and I(2)-statistic were used to evaluate the level of heterogeneity. Sensitivity analysis was conducted to examine the stability of overall estimates. Potential publication bias was detected by Begg's test and Egger's test. Finally, ten articles were identified to be included into our meta-analysis. The pooled analyses suggested that HSP27 expression was significantly associated with the unfavorable conditions for differentiation degree, lymphatic metastasis, clinical stage, squamous cell carcinoma and tumor size. However, HSP27 expression had no significant relationship to gender, age and smoking status. Meanwhile, pooled HRs indicated that HSP27 expression could be a predictor for a lower 5-year overall survival (OS) rate (HR: 1.832; 95 % CI 1.322-2.538; P < 0.001) but not for 1-year OS of NSCLC (HR: 0.885; 95 % CI 0.140-5.599; P = 0.896). In conclusion, our meta-analysis demonstrates that HSP27 expression may be a strong biomarker to predict both the poor clinicopathological and prognostic characteristics in patients with NSCLC.
Collapse
Affiliation(s)
- Shuangjiang Li
- Department of General Thoracic Surgery, West China Hospital, Sichuan University, Guoxue Alley No. 37, Chengdu, 610041 China
| | - Wenbiao Zhang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Jun Fan
- Department of General Thoracic Surgery, West China Hospital, Sichuan University, Guoxue Alley No. 37, Chengdu, 610041 China
| | - Yutian Lai
- Department of General Thoracic Surgery, West China Hospital, Sichuan University, Guoxue Alley No. 37, Chengdu, 610041 China
| | - Guowei Che
- Department of General Thoracic Surgery, West China Hospital, Sichuan University, Guoxue Alley No. 37, Chengdu, 610041 China
| |
Collapse
|
18
|
Noren Hooten N, Martin‐Montalvo A, Dluzen DF, Zhang Y, Bernier M, Zonderman AB, Becker KG, Gorospe M, Cabo R, Evans MK. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell 2016; 15:572-81. [PMID: 26990999 PMCID: PMC4854919 DOI: 10.1111/acel.12469] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2016] [Indexed: 12/20/2022] Open
Abstract
Metformin, an oral hypoglycemic agent, has been used for decades to treat type 2 diabetes mellitus. Recent studies indicate that mice treated with metformin live longer and have fewer manifestations of age‐related chronic disease. However, the molecular mechanisms underlying this phenotype are unknown. Here, we show that metformin treatment increases the levels of the microRNA‐processing protein DICER1 in mice and in humans with diabetes mellitus. Our results indicate that metformin upregulates DICER1 through a post‐transcriptional mechanism involving the RNA‐binding protein AUF1. Treatment with metformin altered the subcellular localization of AUF1, disrupting its interaction with DICER1 mRNA and rendering DICER1 mRNA stable, allowing DICER1 to accumulate. Consistent with the role of DICER1 in the biogenesis of microRNAs, we found differential patterns of microRNA expression in mice treated with metformin or caloric restriction, two proven life‐extending interventions. Interestingly, several microRNAs previously associated with senescence and aging, including miR‐20a, miR‐34a, miR‐130a, miR‐106b, miR‐125, and let‐7c, were found elevated. In agreement with these findings, treatment with metformin decreased cellular senescence in several senescence models in a DICER1‐dependent manner. Metformin lowered p16 and p21 protein levels and the abundance of inflammatory cytokines and oncogenes that are hallmarks of the senescence‐associated secretory phenotype (SASP). These data lead us to hypothesize that changes in DICER1 levels may be important for organismal aging and to propose that interventions that upregulate DICER1 expression (e.g., metformin) may offer new pharmacotherapeutic approaches for age‐related disease.
Collapse
Affiliation(s)
- Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Alejandro Martin‐Montalvo
- Translational Gerontology Branch National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
- Pancreatic Islet Development and Regeneration Unit Department of Stem Cells CABIMER‐Andalusian Center for Molecular Biology and Regenerative Medicine Avenida Americo Vespucio, Parque Científico y Tecnologico Cartuja 93 41092 Sevilla Spain
| | - Douglas F. Dluzen
- Laboratory of Epidemiology and Population Sciences National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Yongqing Zhang
- Laboratory of Genetics National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Michel Bernier
- Translational Gerontology Branch National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Kevin G. Becker
- Laboratory of Genetics National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Myriam Gorospe
- Laboratory of Genetics National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Rafael Cabo
- Translational Gerontology Branch National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences National Institute on Aging National Institutes of Health 251 Bayview Boulevard Baltimore MD 21224 USA
| |
Collapse
|
19
|
He X, Li L, Tan H, Chen J, Zhou Y. Atorvastatin attenuates contrast-induced nephropathy by modulating inflammatory responses through the regulation of JNK/p38/Hsp27 expression. J Pharmacol Sci 2016; 131:18-27. [PMID: 27156929 DOI: 10.1016/j.jphs.2016.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/19/2016] [Accepted: 03/10/2016] [Indexed: 01/08/2023] Open
Abstract
This study aimed to investigate whether atorvastatin reduce the contrast-induced nephropathy inflammatory response and apoptosis of renal tubular epithelial cells and the relationship with MAPK signaling pathway. We utilized the iopamidol-induced contrast-induced nephropathy (CIN) rat model which was induced by a single dose of iopamidol (2.9 g iodine/kg) and a cell model in which human embryonic proximal tubular (HK2) cells were treated with iopamidol. The rats were divided into five groups: (1) control rats (CR); (2) atorvastatin (CA); (3) iopamidol (CM); (4) iopamidol and atorvastatin (20 mg/kg d) (CMA2); (5) iopamidol and atorvastatin (40 mg/kg d) (CMA4). On days 1, 2 and 6 after iopamidol injection, the urea nitrogen and cystatin C increased in CM compared with CR but decreased in CMA compared with CM. Inflammatory parameters and the percentage of apoptotic cells were increased in CM compared with CR and CA, but they were decreased in CMA compared with CM. We also found that atorvastatin ameliorated the renal tubular necrosis, apoptosis, and the deterioration of renal function in a dose dependent manner (P < 0.05). Furthermore, in vivo, both of SP600125 (JNK inhibitor) and SB203580 (p38 inhibitor) could decrease the expression of Bax and caspase-3, but increase Bcl-2 levels in HK2 cells treated with iopamidol. Our study demonstrates that high-dosage atorvastatin treatment attenuates both the inflammatory processes and apoptosis in contrast-induced acute kidney injury, and that the JNK/p38 MAPK pathway participates in the contrast-induced apoptosis of renal tubular cells. Finally, atorvastatin reduces CIN by suppression of apoptosis, which may be through inhibition of JNK/p38 MAPK pathways.
Collapse
Affiliation(s)
- Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Liwen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hong Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Yingling Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Nguyen Ho-Bouldoires TH, Clapéron A, Mergey M, Wendum D, Desbois-Mouthon C, Tahraoui S, Fartoux L, Chettouh H, Merabtene F, Scatton O, Gaestel M, Praz F, Housset C, Fouassier L. Mitogen-activated protein kinase-activated protein kinase 2 mediates resistance to hydrogen peroxide-induced oxidative stress in human hepatobiliary cancer cells. Free Radic Biol Med 2015; 89:34-46. [PMID: 26169728 DOI: 10.1016/j.freeradbiomed.2015.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/25/2015] [Accepted: 07/08/2015] [Indexed: 12/21/2022]
Abstract
The development and progression of liver cancer are characterized by increased levels of reactive oxygen species (ROS). ROS-induced oxidative stress impairs cell proliferation and ultimately leads to cell death. Although liver cancer cells are especially resistant to oxidative stress, mechanisms of such resistance remain understudied. We identified the MAPK-activated protein kinase 2 (MK2)/heat shock protein 27 (Hsp27) signaling pathway mediating defenses against oxidative stress. In addition to MK2 and Hsp27 overexpression in primary liver tumors compared to adjacent nontumorous tissues, the MK2/Hsp27 pathway is activated by hydrogen peroxide-induced oxidative stress in hepatobiliary cancer cells. MK2 inactivation or inhibition of MK2 or Hsp27 expression increases caspase-3 and PARP cleavage and DNA breaks and therefore cell death. Interestingly, MK2/Hsp27 inhibition decreases antioxidant defenses such as heme oxygenase 1 through downregulation of the transcription factor nuclear factor erythroid-derived 2-like 2. Moreover, MK2/Hsp27 inhibition decreases both phosphorylation of epidermal growth factor receptor (EGFR) and expression of its ligand, heparin-binding EGF-like growth factor. A new identified partner of MK2, the scaffold PDZ protein EBP50, could facilitate these effects through MK2/Hsp27 pathway regulation. These findings demonstrate that the MK2/Hsp27 pathway actively participates in resistance to oxidative stress and may contribute to liver cancer progression.
Collapse
Affiliation(s)
- Thanh Huong Nguyen Ho-Bouldoires
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Audrey Clapéron
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Martine Mergey
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Dominique Wendum
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Anatomie et Cytologie Pathologiques, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Christèle Desbois-Mouthon
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Sylvana Tahraoui
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Laetitia Fartoux
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Hamza Chettouh
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Fatiha Merabtene
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Olivier Scatton
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service de Chirurgie Hépato-Biliaire et Transplantation Hépatique, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Matthias Gaestel
- Institute of Physiological Chemistry, Hannover Medical School, D-30625 Hannover, Germany
| | - Françoise Praz
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Chantal Housset
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Laura Fouassier
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France.
| |
Collapse
|
21
|
Tiedje C, Holtmann H, Gaestel M. The role of mammalian MAPK signaling in regulation of cytokine mRNA stability and translation. J Interferon Cytokine Res 2015; 34:220-32. [PMID: 24697200 DOI: 10.1089/jir.2013.0146] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Extracellular-regulated kinases and p38 mitogen-activated protein kinases are activated in innate (and adaptive) immunity and signal via different routes to alter the stability and translation of various cytokine mRNAs, enabling immune cells to respond promptly. This regulation involves mRNA elements, such as AU-rich motifs, and mRNA-binding proteins, such as tristetraprolin (TTP), HuR, and hnRNPK-homology (KH) type splicing regulatory protein (KSRP). Signal-dependent phosphorylation of mRNA-binding proteins often alters their subcellular localization or RNA-binding affinity. Furthermore, it could lead to an altered interaction with other mRNA-binding proteins and altered scaffolding properties for mRNA-modifying enzymes, such as deadenylases, polyadenylases, decapping enzymes, poly(A) binding proteins, exo- or endonucleases, and proteins of the exosome machinery. In many cases, this results in unstable mRNAs being stabilized, with their translational arrest being released and cytokine production being stimulated. Hence, components of these mechanisms are potential targets for the modulation of the inflammatory response.
Collapse
Affiliation(s)
- Christopher Tiedje
- Institute of Physiological Chemistry, Hannover Medical School , Hannover, Germany
| | | | | |
Collapse
|
22
|
Shen ZJ, Malter JS. Regulation of AU-Rich Element RNA Binding Proteins by Phosphorylation and the Prolyl Isomerase Pin1. Biomolecules 2015; 5:412-34. [PMID: 25874604 PMCID: PMC4496679 DOI: 10.3390/biom5020412] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 03/23/2015] [Accepted: 03/31/2015] [Indexed: 01/19/2023] Open
Abstract
The accumulation of 3' untranslated region (3'-UTR), AU-rich element (ARE) containing mRNAs, are predominantly controlled at the post-transcriptional level. Regulation appears to rely on a variable and dynamic interaction between mRNA target and ARE-specific binding proteins (AUBPs). The AUBP-ARE mRNA recognition is directed by multiple intracellular signals that are predominantly targeted at the AUBPs. These include (but are unlikely limited to) methylation, acetylation, phosphorylation, ubiquitination and isomerization. These regulatory events ultimately affect ARE mRNA location, abundance, translation and stability. In this review, we describe recent advances in our understanding of phosphorylation and its impact on conformation of the AUBPs, interaction with ARE mRNAs and highlight the role of Pin1 mediated prolyl cis-trans isomerization in these biological process.
Collapse
Affiliation(s)
- Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8548, USA.
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8548, USA.
| |
Collapse
|
23
|
Low TY, Peng M, Magliozzi R, Mohammed S, Guardavaccaro D, Heck AJR. A systems-wide screen identifies substrates of the SCFβTrCP ubiquitin ligase. Sci Signal 2014; 7:rs8. [PMID: 25515538 DOI: 10.1126/scisignal.2005882] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cellular proteins are degraded by the ubiquitin-proteasome system (UPS) in a precise and timely fashion. Such precision is conferred by the high substrate specificity of ubiquitin ligases. Identification of substrates of ubiquitin ligases is crucial not only to unravel the molecular mechanisms by which the UPS controls protein degradation but also for drug discovery purposes because many established UPS substrates are implicated in disease. We developed a combined bioinformatics and affinity purification-mass spectrometry (AP-MS) workflow for the system-wide identification of substrates of SCF(βTrCP), a member of the SCF family of ubiquitin ligases. These ubiquitin ligases are characterized by a multisubunit architecture typically consisting of the invariable subunits Rbx1, Cul1, and Skp1 and one of 69 F-box proteins. The F-box protein of this member of the family is βTrCP. SCF(βTrCP) binds, through the WD40 repeats of βTrCP, to the DpSGXX(X)pS diphosphorylated motif in its substrates. We recovered 27 previously reported SCF(βTrCP) substrates, of which 22 were verified by two independent statistical protocols, thereby confirming the reliability of this approach. In addition to known substrates, we identified 221 proteins that contained the DpSGXX(X)pS motif and also interacted specifically with the WD40 repeats of βTrCP. Thus, with SCF(βTrCP), as the example, we showed that integration of structural information, AP-MS, and degron motif mining constitutes an effective method to screen for substrates of ubiquitin ligases.
Collapse
Affiliation(s)
- Teck Yew Low
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands. Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Mao Peng
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands. Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Roberto Magliozzi
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Shabaz Mohammed
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands. Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Daniele Guardavaccaro
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands. Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, Netherlands.
| |
Collapse
|
24
|
Rapid proteasomal degradation of posttranscriptional regulators of the TIS11/tristetraprolin family is induced by an intrinsically unstructured region independently of ubiquitination. Mol Cell Biol 2014; 34:4315-28. [PMID: 25246635 DOI: 10.1128/mcb.00643-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The TIS11/tristetraprolin (TTP) CCCH tandem zinc finger proteins are major effectors in the destabilization of mRNAs bearing AU-rich elements (ARE) in their 3' untranslated regions. In this report, we demonstrate that the Drosophila melanogaster dTIS11 protein is short-lived due to its rapid ubiquitin-independent degradation by the proteasome. Our data indicate that this mechanism is tightly associated with the intrinsically unstructured, disordered N- and C-terminal domains of the protein. Furthermore, we show that TTP, the mammalian TIS11/TTP protein prototype, shares the same three-dimensional characteristics and is degraded by the same proteolytic pathway as dTIS11, thereby indicating that this mechanism has been conserved across evolution. Finally, we observed a phosphorylation-dependent inhibition of dTIS11 and TTP degradation by the proteasome in vitro, raising the possibility that such modifications directly affect proteasomal recognition for these proteins. As a group, RNA-binding proteins (RNA-BPs) have been described as enriched in intrinsically disordered regions, thus raising the possibility that the mechanism that we uncovered for TIS11/TTP turnover is widespread among other RNA-BPs.
Collapse
|
25
|
Gao X, Dong H, Lin C, Sheng J, Zhang F, Su J, Xu Z. Reduction of AUF1-mediated follistatin mRNA decay during glucose starvation protects cells from apoptosis. Nucleic Acids Res 2014; 42:10720-30. [PMID: 25159612 PMCID: PMC4176339 DOI: 10.1093/nar/gku778] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Follistatin (FST) performs several vital functions in the cells, including protection from apoptosis during stress. The expression of FST is up-regulated in response to glucose deprivation by an unknown mechanism. We herein showed that the induction of FST by glucose deprivation was due to an increase in the half-life of its mRNA. We further identified an AU-rich element (ARE) in the 3′UTR of FST mRNA that mediated its decay. The expression of FST was elevated after knocking down AUF1 and reduced when AUF1 was further expressed. In vitro binding assays and RNA pull-down assays revealed that AUF1 interacted with FST mRNA directly via its ARE. During glucose deprivation, a majority of AUF1 shuttled from cytoplasm to nucleus, resulting in dissociation of AUF1 from FST mRNA and thus stabilization of FST mRNA. Finally, knockdown of AUF1 decreased whereas overexpression of AUF1 increased glucose deprivation-induced apoptosis. The apoptosis promoting effect of AUF1 was eliminated in FST expressing cells. Collectively, this study provided evidence that AUF1 is a negative regulator of FST expression and participates in the regulation of cell survival under glucose deprivation.
Collapse
Affiliation(s)
- Xiangwei Gao
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Haojie Dong
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Chen Lin
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jinghao Sheng
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Fan Zhang
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jinfeng Su
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zhengping Xu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
26
|
de Verteuil DA, Rouette A, Hardy MP, Lavallée S, Trofimov A, Gaucher É, Perreault C. Immunoproteasomes Shape the Transcriptome and Regulate the Function of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1121-32. [DOI: 10.4049/jimmunol.1400871] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
27
|
Niforou K, Cheimonidou C, Trougakos IP. Molecular chaperones and proteostasis regulation during redox imbalance. Redox Biol 2014; 2:323-32. [PMID: 24563850 PMCID: PMC3926111 DOI: 10.1016/j.redox.2014.01.017] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/11/2014] [Accepted: 01/18/2014] [Indexed: 02/05/2023] Open
Abstract
Free radicals originate from both exogenous environmental sources and as by-products of the respiratory chain and cellular oxygen metabolism. Sustained accumulation of free radicals, beyond a physiological level, induces oxidative stress that is harmful for the cellular homeodynamics as it promotes the oxidative damage and stochastic modification of all cellular biomolecules including proteins. In relation to proteome stability and maintenance, the increased concentration of oxidants disrupts the functionality of cellular protein machines resulting eventually in proteotoxic stress and the deregulation of the proteostasis (homeostasis of the proteome) network (PN). PN curates the proteome in the various cellular compartments and the extracellular milieu by modulating protein synthesis and protein machines assembly, protein recycling and stress responses, as well as refolding or degradation of damaged proteins. Molecular chaperones are key players of the PN since they facilitate folding of nascent polypeptides, as well as holding, folding, and/or degradation of unfolded, misfolded, or non-native proteins. Therefore, the expression and the activity of the molecular chaperones are tightly regulated at both the transcriptional and post-translational level at organismal states of increased oxidative and, consequently, proteotoxic stress, including ageing and various age-related diseases (e.g. degenerative diseases and cancer). In the current review we present a synopsis of the various classes of intra- and extracellular chaperones, the effects of oxidants on cellular homeodynamics and diseases and the redox regulation of chaperones. Free radicals originate from various sources and at physiological concentrations are essential for the modulation of cell signalling pathways. Abnormally high levels of free radicals induce oxidative stress and damage all cellular biomolecules, including proteins. Molecular chaperones facilitate folding of nascent polypeptides, as well as holding, folding, and/or degradation of damaged proteins. The expression and the activity of chaperones during oxidative stress are regulated at both the transcriptional and post-translational level.
Collapse
Key Words
- AGEs, Advanced Glycation End Products
- ALS, Autophagy Lysosome System
- AP-1, Activator Protein-1
- CLU, apolipoprotein J/Clusterin
- Chaperones
- Diseases
- EPMs, Enzymatic Protein Modifications
- ER, Endoplasmic Reticulum
- ERAD, ER-Associated protein Degradation
- Free radicals
- GPx7, Glutathione Peroxidase 7
- GRP78, Glucose Regulated Protein of 78 kDa
- HSF1, Heat Shock transcription Factor-1
- HSP, Heat Shock Protein
- Hb, Haemoglobin
- Keap1, Kelch-like ECH-associated protein 1
- NADH, Nicotinamide Adenine Dinucleotide
- NEPMs, Non-Enzymatic Protein Modifications
- NOS, Nitric Oxide Synthase
- NOx, NAD(P)H Oxidase
- Nrf2, NF-E2-related factor 2
- Oxidative stress
- PDI, Protein Disulfide Isomerase
- PDR, Proteome Damage Responses
- PN, Proteostasis Network
- Proteome
- RNS, Reactive Nitrogen Species
- ROS, Reactive Oxygen Species
- Redox signalling
- UPR, Unfolded Protein Response
- UPS, Ubiquitin Proteasome System
- α(2)M, α(2)-Macroglobulin
Collapse
Affiliation(s)
- Katerina Niforou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Christina Cheimonidou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| |
Collapse
|