101
|
Heat Shock Factor 1 Is a Substrate for p38 Mitogen-Activated Protein Kinases. Mol Cell Biol 2016; 36:2403-17. [PMID: 27354066 PMCID: PMC5007788 DOI: 10.1128/mcb.00292-16] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 12/04/2022] Open
Abstract
Heat shock factor 1 (HSF1) monitors the structural integrity of the proteome. Phosphorylation at S326 is a hallmark for HSF1 activation, but the identity of the kinase(s) phosphorylating this site has remained elusive. We show here that the dietary agent phenethyl isothiocyanate (PEITC) inhibits heat shock protein 90 (Hsp90), the main negative regulator of HSF1; activates p38 mitogen-activated protein kinase (MAPK); and increases S326 phosphorylation, trimerization, and nuclear translocation of HSF1, and the transcription of a luciferase reporter, as well as the endogenous prototypic HSF1 target Hsp70. In vitro, all members of the p38 MAPK family rapidly and stoichiometrically catalyze the S326 phosphorylation. The use of stable knockdown cell lines and inhibitors indicated that among the p38 MAPKs, p38γ is the principal isoform responsible for the phosphorylation of HSF1 at S326 in cells. A protease-mass spectrometry approach confirmed S326 phosphorylation and unexpectedly revealed that p38 MAPK also catalyzes the phosphorylation of HSF1 at S303/307, previously known repressive posttranslational modifications. Thus, we have identified p38 MAPKs as highly efficient catalysts for the phosphorylation of HSF1. Furthermore, our findings suggest that the magnitude and persistence of activation of p38 MAPK are important determinants of the extent and duration of the heat shock response.
Collapse
|
102
|
Goenka A, Sengupta S, Pandey R, Parihar R, Mohanta GC, Mukerji M, Ganesh S. Human satellite-III non-coding RNAs modulate heat-shock-induced transcriptional repression. J Cell Sci 2016; 129:3541-3552. [PMID: 27528402 DOI: 10.1242/jcs.189803] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/10/2016] [Indexed: 12/31/2022] Open
Abstract
The heat shock response is a conserved defense mechanism that protects cells from physiological stress, including thermal stress. Besides the activation of heat-shock-protein genes, the heat shock response is also known to bring about global suppression of transcription; however, the mechanism by which this occurs is poorly understood. One of the intriguing aspects of the heat shock response in human cells is the transcription of satellite-III (Sat3) long non-coding RNAs and their association with nuclear stress bodies (nSBs) of unknown function. Besides association with the Sat3 transcript, the nSBs are also known to recruit the transcription factors HSF1 and CREBBP, and several RNA-binding proteins, including the splicing factor SRSF1. We demonstrate here that the recruitment of CREBBP and SRSF1 to nSBs is Sat3-dependent, and that loss of Sat3 transcripts relieves the heat-shock-induced transcriptional repression of a few target genes. Conversely, forced expression of Sat3 transcripts results in the formation of nSBs and transcriptional repression even without a heat shock. Our results thus provide a novel insight into the regulatory role for the Sat3 transcripts in heat-shock-dependent transcriptional repression.
Collapse
Affiliation(s)
- Anshika Goenka
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Sonali Sengupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Rajesh Pandey
- CSIR Ayurgenomics Unit - TRISUTRA, CSIR - Institute of Genomics and Integrative Biology (IGIB), Mathura Road, New Delhi 110025, India
| | - Rashmi Parihar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Girish Chandra Mohanta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Mitali Mukerji
- CSIR Ayurgenomics Unit - TRISUTRA, CSIR - Institute of Genomics and Integrative Biology (IGIB), Mathura Road, New Delhi 110025, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| |
Collapse
|
103
|
Pincus D. Size doesn't matter in the heat shock response. Curr Genet 2016; 63:175-178. [PMID: 27502399 DOI: 10.1007/s00294-016-0638-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 11/25/2022]
Abstract
Heat shock factor 1 (Hsf1) is a transcription factor that is often described as the master regulator of the heat shock response in all eukaryotes. However, due to its essentiality in yeast, Hsf1's contribution to the transcriptome under basal and heat shock conditions has never been directly determined. Using a chemical genetics approach that allowed rapid Hsf1 inactivation, my colleagues and I have recently shown that the bulk of the heat shock response is Hsf1 independent. Rather than inducing genes responsible for carrying out the various cellular processes required for adaptation to thermal stress, Hsf1 controls a dedicated set of chaperone protein genes devoted to restoring protein-folding homeostasis. The limited scope of the Hsf1 regulon belies its outsize importance in cellular fitness.
Collapse
Affiliation(s)
- David Pincus
- Nine Cambridge Center, Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.
| |
Collapse
|
104
|
Transcriptional control of non-apoptotic developmental cell death in C. elegans. Cell Death Differ 2016; 23:1985-1994. [PMID: 27472063 DOI: 10.1038/cdd.2016.77] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/13/2016] [Accepted: 06/28/2016] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death is an essential aspect of animal development. Mutations in vertebrate genes that mediate apoptosis only mildly perturb development, suggesting that other cell death modes likely have important roles. Linker cell-type death (LCD) is a morphologically conserved cell death form operating during the development of Caenorhabditis elegans and vertebrates. We recently described a molecular network governing LCD in C. elegans, delineating a key role for the transcription factor heat-shock factor 1 (HSF-1). Although HSF-1 functions to protect cells from stress in many settings by inducing expression of protein folding chaperones, it promotes LCD by inducing expression of the conserved E2 ubiquitin-conjugating enzyme LET-70/UBE2D2, which is not induced by stress. Following whole-genome RNA interference and candidate gene screens, we identified and characterized four conserved regulators required for LCD. Here we show that two of these, NOB-1/Hox and EOR-1/PLZF, act upstream of HSF-1, in the context of Wnt signaling. A third protein, NHR-67/TLX/NR2E1, also functions upstream of HSF-1, and has a separate activity that prevents precocious expression of HSF-1 transcriptional targets. We demonstrate that the SET-16/mixed lineage leukemia 3/4 (MLL3/4) chromatin regulation complex functions at the same step or downstream of HSF-1 to control LET-70/UBE2D2 expression. Our results identify conserved proteins governing LCD, and demonstrate that transcriptional regulators influence this process at multiple levels.
Collapse
|
105
|
Yabuta Y. Functions of heat shock transcription factors involved in response to photooxidative stresses in Arabidopsis. Biosci Biotechnol Biochem 2016; 80:1254-63. [DOI: 10.1080/09168451.2016.1176515] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Because plants are continually exposed to various environmental stresses, they possess numerous transcription factors that regulate metabolism to adapt and acclimate to those conditions. To clarify the gene regulation systems activated in response to photooxidative stress, we isolated 76 high light and heat shock stress-inducible genes, including heat shock transcription factor (Hsf) A2 from Arabidopsis. Unlike yeast or animals, more than 20 genes encoding putative Hsfs are present in the genomes of higher plants, and they are categorized into three classes based on their structural characterization. However, the multiplicity of Hsfs in plants remains unknown. Furthermore, the individual functions of Hsfs are also largely unknown because of their genetic redundancy. Recently, the developments of T-DNA insertion knockout mutant lines and chimeric repressor gene-silencing technology have provided effective tools for exploring the individual functions of Hsfs. This review describes the current knowledge on the individual functions and activation mechanisms of Hsfs.
Collapse
Affiliation(s)
- Yukinori Yabuta
- Faculty of Agriculture, School of Agricultural, Biological, and Environmental Sciences, Tottori University, Tottori, Japan
| |
Collapse
|
106
|
Karvinen S, Silvennoinen M, Vainio P, Sistonen L, Koch LG, Britton SL, Kainulainen H. Effects of intrinsic aerobic capacity, aging and voluntary running on skeletal muscle sirtuins and heat shock proteins. Exp Gerontol 2016; 79:46-54. [DOI: 10.1016/j.exger.2016.03.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/03/2016] [Accepted: 03/23/2016] [Indexed: 11/25/2022]
|
107
|
Butler JS, Chan A, Costelha S, Fishman S, Willoughby JLS, Borland TD, Milstein S, Foster DJ, Gonçalves P, Chen Q, Qin J, Bettencourt BR, Sah DW, Alvarez R, Rajeev KG, Manoharan M, Fitzgerald K, Meyers RE, Nochur SV, Saraiva MJ, Zimmermann TS. Preclinical evaluation of RNAi as a treatment for transthyretin-mediated amyloidosis. Amyloid 2016; 23:109-18. [PMID: 27033334 PMCID: PMC4898164 DOI: 10.3109/13506129.2016.1160882] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
ATTR amyloidosis is a systemic, debilitating and fatal disease caused by transthyretin (TTR) amyloid accumulation. RNA interference (RNAi) is a clinically validated technology that may be a promising approach to the treatment of ATTR amyloidosis. The vast majority of TTR, the soluble precursor of TTR amyloid, is expressed and synthesized in the liver. RNAi technology enables robust hepatic gene silencing, the goal of which would be to reduce systemic levels of TTR and mitigate many of the clinical manifestations of ATTR that arise from hepatic TTR expression. To test this hypothesis, TTR-targeting siRNAs were evaluated in a murine model of hereditary ATTR amyloidosis. RNAi-mediated silencing of hepatic TTR expression inhibited TTR deposition and facilitated regression of existing TTR deposits in pathologically relevant tissues. Further, the extent of deposit regression correlated with the level of RNAi-mediated knockdown. In comparison to the TTR stabilizer, tafamidis, RNAi-mediated TTR knockdown led to greater regression of TTR deposits across a broader range of affected tissues. Together, the data presented herein support the therapeutic hypothesis behind TTR lowering and highlight the potential of RNAi in the treatment of patients afflicted with ATTR amyloidosis.
Collapse
Affiliation(s)
| | - Amy Chan
- a Alnylam Pharmaceuticals , Cambridge , MA , USA
| | - Susete Costelha
- b Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal .,c Molecular Neurobiology, IBMC - Institute for Molecular and Cell Biology Porto , Porto , Portugal
| | | | | | | | | | | | - Paula Gonçalves
- b Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal .,c Molecular Neurobiology, IBMC - Institute for Molecular and Cell Biology Porto , Porto , Portugal
| | - Qingmin Chen
- a Alnylam Pharmaceuticals , Cambridge , MA , USA
| | - June Qin
- a Alnylam Pharmaceuticals , Cambridge , MA , USA
| | | | - Dinah W Sah
- a Alnylam Pharmaceuticals , Cambridge , MA , USA
| | - Rene Alvarez
- a Alnylam Pharmaceuticals , Cambridge , MA , USA
| | | | | | | | | | | | - Maria J Saraiva
- b Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal .,c Molecular Neurobiology, IBMC - Institute for Molecular and Cell Biology Porto , Porto , Portugal
| | | |
Collapse
|
108
|
Krishnamurthy K, Glaser S, Alpini GD, Cardounel AJ, Liu Z, Ilangovan G. Heat shock factor-1 knockout enhances cholesterol 7α-hydroxylase (CYP7A1) and multidrug transporter (MDR1) gene expressions to attenuate atherosclerosis. Cardiovasc Res 2016; 111:74-83. [PMID: 27131506 DOI: 10.1093/cvr/cvw094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 04/27/2016] [Indexed: 12/23/2022] Open
Abstract
AIMS Stress response, in terms of activation of stress factors, is known to cause obesity and coronary heart disease such as atherosclerosis in human. However, the underlying mechanism(s) of these pathways are not known. Here, we investigated the effect of heat shock factor-1 (HSF-1) on atherosclerosis. METHODS AND RESULTS HSF-1 and low-density lipoprotein receptor (LDLr) double knockout (HSF-1(-/-)/LDLr(-/-)) and LDLr knockout (LDLr(-/-)) mice were fed with atherogenic western diet (WD) for 12 weeks. WD-induced weight gain and atherosclerotic lesion in aortic arch and carotid regions were reduced in HSF-1(-/-)/LDLr(-/-) mice, compared with LDLr(-/-) mice. Also, repression of PPAR-γ2 and AMPKα expression in adipose tissue, low hepatic steatosis, and lessened plasma adiponectins and lipoproteins were observed. In HSF-1(-/-)/LDLr(-/-) liver, higher cholesterol 7α-hydroxylase (CYP7A1) and multidrug transporter [MDR1/P-glycoprotein (P-gp)] gene expressions were observed, consistent with higher bile acid transport and larger hepatic bile ducts. Luciferase reporter gene assays with wild-type CYP7A1 and MDR1 promoters showed lesser luminescence than with mutant promoters (HSF-1 binding site deleted), indicating that HSF-1 binding is repressive of CYP7A1 and MDR1 gene expressions. CONCLUSION HSF-1 ablation not only eliminates heat shock response, but it also transcriptionally up-regulates CYP7A1 and MDR1/P-gp axis in WD-diet fed HSF-1(-/-)/LDLr(-/-) mice to reduce atherosclerosis.
Collapse
Affiliation(s)
- Karthikeyan Krishnamurthy
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Scott & White Digestive Disease Research Center, Medicine, Scott and White and Texas A&M Health Science Center, Temple, TX 76504, USA
| | - Gianfranco D Alpini
- Research, Central Texas Veterans Health Care System, Scott & White Digestive Disease Research Center, Medicine, Scott and White and Texas A&M Health Science Center, Temple, TX 76504, USA
| | - Arturo J Cardounel
- Department of Anesthesiology, The Ohio State University, Columbus, OH 43210, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Govindasamy Ilangovan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
109
|
HSF1 critically attunes proteotoxic stress sensing by mTORC1 to combat stress and promote growth. Nat Cell Biol 2016; 18:527-39. [PMID: 27043084 PMCID: PMC5341796 DOI: 10.1038/ncb3335] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 03/02/2016] [Indexed: 12/16/2022]
Abstract
To cope with proteotoxic stress, cells attenuate protein synthesis. However, the precise mechanisms underlying this fundamental adaptation remain poorly defined. Here we report that mTORC1 acts as an immediate cellular sensor of proteotoxic stress. Surprisingly, the multifaceted stress-responsive kinase JNK constitutively associates with mTORC1 under normal growth conditions. On activation by proteotoxic stress, JNK phosphorylates both RAPTOR at S863 and mTOR at S567, causing partial disintegration of mTORC1 and subsequent translation inhibition. Importantly, HSF1, the central player in the proteotoxic stress response (PSR), preserves mTORC1 integrity and function by inactivating JNK, independently of its canonical transcriptional action. Thereby, HSF1 translationally augments the PSR. Beyond promoting stress resistance, this intricate HSF1-JNK-mTORC1 interplay, strikingly, regulates cell, organ and body sizes. Thus, these results illuminate a unifying mechanism that controls stress adaptation and growth.
Collapse
|
110
|
Xu D, Sun L, Liu S, Zhang L, Yang H. Molecular cloning of hsf1 and hsbp1 cDNAs, and the expression of hsf1, hsbp1 and hsp70 under heat stress in the sea cucumber Apostichopus japonicus. Comp Biochem Physiol B Biochem Mol Biol 2016; 198:1-9. [PMID: 26952354 DOI: 10.1016/j.cbpb.2016.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 02/07/2023]
Abstract
The heat shock response (HSR) is known for the elevated synthesis of heat shock proteins (HSPs) under heat stress, which is mediated primarily by heat shock factor 1 (HSF1). Heat shock factor binding protein 1 (HSBP1) and feedback control of heat shock protein 70 (HSP70) are major regulators of the activity of HSF1. We obtained full-length cDNA of genes hsf1 and hsbp1 in the sea cucumber Apostichopus japonicus, which are the second available for echinoderm (after Strongylocentrotus purpuratus), and the first available for holothurian. The full-length cDNA of hsf1 was 2208bp, containing a 1326bp open reading frame encoding 441 amino acids. The full-length cDNA of hsbp1 was 2850bp, containing a 225bp open reading frame encoding 74 amino acids. The similarities of A. japonicus HSF1 with other species are low, and much higher similarity identities of A. japonicus HSBP1 were shared. Phylogenetic trees showed that A. japonicus HSF1 and HSBP1 were clustered with sequences from S. purpuratus, and fell into distinct clades with sequences from mollusca, arthropoda and vertebrata. Analysis by real-time PCR showed hsf1 and hsbp1 mRNA was expressed constitutively in all tissues examined. The expression of hsf1, hsbp1 and hsp70 in the intestine at 26°C was time-dependent. The results of this study might provide new insights into the regulation of heat shock response in this species.
Collapse
Affiliation(s)
- Dongxue Xu
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Lina Sun
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China.
| | - Shilin Liu
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
| | - Libin Zhang
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
| | - Hongsheng Yang
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China.
| |
Collapse
|
111
|
The transcriptional coactivator PGC1α protects against hyperthermic stress via cooperation with the heat shock factor HSF1. Cell Death Dis 2016; 7:e2102. [PMID: 26890141 PMCID: PMC5399192 DOI: 10.1038/cddis.2016.22] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 01/19/2023]
Abstract
Heat shock proteins (HSPs) are required for the clearance of damaged and aggregated proteins and have important roles in protein homeostasis. It has been shown that the heat shock transcription factor, HSF1, orchestrates the transcriptional induction of these stress-regulated chaperones; however, the coregulatory factors responsible for the enhancement of HSF1 function on these target genes have not been fully elucidated. Here, we demonstrate that the cold-inducible coactivator, PGC1α, also known for its role as a regulator of mitochondrial and peroxisomal biogenesis, thermogenesis and cytoprotection from oxidative stress, regulates the expression of HSPs in vitro and in vivo and modulates heat tolerance. Mechanistically, we show that PGC1α physically interacts with HSF1 on HSP promoters and that cells and mice lacking PGC1α have decreased HSPs levels and are more sensitive to thermal challenges. Taken together, our findings suggest that PGC1α protects against hyperthermia by cooperating with HSF1 in the induction of a transcriptional program devoted to the cellular protection from thermal insults.
Collapse
|
112
|
Hentze N, Le Breton L, Wiesner J, Kempf G, Mayer MP. Molecular mechanism of thermosensory function of human heat shock transcription factor Hsf1. eLife 2016; 5. [PMID: 26785146 PMCID: PMC4775227 DOI: 10.7554/elife.11576] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/18/2016] [Indexed: 01/06/2023] Open
Abstract
The heat shock response is a universal homeostatic cell autonomous reaction of organisms to cope with adverse environmental conditions. In mammalian cells, this response is mediated by the heat shock transcription factor Hsf1, which is monomeric in unstressed cells and upon activation trimerizes, and binds to promoters of heat shock genes. To understand the basic principle of Hsf1 activation we analyzed temperature-induced alterations in the conformational dynamics of Hsf1 by hydrogen exchange mass spectrometry. We found a temperature-dependent unfolding of Hsf1 in the regulatory region happening concomitant to tighter packing in the trimerization region. The transition to the active DNA binding-competent state occurred highly cooperative and was concentration dependent. Surprisingly, Hsp90, known to inhibit Hsf1 activation, lowered the midpoint temperature of trimerization and reduced cooperativity of the process thus widening the response window. Based on our data we propose a kinetic model of Hsf1 trimerization.
Collapse
Affiliation(s)
- Nikolai Hentze
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Laura Le Breton
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Jan Wiesner
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Georg Kempf
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Matthias P Mayer
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
113
|
Neudegger T, Verghese J, Hayer-Hartl M, Hartl FU, Bracher A. Structure of human heat-shock transcription factor 1 in complex with DNA. Nat Struct Mol Biol 2016; 23:140-6. [PMID: 26727489 DOI: 10.1038/nsmb.3149] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023]
Abstract
Heat-shock transcription factor 1 (HSF1) has a central role in mediating the protective response to protein conformational stresses in eukaryotes. HSF1 consists of an N-terminal DNA-binding domain (DBD), a coiled-coil oligomerization domain, a regulatory domain and a transactivation domain. Upon stress, HSF1 trimerizes via its coiled-coil domain and binds to the promoters of heat shock protein-encoding genes. Here, we present cocrystal structures of the human HSF1 DBD in complex with cognate DNA. A comparative analysis of the HSF1 paralog Skn7 from Chaetomium thermophilum showed that single amino acid changes in the DBD can switch DNA binding specificity, thus revealing the structural basis for the interaction of HSF1 with cognate DNA. We used a crystal structure of the coiled-coil domain of C. thermophilum Skn7 to develop a model of the active human HSF1 trimer in which HSF1 embraces the heat-shock-element DNA.
Collapse
Affiliation(s)
- Tobias Neudegger
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jacob Verghese
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Manajit Hayer-Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
114
|
Crombie TA, Tang L, Choe KP, Julian D. Inhibition of the oxidative stress response by heat stress in Caenorhabditis elegans. J Exp Biol 2016; 219:2201-11. [DOI: 10.1242/jeb.135327] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/09/2016] [Indexed: 12/14/2022]
Abstract
It has long been recognized that simultaneous exposure to heat stress and oxidative stress shows a synergistic interaction that reduces organismal fitness, but relatively little is known about the mechanisms underlying this interaction. We investigated the role of molecular stress responses in driving this synergistic interaction using the nematode Caenorhabditis elegans. To induce oxidative stress, we used the pro-oxidant compounds acrylamide, paraquat, and juglone. As expected, we found that heat stress and oxidative stress interact synergistically to reduce survival. Compared to exposure to each stressor alone, during simultaneous, sub-lethal exposure to heat stress and oxidative stress the normal induction of key oxidative stress response (OxSR) genes was generally inhibited while the induction of key heat shock response (HSR) genes was not. Genetically activating the SKN-1 dependent OxSR increased a marker for protein aggregation and decreased whole-worm survival during heat stress alone, with the latter being independent of HSF-1. In contrast, inactivating the HSR by HSF-1 knockdown, which would be expected to decrease basal heat shock protein expression, increased survival during oxidative stress alone compared to wild- type worms. Taken together, these data suggest that in C. elegans the HSR and OxSR cannot be simultaneously activated to the same extent that each can be activated during a single stressor exposure. We conclude that the observed synergistic reduction in survival during combined exposure to heat stress and oxidative stress is due, at least in part, to inhibition of the OxSR during activation of the HSR.
Collapse
Affiliation(s)
| | - Lanlan Tang
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Keith P. Choe
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - David Julian
- Department of Biology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
115
|
Vijayakumaran R, Tan KH, Miranda PJ, Haupt S, Haupt Y. Regulation of Mutant p53 Protein Expression. Front Oncol 2015; 5:284. [PMID: 26734569 PMCID: PMC4681805 DOI: 10.3389/fonc.2015.00284] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/02/2015] [Indexed: 12/19/2022] Open
Abstract
For several decades, p53 has been detected in cancer biopsies by virtue of its high protein expression level which is considered indicative of mutation. Surprisingly, however, mouse genetic studies revealed that mutant p53 is inherently labile, similar to its wild type (wt) counterpart. Consistently, in response to stress conditions, both wt and mutant p53 accumulate in cells. While wt p53 returns to basal level following recovery from stress, mutant p53 remains stable. In part, this can be explained in mutant p53-expressing cells by the lack of an auto-regulatory loop with Mdm2 and other negative regulators, which are pivotal for wt p53 regulation. Further, additional protective mechanisms are acquired by mutant p53, largely mediated by the co-chaperones and their paralogs, the stress-induced heat shock proteins. Consequently, mutant p53 is accumulated in cancer cells in response to chronic stress and this accumulation is critical for its oncogenic gain of functions (GOF). Building on the extensive knowledge regarding wt p53, the regulation of mutant p53 is unraveling. In this review, we describe the current understanding on the major levels at which mutant p53 is regulated. These include the regulation of p53 protein levels by microRNA and by enzymes controlling p53 proteasomal degradation.
Collapse
Affiliation(s)
- Reshma Vijayakumaran
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Kah Hin Tan
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | | | - Sue Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Ygal Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology and Department of Pathology, The University of Melbourne, Parkville, VIC, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
116
|
Ohno Y, Egawa T, Yokoyama S, Nakai A, Sugiura T, Ohira Y, Yoshioka T, Goto K. Deficiency of heat shock transcription factor 1 suppresses heat stress-associated increase in slow soleus muscle mass of mice. Acta Physiol (Oxf) 2015; 215:191-203. [PMID: 26347147 DOI: 10.1111/apha.12600] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 07/20/2015] [Accepted: 09/03/2015] [Indexed: 01/30/2023]
Abstract
AIM Effects of heat shock transcription factor 1 (HSF1) deficiency on heat stress-associated increase in slow soleus muscle mass of mice were investigated. METHODS Both HSF1-null and wild-type mice were randomly assigned to control and heat-stressed groups. Mice in heat-stressed group were exposed to heat stress (41 °C for 60 min) in an incubator without anaesthesia. RESULTS Significant increase in wet and dry weights, and protein content of soleus muscle in wild-type mice was observed seven days after the application of the heat stress. However, heat stress had no impact on soleus muscle mass in HSF1-null mice. Neither type of mice exhibited much effect of heat stress on HSF mRNA expression (HSF1, HSF2 and HSF4). On the other hand, heat stress upregulated heat shock proteins (HSPs) at the mRNA (HSP72) and protein (HSP72 and HSP110) levels in wild-type mice, but not in HSF1-null mice. The population of Pax7-positive nuclei relative to total myonuclei of soleus muscle in wild-type mice was significantly increased by heat stress, but not in HSF1-null mice. Furthermore, the absence of HSF1 gene suppressed heat stress-associated phosphorylation of Akt and p70 S6 kinase (p-p70S6K) in soleus muscle. CONCLUSION Heat stress-associated increase in skeletal muscle mass may be induced by HSF1 and/or HSF1-mediated stress response that activates muscle satellite cells and Akt/p70S6K signalling pathway.
Collapse
Affiliation(s)
- Y. Ohno
- Laboratory of Physiology; School of Health Sciences; Toyohashi SOZO University; Toyohashi Japan
| | - T. Egawa
- Department of Physiology; Graduate School of Health Sciences; Toyohashi SOZO University; Toyohashi Japan
- Research Fellow of the Japan Society for the Promotion of Science; Tokyo Japan
| | - S. Yokoyama
- Laboratory of Physiology; School of Health Sciences; Toyohashi SOZO University; Toyohashi Japan
| | - A. Nakai
- Department of Molecular Biology; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - T. Sugiura
- Faculty of Education; Yamaguchi University; Yamaguchi Japan
| | - Y. Ohira
- Graduate School of Health and Sports Science; Doshisha University; Kyotanabe Japan
| | | | - K. Goto
- Laboratory of Physiology; School of Health Sciences; Toyohashi SOZO University; Toyohashi Japan
- Department of Physiology; Graduate School of Health Sciences; Toyohashi SOZO University; Toyohashi Japan
| |
Collapse
|
117
|
HSF1: Guardian of Proteostasis in Cancer. Trends Cell Biol 2015; 26:17-28. [PMID: 26597576 DOI: 10.1016/j.tcb.2015.10.011] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Proteomic instability is causally related to human diseases. In guarding proteome stability, the heat shock factor 1 (HSF1)-mediated proteotoxic stress response plays a pivotal role. Contrasting with its beneficial role of enhancing cell survival, recent findings have revealed a compelling pro-oncogenic role for HSF1. However, the mechanisms underlying the persistent activation and function of HSF1 within malignancy remain poorly understood. Emerging evidence reveals that oncogenic signaling mobilizes HSF1 and that cancer cells rely on HSF1 to avert proteomic instability and repress tumor-suppressive amyloidogenesis. In aggregate, these new developments suggest that cancer cells endure chronic proteotoxic stress and that proteomic instability is intrinsically associated with the malignant state, a characteristic that could be exploited to combat cancer.
Collapse
|
118
|
Huang C, Lu X, Tong L, Wang J, Zhang W, Jiang B, Yang R. Requirement for endogenous heat shock factor 1 in inducible nitric oxide synthase induction in murine microglia. J Neuroinflammation 2015; 12:189. [PMID: 26467650 PMCID: PMC4607096 DOI: 10.1186/s12974-015-0406-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/28/2015] [Indexed: 01/17/2023] Open
Abstract
Background Inducible nitric oxide synthase (iNOS) makes a great contribution to host defense and inflammation. In many settings, lipopolysaccharide (LPS) induces iNOS expression through activation of the inhibitor of κB-α (IκB-α)-nuclear factor-κB (NF-κB) cascade, whereas interferon-γ (IFN-γ) acts through Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) signals. Heat shock factor 1 (HSF1), a major regulator of heat shock protein transcription, has been shown to regulate the production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), but it remains obscure whether and how HSF1 affects iNOS induction. Methods Western blot was used to measure the protein expression. The mRNA level was measured by real-time PCR. Silence of HSF1 was achieved by small interfering RNA. Nitric oxide (NO) content and NF-κB binding activity were assayed by commercial kits. Chromatin immunoprecipitation (ChIP) was used to measure the binding activity of NF-κB and STAT1 to iNOS promoters. Results HSF1 inhibition or knockdown prevented the LPS- and/or IFN-γ-stimulated iNOS protein expression in cultured microglia. HSF1 inhibition blocked iNOS mRNA transcription. These inhibitory effects of HSF1 inhibition on iNOS expression were confirmed in brain tissues from endotoxemic mice. Further analysis showed that HSF1 inhibition had no effect on IκB-α degradation and NF-κB or STAT1 phosphorylation in LPS/IFN-γ-stimulated cells. The nuclear transport of active NF-κB or STAT1 was also not affected by HSF1 inhibition, but HSF1 inhibition reduced the binding of NF-κB and STAT1 to their DNA elements. In addition, HSF1 inhibition reduced NF-κB and STAT1 bindings to iNOS promoter inside the LPS/IFN-γ-stimulated cells. Conclusions This preventing effect of HSF1 inhibition on iNOS mRNA transcription presents the necessary role of HSF1 in iNOS induction.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Jili Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20Xisi Road, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
119
|
Direct link between metabolic regulation and the heat-shock response through the transcriptional regulator PGC-1α. Proc Natl Acad Sci U S A 2015; 112:E5669-78. [PMID: 26438876 DOI: 10.1073/pnas.1516219112] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years an extensive effort has been made to elucidate the molecular pathways involved in metabolic signaling in health and disease. Here we show, surprisingly, that metabolic regulation and the heat-shock/stress response are directly linked. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a critical transcriptional coactivator of metabolic genes, acts as a direct transcriptional repressor of heat-shock factor 1 (HSF1), a key regulator of the heat-shock/stress response. Our findings reveal that heat-shock protein (HSP) gene expression is suppressed during fasting in mouse liver and in primary hepatocytes dependent on PGC-1α. HSF1 and PGC-1α associate physically and are colocalized on several HSP promoters. These observations are extended to several cancer cell lines in which PGC-1α is shown to repress the ability of HSF1 to activate gene-expression programs necessary for cancer survival. Our study reveals a surprising direct link between two major cellular transcriptional networks, highlighting a previously unrecognized facet of the activity of the central metabolic regulator PGC-1α beyond its well-established ability to boost metabolic genes via its interactions with nuclear hormone receptors and nuclear respiratory factors. Our data point to PGC-1α as a critical repressor of HSF1-mediated transcriptional programs, a finding with possible implications both for our understanding of the full scope of metabolically regulated target genes in vivo and, conceivably, for therapeutics.
Collapse
|
120
|
Lamech LT, Haynes CM. The unpredictability of prolonged activation of stress response pathways. J Cell Biol 2015; 209:781-7. [PMID: 26101215 PMCID: PMC4477854 DOI: 10.1083/jcb.201503107] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In response to stress, cellular compartments activate signaling pathways that mediate transcriptional programs to promote survival and reestablish homeostasis. Manipulation of the magnitude and duration of the activation of stress responses has been proposed as a strategy to prevent or repair the damage associated with aging or degenerative diseases. However, as these pathways likely evolved to respond specifically to transient perturbations, the unpredictability of prolonged activation should be considered.
Collapse
Affiliation(s)
- Lilian T Lamech
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Cole M Haynes
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 BCMB Allied Program, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
121
|
Abstract
Mammalian tissues are always exposed to diverse threats from pathological conditions and
aging. Therefore, the molecular systems that protect the cells from these threats are
indispensable for cell survival. A variety of diseases, including neurodegenerative
diseases, cause intracellular damage and disturb homeostasis. Heat shock transcription
factor 1 (HSF1) positively regulates heat shock protein (Hsp) and maintains the precise
folding of proteins. Moreover, HSF1 induces the non-Hsp genes expression, and degrades
damaged/misfolded protein. Recently, my colleagues and I revealed non-Hsp genes have more
protective roles than Hsps at the cellular level. However, whether these protective
systems are similarly important to cellular defense in each tissue is still elusive. In
this study, I compared polyglutamine (polyQ) protein aggregations/inclusion development in
each tissue of WT- and HSF1KO-Huntington’s disease (HD) mice, and examined the expression
of the eight non-Hsp HSF1 target genes that have a strong suppressive effect on polyQ
protein aggregation. Of these genes, Nfatc2, Pdzk3, Cryab, Csrp2, and Prame were detected
in most tissues, but the other genes were not. Surprisingly, the obvious effect of HSF1
deficiency on the expression of these five genes was detected in only heart, spleen, and
stomach. In addition, polyQ protein aggregations/inclusion was not detected in any tissues
of WT-HD and HSF1KO-HD mice, but higher level of pre-aggregative polyQ protein was
detected in HSF1KO-HD tissues. These results indicate non-Hsp genes are indispensable for
the maintenance of intracellular homeostasis in mammalian tissues, resulting in whole body
homeostasis.
Collapse
Affiliation(s)
- Naoki Hayashida
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
122
|
Uncoupling Stress-Inducible Phosphorylation of Heat Shock Factor 1 from Its Activation. Mol Cell Biol 2015; 35:2530-40. [PMID: 25963659 DOI: 10.1128/mcb.00816-14] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 05/04/2015] [Indexed: 12/31/2022] Open
Abstract
In mammals the stress-inducible expression of genes encoding heat shock proteins is under the control of the heat shock transcription factor 1 (HSF1). Activation of HSF1 is a multistep process, involving trimerization, acquisition of DNA-binding and transcriptional activities, which coincide with several posttranslational modifications. Stress-inducible phosphorylation of HSF1, or hyperphosphorylation, which occurs mainly within the regulatory domain (RD), has been proposed as a requirement for HSF-driven transcription and is widely used for assessing HSF1 activation. Nonetheless, the contribution of hyperphosphorylation to the activity of HSF1 remains unknown. In this study, we generated a phosphorylation-deficient HSF1 mutant (HSF1Δ∼PRD), where the 15 known phosphorylation sites within the RD were disrupted. Our results show that the phosphorylation status of the RD does not affect the subcellular localization and DNA-binding activity of HSF1. Surprisingly, under stress conditions, HSF1Δ∼PRD is a potent transactivator of both endogenous targets and a reporter gene, and HSF1Δ∼PRD has a reduced activation threshold. Our results provide the first direct evidence for uncoupling stress-inducible phosphorylation of HSF1 from its activation, and we propose that the phosphorylation signature alone is not an appropriate marker for HSF1 activity.
Collapse
|
123
|
Chen X, Zhang J, Han C, Dai H, Kong X, Xu L, Xia Q, Zhang M, Zhang J. A sexual dimorphism influences bicyclol-induced hepatic heat shock factor 1 activation and hepatoprotection. Mol Pharmacol 2015; 88:38-47. [PMID: 25901028 DOI: 10.1124/mol.114.097584] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/21/2015] [Indexed: 01/10/2023] Open
Abstract
Bicyclol [4,4'-dimethoxy-5,6,5',6'-bis(methylenedioxy)-2-hydroxy-methyl-2'-methoxycarbonyl biphenyl] is a synthetic hepatoprotectant widely used in clinical practice, but resistance to this treatment is often observed. We found that the hepatoprotective effect of bicyclol was greatly compromised in female and castrated male mice. This study was to dissect the molecular basis behind the sex difference, which might underlie the clinical uncertainty. We compared bicyclol-induced hepatoprotection between male and female mice using acute liver damage models. Inducible knockout by the Cre/loxp system was used to decipher the role of heat shock transcription factor 1 (HSF1). Functional experiments, western blot, and histopathological analysis were used to determine the key causative factors which might antagonize bicyclol in female livers. HSF1 activation and heat shock protein 70 (Hsp70) expression, which were responsible for bicyclol-induced hepatoprotection, were compromised in female and castrated male livers. Compromised HSF1 activation was a result of HSF1 phosphorylation at serine 303, which was catalyzed by glycogen synthase kinase 3β (GSK3β). Testosterone was necessary for bicyclol to inhibit hepatic GSK3β activity. Administration of testosterone or GSK3β inhibitors restored bicyclol-induced protection in females. Bicyclol induces sex-specific hepatoprotection based on a sex-specific HSF1/Hsp70 response, in which testosterone and GSK3β play key roles. Because a lot of patients suffering from liver diseases have very low testosterone levels, our results give a possible explanation for the clinical variation in bicyclol-induced hepatoprotection, as well as practicable solutions to improve the effect of bicyclol.
Collapse
Affiliation(s)
- Xiaosong Chen
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Jianjian Zhang
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Conghui Han
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Huijuan Dai
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Xianming Kong
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Longmei Xu
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Qiang Xia
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Ming Zhang
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| | - Jianjun Zhang
- Department of Transplantation and Hepatic Surgery, Renji Hospital (X.C., Jianjian Z., H.D., X.K., Q.X., M.Z., Jianjun Z.), and The Central Laboratory of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (L.X.); and Department of Urology, Xuzhou Central Hospital, Xuzhou Medical University School of Clinical Medicine, Xuzhou, China (C.H.)
| |
Collapse
|
124
|
Zhou X, Fan LX, Peters DJM, Trudel M, Bradner JE, Li X. Therapeutic targeting of BET bromodomain protein, Brd4, delays cyst growth in ADPKD. Hum Mol Genet 2015; 24:3982-93. [PMID: 25877301 DOI: 10.1093/hmg/ddv136] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023] Open
Abstract
In this study, we identified a BET bromodomain (BRD) protein, Brd4, not only as a novel epigenetic regulator of autosomal dominant polycystic kidney disease (ADPKD) but also as a novel client protein of Hsp90. We found that Brd4 was upregulated in Pkd1 mutant mouse renal epithelial cells and tissues. This upregulation of Brd4 appears to result from the chaperone activity of Hsp90 and escape proteasomal degradation. We further identify that Brd4 is an upstream regulator of the expression of c-Myc which has been upregulated in all rodent models of PKD and ADPKD patients with unknown mechanism. Inhibition of Brd4 in Pkd1 mutant renal epithelial cells with JQ1, a selective small-molecular inhibitor of BET BRD protein(s), (1) decreased the levels of c-Myc mRNA and protein; (2) increased the levels of p21 mRNA and protein, which was transcriptionally repressed by c-Myc; (3) decreased the phosphorylation of Rb; and (4) decreased cystic epithelial cell proliferation as shown by inhibition of S-phase entry. Most importantly, treatment with JQ1 strikingly delayed cyst growth and kidney enlargement, and preserved renal function in two early stage genetic mouse strains with Pkd1 mutations. This study not only provides one of the mechanisms of how c-Myc is upregulated in PKD but also suggests that targeting Brd4 with JQ1 may function as a novel epigenetic approach in ADPKD. The unraveled link between Brd4 and Hsp90 in ADPKD may also be a general mechanism for the upregulation of Brd4 in cancer cells and opens up avenues for combination therapies against ADPKD and cancer.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Internal Medicine, Kidney Institute
| | - Lucy X Fan
- Department of Internal Medicine, Kidney Institute
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Trudel
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montreal, Université de Montreal, Faculté de Médecine, Montreal, Quebec, Canada and
| | - James E Bradner
- Department of Medicine, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaogang Li
- Department of Internal Medicine, Kidney Institute, Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA,
| |
Collapse
|
125
|
MEK guards proteome stability and inhibits tumor-suppressive amyloidogenesis via HSF1. Cell 2015; 160:729-744. [PMID: 25679764 DOI: 10.1016/j.cell.2015.01.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/09/2014] [Accepted: 12/29/2014] [Indexed: 01/04/2023]
Abstract
Signaling through RAS/MAP kinase pathway is central to biology. ERK has long been perceived as the only substrate for MEK. Here, we report that HSF1, the master regulator of the proteotoxic stress response, is a new MEK substrate. Beyond mediating cell-environment interactions, the MEK-HSF1 regulation impacts malignancy. In tumor cells, MEK blockade inactivates HSF1 and thereby provokes proteomic chaos, presented as protein destabilization, aggregation, and, strikingly, amyloidogenesis. Unlike their non-transformed counterparts, tumor cells are particularly susceptible to proteomic perturbation and amyloid induction. Amyloidogenesis is tumor suppressive, reducing in vivo melanoma growth and contributing to the potent anti-neoplastic effects of proteotoxic stressors. Our findings unveil a key biological function of the oncogenic RAS-MEK signaling in guarding proteostasis and suppressing amyloidogenesis. Thus, proteomic instability is an intrinsic feature of malignant state, and disrupting the fragile tumor proteostasis to promote amyloidogenesis may be a feasible therapeutic strategy.
Collapse
|
126
|
Gregori S, Amodio G, Quattrone F, Panina-Bordignon P. HLA-G Orchestrates the Early Interaction of Human Trophoblasts with the Maternal Niche. Front Immunol 2015; 6:128. [PMID: 25870595 PMCID: PMC4378286 DOI: 10.3389/fimmu.2015.00128] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/09/2015] [Indexed: 01/13/2023] Open
Abstract
Extravillous trophoblasts (EVTs) play a central role in educating maternal leukocytes, endometrial stromal and endothelial cells to generate a receptive decidual microenvironment tailored to accept the semi-allogeneic fetus. HLA-G, a non-classical HLA class I molecule endowed with immune-regulatory functions, is primarily expressed on EVTs lining the placenta and on the naturally occurring tolerogenic dendritic cells, named DC-10, which are enriched in the human first trimester decidua. Decidual DC-10 are involved in HLA-G-mediated tolerance at the maternal–fetal interface. EVTs not only establish a tolerogenic microenvironment through the interaction with maternal innate and adaptive cells but also orchestrate placenta vascular and tissue remodeling, leading to a successful pregnancy. Here, we discuss the potential implications of the HLA-G-mediated cross-talk among the cells present at the maternal–fetal interface, and its role in maintaining a positive relationship between the mother and the fetus.
Collapse
Affiliation(s)
- Silvia Gregori
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Giada Amodio
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Federica Quattrone
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, IRCCS San Raffaele Hospital , Milan , Italy
| | - Paola Panina-Bordignon
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, IRCCS San Raffaele Hospital , Milan , Italy
| |
Collapse
|
127
|
Abstract
Loss of protein homeostasis (proteostasis) is a common feature of aging and disease that is characterized by the appearance of nonnative protein aggregates in various tissues. Protein aggregation is routinely suppressed by the proteostasis network (PN), a collection of macromolecular machines that operate in diverse ways to maintain proteome integrity across subcellular compartments and between tissues to ensure a healthy life span. Here, we review the composition, function, and organizational properties of the PN in the context of individual cells and entire organisms and discuss the mechanisms by which disruption of the PN, and related stress response pathways, contributes to the initiation and progression of disease. We explore emerging evidence that disease susceptibility arises from early changes in the composition and activity of the PN and propose that a more complete understanding of the temporal and spatial properties of the PN will enhance our ability to develop effective treatments for protein conformational diseases.
Collapse
Affiliation(s)
- Johnathan Labbadia
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208;
| | | |
Collapse
|
128
|
Tan J, Tan S, Zheng H, Liu M, Chen G, Zhang H, Wang K, Tan S, Zhou J, Xiao XZ. HSF1 functions as a transcription regulator for Dp71 expression. Cell Stress Chaperones 2015; 20:371-9. [PMID: 25430510 PMCID: PMC4326382 DOI: 10.1007/s12192-014-0558-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 11/25/2022] Open
Abstract
Heat shock factor 1 (HSF1) is one of the most important transcriptional molecules in the heat shock process; however, HSF1 can also regulate the expression of other proteins. Dystrophin Dp71 is one of the most widely expressed isoforms of the dystrophin gene family. In our experiments, we showed for the first time that HSF1 can function as a transcriptional factor for endogenous Dp71 expression in vivo and in vitro. We demonstrated that the messenger RNA (mRNA) and protein expression of Dp71 were significantly reduced in HSF1-knockout mice compared with wild-type mice in brain, lung, liver, spleen, and kidney. Overexpression of HSF1 significantly enhanced the mRNA and protein expression of Dp71 in HeLa cells. Inhibiting the expression of HSF1 in HeLa cells significantly reduced the expression of Dp71. By use of the EMSA technique, the chromatin immunoprecipitation assay, and the luciferase reporter system, we demonstrated that HSF1 can directly bind the HSE in the Dp71 promoter region. We concluded from our data that HSF1 functions as a transcriptional regulator of Dp71 expression.
Collapse
Affiliation(s)
- Jin Tan
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Sichuang Tan
- />Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital, 139# Ren Ming Road, Changsha, Hunan 410011 People’s Republic of China
| | - Hexin Zheng
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- />Key Laboratory of Genetics and Birth Health of Hunan Province, Family Planning, Institute of Hunan Province, Changsha, 410126 China
| | - Meidong Liu
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Guangwen Chen
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Huali Zhang
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Kangkai Wang
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Sipin Tan
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- />Molecular and Cell Experimental Center, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 People’s Republic of China
| | - Jiang Zhou
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Xian-zhong Xiao
- />Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110# Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| |
Collapse
|
129
|
The chaperone balance hypothesis: the importance of the extracellular to intracellular HSP70 ratio to inflammation-driven type 2 diabetes, the effect of exercise, and the implications for clinical management. Mediators Inflamm 2015; 2015:249205. [PMID: 25814786 PMCID: PMC4357135 DOI: 10.1155/2015/249205] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/12/2015] [Indexed: 01/01/2023] Open
Abstract
Recent evidence shows divergence between the concentrations of extracellular 70 kDa heat shock protein [eHSP70] and its intracellular concentrations [iHSP70] in people with type 2 diabetes (T2DM). A vital aspect regarding HSP70 physiology is its versatility to induce antagonistic actions, depending on the location of the protein. For example, iHSP70 exerts a powerful anti-inflammatory effect, while eHSP70 activates proinflammatory pathways. Increased eHSP70 is associated with inflammatory and oxidative stress conditions, whereas decreased iHSP70 levels are related to insulin resistance in skeletal muscle. Serum eHSP70 concentrations are positively correlated with markers of inflammation, such as C-reactive protein, monocyte count, and TNF-α, while strategies to enhance iHSP70 (e.g., heat treatment, chemical HSP70 inducers or coinducers, and physical exercise) are capable of reducing the inflammatory profile and the insulin resistance state. Here, we present recent findings suggesting that imbalances in the HSP70 status, described by the [eHSP70]/[iHSP70] ratio, may be determinant to trigger a chronic proinflammatory state that leads to insulin resistance and T2DM development. This led us to hypothesize that changes in this ratio value could be used as a biomarker for the management of the inflammatory response in insulin resistance and diabetes.
Collapse
|
130
|
Affiliation(s)
- Robert J. Collier
- School of Animal and Comparative Biomedical Science, University of Arizona, Tucson, Arizona 85719;
| | - Kifle G. Gebremedhin
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853
| |
Collapse
|
131
|
Kaitsuka T, Matsushita M. Regulation of translation factor EEF1D gene function by alternative splicing. Int J Mol Sci 2015; 16:3970-9. [PMID: 25686034 PMCID: PMC4346937 DOI: 10.3390/ijms16023970] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/04/2015] [Indexed: 11/16/2022] Open
Abstract
Alternative splicing is an exquisite mechanism that allows one coding gene to have multiple functions. The alternative splicing machinery is necessary for proper development, differentiation and stress responses in a variety of organisms, and disruption of this machinery is often implicated in human diseases. Previously, we discovered a long form of eukaryotic elongation factor 1Bδ (eEF1Bδ; this long-form eEF1Bδ results from alternative splicing of EEF1D transcripts and regulates the cellular stress response by transcriptional activation, not translational enhancement, of heat-shock responsive genes. In this review, we discuss the molecular function of EEF1D alternative splicing products and the estimated implication of human diseases.
Collapse
Affiliation(s)
- Taku Kaitsuka
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan.
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| |
Collapse
|
132
|
Atarod S, Turner B, Pearce KF, Ahmed SS, Norden J, Bogunia-Kubik K, Wang XN, Collin M, Dickinson AM. Elevated level of HSPA1L mRNA correlates with graft-versus-host disease. Transpl Immunol 2015; 32:188-94. [PMID: 25680846 DOI: 10.1016/j.trim.2015.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
Graft-versus-host disease (GVHD) can be a fatal complication of allogeneic stem cell transplantation (allo-HSCT). GVHD can be classified as acute (aGVHD: up to 100 days) or chronic (cGVHD: after 100 days) based on the time-point of disease occurrence. At present there are a limited number of biomarkers available for use in the clinic. Thus, the aim of this research was to evaluate the biomarker potential of the extensively studied Heat Shock Protein 70 family members (HSPA1A/HSPA1B and HSPA1L) at the messenger RNA (mRNA) level in acute and cGVHD patient cohorts. In the skin biopsies, HSPA1L mRNA expression was lower in patients with severe aGVHD (grades II-III) when compared to those with none or low grade aGVHD (grades 0-I) and normal controls. In whole blood, HSPA1L mRNA expression level was significantly (p = 0.008) up-regulated at 28 days post-transplant in cGVHD patients with a significant area under the curve (AUC = 0.773). In addition, HSPA1B expression in whole blood was significantly higher at 3 months post-transplant in both the aGVHD grade II-III (p = 0.012) and cGVHD (p = 0.027) patients. Our initial results in this small cohort show that quantifying HSPA1L mRNA expression in the whole blood of allo-HSCT patients at day 28 post-allo-HSCT may be a useful predictive biomarker for cGVHD.
Collapse
Affiliation(s)
- Sadaf Atarod
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Brie Turner
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Kim Frances Pearce
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Shaheda S Ahmed
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Jean Norden
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Katarzyna Bogunia-Kubik
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.
| | - Xiao-nong Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Matthew Collin
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Anne Mary Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
133
|
Dai C. Metabolic stressors disrupt proteome homeostasis to suppress malignancy. Mol Cell Oncol 2015; 2. [PMID: 26973866 PMCID: PMC4786015 DOI: 10.1080/23723556.2014.1002718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
Within tumor cells the heat shock factor 1 (HSF1)-mediated stress response is constitutively mobilized to counter persistent proteotoxic stress, hence sustaining their fragile proteome homeostasis and supporting robust malignant phenotypes. Intriguingly, our new studies reveal that metabolic stressors, such as metformin, inactivate HSF1 and provoke proteomic chaos, thereby impeding tumorigenesis.
Collapse
Affiliation(s)
- Chengkai Dai
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
134
|
Bridges TM, Scheraga RG, Tulapurkar ME, Suffredini D, Liggett SB, Ramarathnam A, Potla R, Singh IS, Hasday JD. Polymorphisms in human heat shock factor-1 and analysis of potential biological consequences. Cell Stress Chaperones 2015; 20:47-59. [PMID: 25023647 PMCID: PMC4255257 DOI: 10.1007/s12192-014-0524-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 06/24/2014] [Accepted: 06/30/2014] [Indexed: 11/30/2022] Open
Abstract
The stress-activated transcription factor, heat shock factor-1 (HSF1), regulates many genes including cytoprotective heat shock proteins (HSPs). We hypothesized that polymorphisms in HSF1 may alter the level or function of HSF1 protein accounting for interindividual viability in disease susceptibility or prognosis. We searched for exomic variants in HSF1 by querying human genome databases and directly sequencing DNA from 80 anonymous genomic DNA samples. Overall, HSF1 sequence was highly conserved, with no common variations. We found 31 validated deviations from a reference sequence in the dbSNP database and an additional 5 novel variants by sequencing, with allele frequencies that were 0.06 or less. Of these 36, 2 were in 5'-untranslated region (5'UTR), 10 in 3'UTR, and 24 in the coding region. The potential effects of 5'UTR on secondary structure, protein structure/function, and 3'UTR targets of microRNAs were analyzed using RNAFold, PolyPhen-2, SIFT, and MicroSNiper. One of the 5'UTR variants was predicted to strengthen secondary structure. Eight of 3'UTR variants were predicted to modify microRNA target sequences. Eight of the coding region variants were predicted to modify HSF1 structure/function. Reducing HSF1 levels in A549 cells using short hairpin RNA (shRNA) increased sensitivity to heat-induced killing demonstrating the impact that genetic variants that reduce HSF1 levels might have. Using the pmirGLO expression system, we found that the wild-type HSF1 3'UTR suppressed translation of a firefly luciferase reporter plasmid by 65 %. Introducing two of four 3'UTR single nucleotide polymorphisms (SNPs) increased HSF1 3'UTR translational suppression by 27-44 % compared with the wild-type HSF1 3'UTR sequence while a third SNP reduced suppression by 25 %. HSF1 variants may alter HSF1 protein levels or function with potential effects on cell functions, including sensitivity to stress.
Collapse
Affiliation(s)
- Tiffany M. Bridges
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Rachel G. Scheraga
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Mohan E. Tulapurkar
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Dante Suffredini
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Stephen B. Liggett
- />Departments of Medicine and Molecular Physiology and Pharmacology, University of South Florida, Tampa, FL 22612 USA
| | - Aparna Ramarathnam
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Ratnakar Potla
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
| | - Ishwar S. Singh
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
- />Medicine and Research services of the Baltimore VA Medical Center, Baltimore, MD 21201 USA
| | - Jeffrey D. Hasday
- />Pulmonary and Critical Care Medicine Division, Department of Medicine, University of Maryland School of Medicine, 110 S. Paca St. 2nd floor, Baltimore, Maryland USA
- />Medicine and Research services of the Baltimore VA Medical Center, Baltimore, MD 21201 USA
| |
Collapse
|
135
|
Luan Q, Jin L, Jiang CC, Tay KH, Lai F, Liu XY, Liu YL, Guo ST, Li CY, Yan XG, Tseng HY, Zhang XD. RIPK1 regulates survival of human melanoma cells upon endoplasmic reticulum stress through autophagy. Autophagy 2015; 11:975-94. [PMID: 26018731 PMCID: PMC4590596 DOI: 10.1080/15548627.2015.1049800] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 04/28/2015] [Accepted: 05/05/2015] [Indexed: 12/11/2022] Open
Abstract
Although RIPK1 (receptor [TNFRSF]-interacting protein kinase 1) is emerging as a critical determinant of cell fate in response to cellular stress resulting from activation of death receptors and DNA damage, its potential role in cell response to endoplasmic reticulum (ER) stress remains undefined. Here we report that RIPK1 functions as an important prosurvival mechanism in melanoma cells undergoing pharmacological ER stress induced by tunicamycin (TM) or thapsigargin (TG) through activation of autophagy. While treatment with TM or TG upregulated RIPK1 and triggered autophagy in melanoma cells, knockdown of RIPK1 inhibited autophagy and rendered the cells sensitive to killing by TM or TG, recapitulating the effect of inhibition of autophagy. Consistently, overexpression of RIPK1 enhanced induction of autophagy and conferred resistance of melanoma cells to TM- or TG-induced cell death. Activation of MAPK8/JNK1 or MAPK9/JNK2, which phosphorylated BCL2L11/BIM leading to its dissociation from BECN1/Beclin 1, was involved in TM- or TG-induced, RIPK1-mediated activation of autophagy; whereas, activation of the transcription factor HSF1 (heat shock factor protein 1) downstream of the ERN1/IRE1-XBP1 axis of the unfolded protein response was responsible for the increase in RIPK1 in melanoma cells undergoing pharmacological ER stress. Collectively, these results identify upregulation of RIPK1 as an important resistance mechanism of melanoma cells to TM- or TG-induced ER stress by protecting against cell death through activation of autophagy, and suggest that targeting the autophagy-activating mechanism of RIPK1 may be a useful strategy to enhance sensitivity of melanoma cells to therapeutic agents that induce ER stress.
Collapse
Key Words
- 3-MA, 3-methyladenine
- AMPK, AMP-activated protein kinase
- ATF6, activating transcription factor 6
- Baf A1, bafilomycin A1
- CAMKK2, calcium/calmodulin-dependent protein kinase kinase 2: β
- EIF2AK3/PERK, eukaryotic translation initiation factor 2-α kinase 3
- ER, endoplasmic reticulum
- ERN1/IRE1, endoplasmic reticulum to nucleus signaling 1
- HSF1, heat shock transcription factor 1
- HSPA5, heat shock 70kDa protein 5 (glucose-regulated protein: 78kDa)
- MAP2K1/MEK1, mitogen-activated protein kinase kinase 1
- MAPK, mitogen-activated protein kinase
- MAPK1/ERK2, mitogen-activated protein kinase 1
- MAPK11/p38β, mitogen-activated protein kinase 11
- MAPK12/p38γ, mitogen-activated protein kinase 12
- MAPK13/p38δ, mitogen-activated protein kinase 13
- MAPK14/p38α, mitogen-activated protein kinase 14
- MAPK3/ERK1, mitogen-activated protein kinase 3
- MAPK8/JNK1, mitogen-activated protein kinase 8
- MAPK9/JNK2, mitogen-activated protein kinase 9
- NFKB1, nuclear factor of kappa light polypeptide gene enhancer in B-cells 1
- PRKAA1, protein kinase AMP-activated: α 1 catalytic subunit
- RIPK1
- RIPK1, receptor (TNFRSF)-interacting protein kinase 1
- SQSTM1/p62, sequestosome 1
- TG, thapsigargin
- TM, tunicamycin
- TNFRSF1A/TNFR1, tumor necrosis factor receptor superfamily: member 1A
- UPR, unfolded protein response
- XBP1, x-box binding protein 1
- autophagy
- cell death
- endoplasmic reticulum stress
- melanoma
Collapse
Affiliation(s)
- Qi Luan
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
- Department of Dermatology; Xijing Hospital; Fourth Military Medical University; Xi'an; China
| | - Lei Jin
- School of Medicine and Public Health; University of Newcastle; NSW, Australia
| | - Chen Chen Jiang
- School of Medicine and Public Health; University of Newcastle; NSW, Australia
| | - Kwang Hong Tay
- School of Medicine and Public Health; University of Newcastle; NSW, Australia
| | - Fritz Lai
- School of Medicine and Public Health; University of Newcastle; NSW, Australia
| | - Xiao Ying Liu
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| | - Yi Lun Liu
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| | - Su Tang Guo
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| | - Chun Ying Li
- Department of Dermatology; Xijing Hospital; Fourth Military Medical University; Xi'an; China
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| | - Hsin-Yi Tseng
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy; University of Newcastle; NSW, Australia
| |
Collapse
|
136
|
Abstract
The heat shock response (HSR) is an ancient and highly conserved process that is essential for coping with environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review the phylogenetically conserved mechanisms that regulate fever and discuss the effects that febrile-range temperatures have on multiple biological processes involved in host defense and cell death and survival, including the HSR and its implications for patients with severe sepsis, trauma, and other acute systemic inflammatory states. Heat shock factor-1, a heat-induced transcriptional enhancer is not only the central regulator of the HSR but also regulates expression of pivotal cytokines and early response genes. Febrile-range temperatures exert additional immunomodulatory effects by activating mitogen-activated protein kinase cascades and accelerating apoptosis in some cell types. This results in accelerated pathogen clearance, but increased collateral tissue injury, thus the net effect of exposure to febrile range temperature depends in part on the site and nature of the pathologic process and the specific treatment provided.
Collapse
Affiliation(s)
- Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore V.A. Medical Center, Baltimore, Maryland
| | | | | |
Collapse
|
137
|
Dai S, Tang Z, Cao J, Zhou W, Li H, Sampson S, Dai C. Suppression of the HSF1-mediated proteotoxic stress response by the metabolic stress sensor AMPK. EMBO J 2014; 34:275-93. [PMID: 25425574 PMCID: PMC4339117 DOI: 10.15252/embj.201489062] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Numerous extrinsic and intrinsic insults trigger the HSF1-mediated proteotoxic stress response (PSR), an ancient transcriptional program that is essential to proteostasis and survival under such conditions. In contrast to its well-recognized mobilization by proteotoxic stress, little is known about how this powerful adaptive mechanism reacts to other stresses. Surprisingly, we discovered that metabolic stress suppresses the PSR. This suppression is largely mediated through the central metabolic sensor AMPK, which physically interacts with and phosphorylates HSF1 at Ser121. Through AMPK activation, metabolic stress represses HSF1, rendering cells vulnerable to proteotoxic stress. Conversely, proteotoxic stress inactivates AMPK and thereby interferes with the metabolic stress response. Importantly, metformin, a metabolic stressor and popular anti-diabetic drug, inactivates HSF1 and provokes proteotoxic stress within tumor cells, thereby impeding tumor growth. Thus, these findings uncover a novel interplay between the metabolic stress sensor AMPK and the proteotoxic stress sensor HSF1 that profoundly impacts stress resistance, proteostasis, and malignant growth.
Collapse
Affiliation(s)
- Siyuan Dai
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Zijian Tang
- The Jackson Laboratory, Bar Harbor, ME, USA Graduate Programs, Department of Molecular & Biomedical Sciences, The University of Maine, Orono, ME, USA
| | - Junyue Cao
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Wei Zhou
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Huawen Li
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | |
Collapse
|
138
|
Soares MP, Gozzelino R, Weis S. Tissue damage control in disease tolerance. Trends Immunol 2014; 35:483-94. [PMID: 25182198 DOI: 10.1016/j.it.2014.08.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/09/2014] [Accepted: 08/11/2014] [Indexed: 02/07/2023]
Abstract
Immune-driven resistance mechanisms are the prevailing host defense strategy against infection. By contrast, disease tolerance mechanisms limit disease severity by preventing tissue damage or ameliorating tissue function without interfering with pathogen load. We propose here that tissue damage control underlies many of the protective effects of disease tolerance. We explore the mechanisms of cellular adaptation that underlie tissue damage control in response to infection as well as sterile inflammation, integrating both stress and damage responses. Finally, we discuss the potential impact of targeting these mechanisms in the treatment of disease.
Collapse
|
139
|
Ishida K, Varrecchia M, Knudsen GM, Jolly ER. Immunolocalization of anti-hsf1 to the acetabular glands of infectious schistosomes suggests a non-transcriptional function for this transcriptional activator. PLoS Negl Trop Dis 2014; 8:e3051. [PMID: 25078989 PMCID: PMC4117452 DOI: 10.1371/journal.pntd.0003051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/16/2014] [Indexed: 11/19/2022] Open
Abstract
Schistosomiasis is a chronically debilitating disease caused by parasitic worms of the genus Schistosoma, and it is a global problem affecting over 240 million people. Little is known about the regulatory proteins and mechanisms that control schistosome host invasion, gene expression, and development. Schistosome larvae, cercariae, are transiently free-swimming organisms and infectious to man. Cercariae penetrate human host skin directly using proteases that degrade skin connective tissue. These proteases are secreted from anucleate acetabular glands that contain many proteins, including heat shock proteins. Heat shock transcription factors are strongly conserved activators that play crucial roles in the maintenance of cell homeostasis by transcriptionally regulating heat shock protein expression. In this study, we clone and characterize the schistosome Heat shock factor 1 gene (SmHSF1). We verify its ability to activate transcription using a modified yeast one-hybrid system, and we show that it can bind to the heat shock binding element (HSE) consensus DNA sequence. Our quantitative RT-PCR analysis shows that SmHSF1 is expressed throughout several life-cycle stages from sporocyst to adult worm. Interestingly, using immunohistochemistry, a polyclonal antibody raised against an Hsf1-peptide demonstrates a novel localization for this conserved, stress-modulating activator. Our analysis suggests that schistosome Heat shock factor 1 may be localized to the acetabular glands of infective cercariae. Schistosome parasites are the cause of human schistosomiasis and infect more than 240 million people worldwide. Schistosome larvae, termed cercariae, are a free-swimming mobile developmental stage responsible for host infection. These larvae produce enzymes that degrade human skin, allowing them to pass into the human host. After invasion, they continue to evade the immune system and develop into adult worms. The transition from free-swimming larvae in freshwater to invasion into a warm-blooded saline environment requires that the parasite regulate genes to adapt to these changes. Heat shock factor 1 is a well-characterized activator of stress and heat response that functions in cellular nuclei. Using immunohistochemistry, we observed non-nuclear localization for anti-Heat shock factor 1 signal in the secretory glands necessary for the invasive function of schistosome larvae. This observation expands the potential mechanistic roles for Heat shock factor 1 and may aid in our understanding of schistosome host invasion and early development.
Collapse
Affiliation(s)
- Kenji Ishida
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Melissa Varrecchia
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Giselle M. Knudsen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Emmitt R. Jolly
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
140
|
Lin Y, Cheng Y, Jin J, Jin X, Jiang H, Yan H, Cheng B. Genome duplication and gene loss affect the evolution of heat shock transcription factor genes in legumes. PLoS One 2014; 9:e102825. [PMID: 25047803 PMCID: PMC4105503 DOI: 10.1371/journal.pone.0102825] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 06/25/2014] [Indexed: 11/18/2022] Open
Abstract
Whole-genome duplication events (polyploidy events) and gene loss events have played important roles in the evolution of legumes. Here we show that the vast majority of Hsf gene duplications resulted from whole genome duplication events rather than tandem duplication, and significant differences in gene retention exist between species. By searching for intraspecies gene colinearity (microsynteny) and dating the age distributions of duplicated genes, we found that genome duplications accounted for 42 of 46 Hsf-containing segments in Glycine max, while paired segments were rarely identified in Lotus japonicas, Medicago truncatula and Cajanus cajan. However, by comparing interspecies microsynteny, we determined that the great majority of Hsf-containing segments in Lotus japonicas, Medicago truncatula and Cajanus cajan show extensive conservation with the duplicated regions of Glycine max. These segments formed 17 groups of orthologous segments. These results suggest that these regions shared ancient genome duplication with Hsf genes in Glycine max, but more than half of the copies of these genes were lost. On the other hand, the Glycine max Hsf gene family retained approximately 75% and 84% of duplicated genes produced from the ancient genome duplication and recent Glycine-specific genome duplication, respectively. Continuous purifying selection has played a key role in the maintenance of Hsf genes in Glycine max. Expression analysis of the Hsf genes in Lotus japonicus revealed their putative involvement in multiple tissue-/developmental stages and responses to various abiotic stimuli. This study traces the evolution of Hsf genes in legume species and demonstrates that the rates of gene gain and loss are far from equilibrium in different species.
Collapse
Affiliation(s)
- Yongxiang Lin
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
- Crop Research Institute, Anhui Academy of Agricultural Sciences, Hefei, Anhui, China
| | - Ying Cheng
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| | - Jing Jin
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| | - Xiaolei Jin
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| | - Haiyang Jiang
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| | - Hanwei Yan
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| | - Beijiu Cheng
- Key Lab of Crop Biology of Anhui Province, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, China
| |
Collapse
|
141
|
Ferat-Osorio E, Sánchez-Anaya A, Gutiérrez-Mendoza M, Boscó-Gárate I, Wong-Baeza I, Pastelin-Palacios R, Pedraza-Alva G, Bonifaz LC, Cortés-Reynosa P, Pérez-Salazar E, Arriaga-Pizano L, López-Macías C, Rosenstein Y, Isibasi A. Heat shock protein 70 down-regulates the production of toll-like receptor-induced pro-inflammatory cytokines by a heat shock factor-1/constitutive heat shock element-binding factor-dependent mechanism. JOURNAL OF INFLAMMATION-LONDON 2014; 11:19. [PMID: 25053922 PMCID: PMC4105516 DOI: 10.1186/1476-9255-11-19] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 07/05/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Heat shock protein 70 (Hsp70) is an intracellular chaperone protein with regulatory and cytoprotective functions. Hsp70 can also be found in the extracellular milieu, as a result of active secretion or passive release from damaged cells. The role of extracellular Hsp70 is not fully understood. Some studies report that it activates monocytes, macrophages and dendritic cells through innate immune receptors (such as Toll-like receptors, TLRs), while others report that Hsp70 is a negative regulator of the inflammatory response. In order to address this apparent inconsistency, in this study we evaluated the response of human monocytes to a highly purified recombinant Hsp70. METHODS Human peripheral blood monocytes were stimulated with Hsp70, alone or in combination with TLR agonists. Cytokines were quantified in culture supernatants, their mRNAs were measured by RT-PCR, and the binding of transcription factors was evaluated by electrophoretic mobility shift assay (EMSA). Kruskal-Wallis test or one-way or two-way ANOVA were used to analyze the data. RESULTS The addition of Hsp70 to TLR-activated monocytes down-regulated TNF-α as well as IL-6 levels. This effect was independent of a physical interaction between Hsp70 and TLR agonists; instead it resulted of changes at the TNF-α gene expression level. The decrease in TNF-α expression correlated with the binding of HSF-1 (heat shock transcription factor 1, a transcription factor activated in response to Hsp70) and CHBF (constitutive HSE-binding factor) to the TNF-α gene promoter. CONCLUSION Extracellular Hsp70 negatively regulates the production of pro-inflammatory cytokines of monocytes exposed to TLR agonists and contributes to dampen the inflammatory response.
Collapse
Affiliation(s)
- Eduardo Ferat-Osorio
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México ; Servicio de Cirugía Gastrointestinal, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Aldair Sánchez-Anaya
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Mireille Gutiérrez-Mendoza
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Ilka Boscó-Gárate
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Isabel Wong-Baeza
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México ; Departamento de Inmunología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, México D.F., México
| | | | - Gustavo Pedraza-Alva
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Pedro Cortés-Reynosa
- Departamento de Biología Celular, (CINVESTAV) Instituto Politécnico Nacional, Centro de Investigación y Estudios Avanzados, México D.F., México
| | - Eduardo Pérez-Salazar
- Departamento de Biología Celular, (CINVESTAV) Instituto Politécnico Nacional, Centro de Investigación y Estudios Avanzados, México D.F., México
| | - Lourdes Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México ; Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, Cuernavaca Mor. 62210, México
| | - Armando Isibasi
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México ; Coordinación de Investigación en Salud, Piso 4 Bloque B Unidad de Congresos Centro Médico Nacional Siglo XXI, Av. Cuauhtémoc 330, Col. Doctores, México D.F. CP 06020, México
| |
Collapse
|
142
|
Engerud H, Tangen IL, Berg A, Kusonmano K, Halle MK, Oyan AM, Kalland KH, Stefansson I, Trovik J, Salvesen HB, Krakstad C. High level of HSF1 associates with aggressive endometrial carcinoma and suggests potential for HSP90 inhibitors. Br J Cancer 2014; 111:78-84. [PMID: 24853175 PMCID: PMC4090731 DOI: 10.1038/bjc.2014.262] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/11/2014] [Accepted: 04/23/2014] [Indexed: 11/12/2022] Open
Abstract
Background: Recent identification of a specific role of HSF1 in cancer progression has led to new relevance of HSF1 as both a prognostic and a predictive marker. The role of HSF1 in endometrial cancer has so far been unexplored. Methods: A total of 823 lesions from endometrial carcinoma precursors, primary tumours and metastases were prospectively collected and explored for HSF1 protein expression in relation to established markers for aggressive disease and survival. Transcriptional alterations related to HSF1 protein level were investigated by microarray analysis for 224 freshly frozen samples in parallel. Results: High expression of HSF1 protein in endometrial carcinoma is significantly associated with aggressive disease and poor survival (all P-values ⩽0.02), also among ERα-positive patients presumed to have good prognosis. The HSF1-related gene signatures increase during disease progression and were also found to have prognostic value. Gene expression analyses identified HSP90 inhibition as a potential novel therapeutic approach for cases with high protein expression of HSF1. Conclusions: We demonstrate for the first time in endometrial cancer that high expression of HSF1 and measures for transcriptional activation of HSF1 associate with poor outcome and disease progression. The HSP90 inhibitors are suggested as new targeted therapeutics for patients with high HSF1 levels in tumour in particular.
Collapse
Affiliation(s)
- H Engerud
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - I L Tangen
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - A Berg
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - K Kusonmano
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway [3] Computational Biology Unit, University of Bergen, Bergen, Norway
| | - M K Halle
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - A M Oyan
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - K H Kalland
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - I Stefansson
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - J Trovik
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - H B Salvesen
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - C Krakstad
- 1] Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway [2] Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
143
|
Sreedharan R, Chen S, Miller M, Haribhai D, Williams CB, Van Why SK. Mice with an absent stress response are protected against ischemic renal injury. Kidney Int 2014; 86:515-24. [PMID: 24805105 PMCID: PMC4149847 DOI: 10.1038/ki.2014.73] [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: 03/12/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 01/18/2023]
Abstract
Inducible heat shock proteins (HSP), regulated by heat shock factor-1 (HSF-1), protect against renal cell injury in vitro. To determine whether HSPs ameliorate ischemic renal injury in vivo, HSF-1functional knock-out mice (HSF-KO) were compared with wild-type mice following bilateral ischemic renal injury. Following injury, the kidneys of wild-type mice had the expected induction of HSP70 and HSP25; a response absent in the kidneys of HSF-KO mice. Baseline serum creatinine was equivalent between strains. Serum creatinines at 24 hours reflow in HSF-KO mice were significantly lower than in the wild-type. Histology showed similar tubule injury in both strains after ischemic renal injury but increased medullary vascular congestion in wild-type compared with HSF-KO mice. Flow-cytometry of mononuclear cells isolated from kidneys showed no difference between strains in the number of CD4+ and CD8+ T cells in sham operated animals. At 1 hour of reflow, CD4+ and CD8+ cells were doubled in the kidneys of wild type but not HSF-KO mice. Foxp3+ T regulatory cells were significantly more abundant in the kidneys of sham-operated HSF-KO than wild-type mice. Suppression of CD25+Foxp3+ cells in HSF-KO kidneys with the anti-CD25 antibody PC61 reversed the protection against ischemic renal injury. Thus, HSF-KO mice are protected from ischemic renal injury by a mechanism that depends on an increase in the T regulatory cells in the kidney associated with altered T cell infiltration early in reflow. Hence, stress response activation may contribute to early injury by facilitating T cell infiltration into ischemic kidney.
Collapse
Affiliation(s)
- Rajasree Sreedharan
- Division of Nephrology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Shaoying Chen
- Division of Nephrology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Melody Miller
- Division of Nephrology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Dipica Haribhai
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Calvin B Williams
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Scott K Van Why
- Division of Nephrology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| |
Collapse
|
144
|
Inhibiting heat shock factor 1 in human cancer cells with a potent RNA aptamer. PLoS One 2014; 9:e96330. [PMID: 24800749 PMCID: PMC4011729 DOI: 10.1371/journal.pone.0096330] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 04/04/2014] [Indexed: 11/19/2022] Open
Abstract
Heat shock factor 1 (HSF1) is a master regulator that coordinates chaperone protein expression to enhance cellular survival in the face of heat stress. In cancer cells, HSF1 drives a transcriptional program distinct from heat shock to promote metastasis and cell survival. Its strong association with the malignant phenotype implies that HSF1 antagonists may have general and effective utilities in cancer therapy. For this purpose, we had identified an avid RNA aptamer for HSF1 that is portable among different model organisms. Extending our previous work in yeast and Drosophila, here we report the activity of this aptamer in human cancer cell lines. When delivered into cells using a synthetic gene and strong promoter, this aptamer was able to prevent HSF1 from binding to its DNA regulation elements. At the cellular level, expression of this aptamer induced apoptosis and abolished the colony-forming capability of cancer cells. At the molecular level, it reduced chaperones and attenuated the activation of the MAPK signaling pathway. Collectively, these data demonstrate the advantage of aptamers in drug target validation and support the hypothesis that HSF1 DNA binding activity is a potential target for controlling oncogenic transformation and neoplastic growth.
Collapse
|
145
|
HSF-1 is Involved in Attenuating the Release of Inflammatory Cytokines Induced by LPS Through Regulating Autophagy. Shock 2014; 41:449-53. [DOI: 10.1097/shk.0000000000000118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
146
|
Roles of heat shock factor 1 in neuronal response to fetal environmental risks and its relevance to brain disorders. Neuron 2014; 82:560-72. [PMID: 24726381 DOI: 10.1016/j.neuron.2014.03.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 12/25/2022]
Abstract
Prenatal exposure of the developing brain to various environmental challenges increases susceptibility to late onset of neuropsychiatric dysfunction; still, the underlying mechanisms remain obscure. Here we show that exposure of embryos to a variety of environmental factors such as alcohol, methylmercury, and maternal seizure activates HSF1 in cerebral cortical cells. Furthermore, Hsf1 deficiency in the mouse cortex exposed in utero to subthreshold levels of these challenges causes structural abnormalities and increases seizure susceptibility after birth. In addition, we found that human neural progenitor cells differentiated from induced pluripotent stem cells derived from schizophrenia patients show higher variability in the levels of HSF1 activation induced by environmental challenges compared to controls. We propose that HSF1 plays a crucial role in the response of brain cells to prenatal environmental insults and may be a key component in the pathogenesis of late-onset neuropsychiatric disorders.
Collapse
|
147
|
Chai RC, Kouspou MM, Lang BJ, Nguyen CH, van der Kraan AGJ, Vieusseux JL, Lim RC, Gillespie MT, Benjamin IJ, Quinn JMW, Price JT. Molecular stress-inducing compounds increase osteoclast formation in a heat shock factor 1 protein-dependent manner. J Biol Chem 2014; 289:13602-14. [PMID: 24692538 DOI: 10.1074/jbc.m113.530626] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Many anticancer therapeutic agents cause bone loss, which increases the risk of fractures that severely reduce quality of life. Thus, in drug development, it is critical to identify and understand such effects. Anticancer therapeutic and HSP90 inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) causes bone loss by increasing osteoclast formation, but the mechanism underlying this is not understood. 17-AAG activates heat shock factor 1 (Hsf1), the master transcriptional regulator of heat shock/cell stress responses, which may be involved in this negative action of 17-AAG upon bone. Using mouse bone marrow and RAW264.7 osteoclast differentiation models we found that HSP90 inhibitors that induced a heat shock response also enhanced osteoclast formation, whereas HSP90 inhibitors that did not (including coumermycin A1 and novobiocin) did not affect osteoclast formation. Pharmacological inhibition or shRNAmir knockdown of Hsf1 in RAW264.7 cells as well as the use of Hsf1 null mouse bone marrow cells demonstrated that 17-AAG-enhanced osteoclast formation was Hsf1-dependent. Moreover, ectopic overexpression of Hsf1 enhanced 17-AAG effects upon osteoclast formation. Consistent with these findings, protein levels of the essential osteoclast transcription factor microphthalmia-associated transcription factor were increased by 17-AAG in an Hsf1-dependent manner. In addition to HSP90 inhibitors, we also identified that other agents that induced cellular stress, such as ethanol, doxorubicin, and methotrexate, also directly increased osteoclast formation, potentially in an Hsf1-dependent manner. These results, therefore, indicate that cellular stress can enhance osteoclast differentiation via Hsf1-dependent mechanisms and may significantly contribute to pathological and therapeutic related bone loss.
Collapse
Affiliation(s)
- Ryan C Chai
- From the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Carnemolla A, Labbadia JP, Lazell H, Neueder A, Moussaoui S, Bates GP. Contesting the dogma of an age-related heat shock response impairment: implications for cardiac-specific age-related disorders. Hum Mol Genet 2014; 23:3641-56. [PMID: 24556212 PMCID: PMC4065144 DOI: 10.1093/hmg/ddu073] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ageing is associated with the reduced performance of physiological processes and has been proposed as a major risk factor for disease. An age-related decline in stress response pathways has been widely documented in lower organisms. In particular, the heat shock response (HSR) becomes severely compromised with age in Caenorhabditis elegans. However, a comprehensive analysis of the consequences of ageing on the HSR in higher organisms has not been documented. We used both HS and inhibition of HSP90 to induce the HSR in wild-type mice at 3 and 22 months of age to investigate the extent to which different brain regions, and peripheral tissues can sustain HSF1 activity and HS protein (HSP) expression with age. Using chromatin immunoprecipitation, quantitative reverse transcription polymerase chain reaction, western blotting and enzyme linked immunosorbent assay (ELISA), we were unable to detect a difference in the level or kinetics of HSP expression between young and old mice in all brain regions. In contrast, we did observe an age-related reduction in chaperone levels and HSR-related proteins in the heart. This could result in a decrease in the protein folding capacity of old hearts with implications for age-related cardiac disorders.
Collapse
Affiliation(s)
- Alisia Carnemolla
- Department Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hosptial, Great Maze Pond, London SE1 9RT, UK
| | - John P Labbadia
- Department Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hosptial, Great Maze Pond, London SE1 9RT, UK
| | - Hayley Lazell
- Department Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hosptial, Great Maze Pond, London SE1 9RT, UK
| | - Andreas Neueder
- Department Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hosptial, Great Maze Pond, London SE1 9RT, UK
| | - Saliha Moussaoui
- Novartis Institute for Biomedical Research, Neuroscience Discovery, Basel CH-4002, Switzerland
| | - Gillian P Bates
- Department Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hosptial, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
149
|
Barber RC, Maass DL, White DJ, Horton JW, Wolf SE, Minei JP, Zang QS. Deficiency in Heat Shock Factor 1 (HSF-1) Expression Exacerbates Sepsis-induced Inflammation and Cardiac Dysfunction. ACTA ACUST UNITED AC 2014; 1. [PMID: 30701190 DOI: 10.15226/2376-4570/1/1/00103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the present study, we investigated whether absence of heat shock factor 1 (HSF-1) and inability to increase myocardial expression of heat shock proteins alter septic responses of inflammatory cytokines and myocardial contractility. HSF-1 knockout (hsf -/-) mice and wild type litter mates underwent a sterile (lipopolysaccharide; LPS) or infectious (Streptococcus pneumoniae or Klebsiella pneumoniae) septic challenge. Production of cytokines, TNF, IL-1β, IL-6 and IL-10, in the blood and from cardiomyocytes was exaggerated in the hsf -/- mice compared to responses measured in wild type mice given an identical septic challenge. This enhanced compartmentalized myocardial inflammation was associated with significantly decreased cardiac contraction and diminished relaxation in the hsf -/- mice. However, lacking HSF-1 expression did not affect intracellular calcium and sodium responses in cardiomyocytes isolated from septic challenged mice, suggesting that ion loading was not a major or sustaining cause of the greater myocardial contractile defects in hsf -/- mice. In conclusion, our data indicated that HSF-1 and downstream heat shock proteins are essential components to support cardiac function in sepsis. Further studies are warranted to further define the precise mechanisms of HSF-1 mediated cardiac protection.
Collapse
Affiliation(s)
- Robert C Barber
- University of North Texas Health Science Center, Department of Pharmacology and Neurosciences, Fort Worth, TX, USA
| | - David L Maass
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - D Jean White
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Jureta W Horton
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Steven E Wolf
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Joseph P Minei
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Qun S Zang
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| |
Collapse
|
150
|
Sulphoxythiocarbamates modify cysteine residues in HSP90 causing degradation of client proteins and inhibition of cancer cell proliferation. Br J Cancer 2013; 110:71-82. [PMID: 24322890 PMCID: PMC3887302 DOI: 10.1038/bjc.2013.710] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/11/2013] [Accepted: 10/17/2013] [Indexed: 12/15/2022] Open
Abstract
Background: Heat shock protein 90 (HSP90) has a key role in the maintenance of the cellular proteostasis. However, HSP90 is also involved in stabilisation of oncogenic client proteins and facilitates oncogene addiction and cancer cell survival. The development of HSP90 inhibitors for cancer treatment is an area of growing interest as such agents can affect multiple pathways that are linked to all hallmarks of cancer. This study aimed to test the hypothesis that targeting cysteine residues of HSP90 will lead to degradation of client proteins and inhibition of cancer cell proliferation. Methods: Combining chemical synthesis, biological evaluation, and structure–activity relationship analysis, we identified a new class of HSP90 inhibitors. Click chemistry and protease-mass spectrometry established the sites of modification of the chaperone. Results: The mildly electrophilic sulphoxythiocarbamate alkyne (STCA) selectively targets cysteine residues of HSP90, forming stable thiocarbamate adducts. Without interfering with the ATP-binding ability of the chaperone, STCA destabilises the client proteins RAF1, HER2, CDK1, CHK1, and mutant p53, and decreases proliferation of breast cancer cells. Addition of a phenyl or a tert-butyl group in tandem with the benzyl substituent at nitrogen increased the potency. A new compound, S-4, was identified as the most robust HSP90 inhibitor within a series of 19 derivatives. Conclusion: By virtue of their cysteine reactivity, sulphoxythiocarbamates target HSP90, causing destabilisation of its client oncoproteins and inhibiting cell proliferation.
Collapse
|