151
|
Yang J, Bridges K, Chen KY, Liu AYC. Riluzole increases the amount of latent HSF1 for an amplified heat shock response and cytoprotection. PLoS One 2008; 3:e2864. [PMID: 18682744 PMCID: PMC2481402 DOI: 10.1371/journal.pone.0002864] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Accepted: 07/04/2008] [Indexed: 11/19/2022] Open
Abstract
Background Induction of the heat shock response (HSR) and increased expression of the heat shock proteins (HSPs) provide mechanisms to ensure proper protein folding, trafficking, and disposition. The importance of HSPs is underscored by the understanding that protein mis-folding and aggregation contribute centrally to the pathogenesis of neurodegenerative diseases. Methodology/Principal Findings We used a cell-based hsp70-luciferease reporter gene assay system to identify agents that modulate the HSR and show here that clinically relevant concentrations of the FDA-approved ALS drug riluzole significantly increased the heat shock induction of hsp70-luciferse reporter gene. Immuno-Western and -cytochemical analysis of HSF1 show that riluzole increased the amount of cytosolic HSF1 to afford a greater activation of HSF1 upon heat shock. The increased HSF1 contributed centrally to the cytoprotective activity of riluzole as hsf1 gene knockout negated the synergistic activity of riluzole and conditioning heat shock to confer cell survival under oxidative stress. Evidence of a post-transcriptional mechanism for the increase in HSF1 include: quantitation of mRNAhsf1 by RT-PCR showed no effect of either heat shock or riluzole treatment; riluzole also increased the expression of HSF1 from a CMV-promoter; analysis of the turnover of HSF1 by pulse chase and immunoprecipitation show that riluzole slowed the decay of [35S]labeled-HSF1. The effect of riluzole on HSF1 was qualitatively different from that of MG132 and chloroquine, inhibitors of the proteasome and lysosome, respectively, and appeared to involve the chaperone-mediated autophagy pathway as RNAi-mediated knockdown of CMA negated its effect. Conclusion/Significance We show that riluzole increased the amount of HSF1 to amplify the HSR for cytoprotection. Our study provides novel insight into the mechanism that regulates HSF1 turnover, and identifies the degradation of HSF1 as a target for therapeutics intervention.
Collapse
Affiliation(s)
- Jingxian Yang
- Department of Cell Biology and Neuroscience, Rutgers State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kristen Bridges
- Department of Cell Biology and Neuroscience, Rutgers State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kuang Yu Chen
- Department of Chemistry and Chemical Biology, Rutgers State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Alice Y.-C. Liu
- Department of Cell Biology and Neuroscience, Rutgers State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
152
|
Promoter ChIP-chip analysis in mouse testis reveals Y chromosome occupancy by HSF2. Proc Natl Acad Sci U S A 2008; 105:11224-9. [PMID: 18682557 DOI: 10.1073/pnas.0800620105] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mammalian Y chromosome is essential for spermatogenesis, which is characterized by sperm cell differentiation and chromatin condensation for acquisition of correct shape of the sperm. Deletions of the male-specific region of the mouse Y chromosome long arm (MSYq), harboring multiple copies of a few genes, lead to sperm head defects and impaired fertility. Using chromatin immunoprecipitation on promoter microarray (ChIP-chip) on mouse testis, we found a striking in vivo MSYq occupancy by heat shock factor 2 (HSF2), a transcription factor involved in spermatogenesis. HSF2 was also found to regulate the transcription of MSYq resident genes, whose transcriptional regulation has been unknown. Importantly, disruption of Hsf2 caused a similar phenotype as the 2/3 deletion of MSYq, i.e., altered expression of the multicopy genes and increased mild sperm head abnormalities. Consequently, aberrant levels of chromatin packing proteins and more frequent DNA fragmentation were detected, implying that HSF2 is required for correct chromatin organization in the sperm. Our findings define a physiological role for HSF2 in the regulation of MSYq resident genes and the quality of sperm.
Collapse
|
153
|
Wilkerson DC, Murphy LA, Sarge KD. Interaction of HSF1 and HSF2 with the Hspa1b promoter in mouse epididymal spermatozoa. Biol Reprod 2008; 79:283-8. [PMID: 18434628 PMCID: PMC2574705 DOI: 10.1095/biolreprod.107.066241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 11/25/2007] [Accepted: 04/11/2008] [Indexed: 11/01/2022] Open
Abstract
The Hspa1b gene is one of the first genes expressed after fertilization, with expression observed in the male pronucleus as early as the one-cell stage of embryogenesis. This expression can occur in the absence of stress and is initiated during the minor zygotic genome activation. There is a significant reduction in the number of embryos developing to the blastocyte stage when HSPA1B levels are depleted, which supports the importance of this protein for embryonic viability. However, the mechanism responsible for allowing expression of Hspa1b during the minor zygotic genome activation (ZGA) is unknown. In this report, we investigated the role of HSF1 and HSF2 in bookmarking Hspa1b during late spermatogenesis. Western blot results show that both HSF1 and HSF2 are present in epididymal spermatozoa, and immunofluorescence analysis revealed that some of the HSF1 and HSF2 proteins in these cells overlap the 4',6'-diamidino-2-phenylindole-stained DNA region. Results from chromatin immunoprecipitation assays showed that HSF1, HSF2, and SP1 are bound to the Hspa1b promoter in epididymal spermatozoa. Furthermore, we observed an increase in HSF2 binding to the Hspa1b promoter in late spermatids versus early spermatids, suggesting a likely period during spermatogenesis when transcription factor binding could occur. These results support a model in which the binding of HSF1, HSF2, and SP1 to the promoter of Hspa1b would allow the rapid formation of a transcription-competent state during the minor ZGA, thereby allowing Hspa1b expression.
Collapse
Affiliation(s)
- Donald C. Wilkerson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536
| | - Lynea A. Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536
| | - Kevin D. Sarge
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
154
|
Jung AE, Fitzsimons HL, Bland RJ, During MJ, Young D. HSP70 and constitutively active HSF1 mediate protection against CDCrel-1-mediated toxicity. Mol Ther 2008; 16:1048-55. [PMID: 18398426 PMCID: PMC3793395 DOI: 10.1038/mt.2008.68] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Defects in cellular quality control mechanisms are thought to contribute to the neuropathology of Parkinson's disease (PD). Overexpressing heat shock proteins (HSPs) may constitute a powerful therapeutic strategy for PD, because they boost the ability of the cell to eliminate unwanted proteins. We investigated the neuroprotective potential of HSP70, HSP40, and H-BH, a constitutively active form of heat shock factor 1, in a rat model of PD based on adeno-associated virus (AAV) vector-mediated overexpression of CDCrel-1, a parkin substrate known to be toxic to dopaminergic neurons. AAV vector-mediated overexpression of H-BH and of HSP70 afforded similar levels of protection against CDCrel-1 toxicity, with approximately 20% improvement in survival of dopaminergic neurons as compared to the controls. The assessment of protection conferred was made using tyrosine hydroxylase (TH) and HuC/D immunohistochemistry and Fluoro-Gold retrograde tracing, and by observing the extent of preservation of spontaneous function and also the extent of drug-induced motor function. In contrast to H-BH and HSP70, HSP40 overexpression exacerbated CDCrel-1-mediated cell death. Real-time reverse transcriptase (RT)-PCR analysis showed that H-BH had the effect of upregulating endogenous HSP70 and HSP40 mRNA levels 10-fold and 4-fold over basal levels, respectively, whereas AAV vector-mediated HSP70 and HSP40 mRNA levels were over 100-fold higher. Our results suggest that a comparatively modest upregulation of multiple HSPs may be an effective approach for achieving significant neuroprotection in PD.
Collapse
Affiliation(s)
- Alisha E Jung
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | | | | | | |
Collapse
|
155
|
Singh IS, Gupta A, Nagarsekar A, Cooper Z, Manka C, Hester L, Benjamin IJ, He JR, Hasday JD. Heat shock co-activates interleukin-8 transcription. Am J Respir Cell Mol Biol 2008; 39:235-42. [PMID: 18367728 DOI: 10.1165/rcmb.2007-0294oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The heat shock (HS) response is a phylogenetically ancient cellular response to stress, including heat, that shifts gene expression to a set of conserved HS protein (HSP) genes. In our earlier studies, febrile-range hyperthermia (FRH) not only activated HSP gene expression, but also increased expression of CXC chemokines in mice, leading us to hypothesize that the CXC chemokine family of genes might be HS-responsive. To address this hypothesis we analyzed the effect of HS on the expression of IL-8/CXCL-8, a member of the human CXC family of ELR(+) chemokines. HS markedly enhanced TNF-alpha-induced IL-8 secretion in human A549 respiratory epithelial-like cells and in primary human small airway epithelial cells. IL-8 mRNA was also up-regulated by HS, but the stability of IL-8 mRNA was not affected. TNF-alpha-induced reporter activity of an IL-8 promoter construct IL8(-1471/+44)-luc stably transfected in A549 cells was also enhanced by HS. Electrophoretic mobility and chromatin immunoprecipitation assays showed that the stress-activated transcription factor heat shock factor-1 (HSF-1) binds to at least two putative heat shock response elements (HSE) present in the IL-8 promoter. Deletional reporter constructs lacking either one or both of these sites showed reduced HS responsiveness. Furthermore, depletion of HSF-1 using siRNA also reduced the effects HS on TNF-alpha-induced IL-8 expression, demonstrating that HSF-1 could also act to regulate IL-8 gene transcription. We speculate that during evolution the CXC chemokine genes may have co-opted elements of the HS response to amplify their expression and enhance neutrophil delivery during febrile illnesses.
Collapse
Affiliation(s)
- Ishwar S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Hwang SO, Boswell SA, Seo JS, Lee SW. Novel oxidative stress-responsive gene ERS25 functions as a regulator of the heat-shock and cell death response. J Biol Chem 2008; 283:13063-9. [PMID: 18326488 DOI: 10.1074/jbc.m709656200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the yeast p24 family, including Emp24p and Erv25p, exist as heteromeric complexes that have been proposed to cycle between the endoplasmic reticulum (ER) and Golgi compartments. The specific functions and sites of action of p24 proteins are still unknown. Here we identified a human homolog of the yeast p24 family of proteins, named ERS25 (endoplasmic reticulum stress-response protein 25), and investigated its role in stress response. ERS25 is predicted to have an ER localization signal peptide, a GOLD (Golgi dynamics) domain, which is found in several eukaryotic Golgi and lipid-trafficking proteins, a coiled-coil region, and a transmembrane domain. We demonstrate that ERS25 is localized to the ER and is induced by ER-specific stress, heat shock, and oxidative stress. The selective induction of ERS25 by brefeldin A, but not tunicamycin, implicates the involvement of ERS25 in protein trafficking between the ER and the Golgi. Small interfering RNA-mediated inhibition of ERS25 results in a significant decrease in apoptosis as well as a reduction of reactive oxygen species induced by oxidative stress. Moreover, ERS25 depletion results in a significant increase in the levels of the ER chaperone HSP70 in response to heat-shock stress through increased levels of HSF-1. We also found that inhibition of ERS25 induction in response to heat shock enhanced the binding of HSP70 to Apaf-1, which is likely to interfere in stress-mediated apoptosis. Together, the data presented here demonstrate that ERS25 may play a critical role in regulation of heat-shock response and apoptosis.
Collapse
Affiliation(s)
- Sun Ok Hwang
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
157
|
Dokladny K, Ye D, Kennedy JC, Moseley PL, Ma TY. Cellular and molecular mechanisms of heat stress-induced up-regulation of occludin protein expression: regulatory role of heat shock factor-1. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:659-70. [PMID: 18276783 DOI: 10.2353/ajpath.2008.070522] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The heat stress (HS)-induced increase in occludin protein expression has been postulated to be a protective response against HS-induced disruption of the intestinal epithelial tight junction barrier. The aim of this study was to elucidate the cellular and molecular processes that mediate the HS-induced up-regulation of occludin expression in Caco-2 cells. Exposure to HS (39 degrees C or 41 degrees C) resulted in increased expression of occludin protein; this was preceded by an increase in occludin mRNA transcription and promoter activity. HS-induced activation of heat shock factor-1 (HSF-1) resulted in cytoplasmic-to-nuclear translocation of HSF-1 and binding to its binding motif in the occludin promoter region. HSF-1 activation was associated with an increase in occludin promoter activity, mRNA transcription, and protein expression; which were abolished by the HSF-1 inhibitor quercetin. Targeted HSF-1 knock-down by siRNA transfection inhibited the HSF-1-induced increase in occulin expression and junctional localization of occulin protein. Site-directed mutagenesis of the HSF-1 binding motif in the occludin promoter region inhibited HS-induced binding of HSF-1 to the occludin promoter region and subsequent promoter activity. In conclusion, our data show for the first time that the HS-induced increase in occludin protein expression is mediated by HSF-1 activation and subsequent binding of HSF-1 to the occludin promoter, which initiates a series of molecular and cellular events culminating in increased junctional localization of occludin protein.
Collapse
Affiliation(s)
- Karol Dokladny
- Internal Medicine-Gastroenterology and Hepatology, MSC10 5550, 1 University of New Mexico, Albuquerque, NM 87131-0001, USA
| | | | | | | | | |
Collapse
|
158
|
Zhang YQ, Sarge KD. Celastrol inhibits polyglutamine aggregation and toxicity though induction of the heat shock response. J Mol Med (Berl) 2007; 85:1421-8. [PMID: 17943263 PMCID: PMC2262918 DOI: 10.1007/s00109-007-0251-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 07/06/2007] [Accepted: 07/19/2007] [Indexed: 11/27/2022]
Abstract
Heat shock proteins (hsps) are protective against the harmful effects of mutant expanded polyglutamine repeat proteins that occur in diseases such as Huntington's, prompting the search for pharmacologic compounds that increase hsp expression in cells as potential treatments for this and related diseases. In this paper, we show that celastrol, a compound recently shown to up-regulate hsp gene expression, significantly decreases killing of cells expressing mutant polyglutamine protein. This effect requires the presence of the transcription factor responsible for mediating inducible hsp gene expression, HSF1, and is correlated with decreased amounts and increased sodium dodecyl sulfate (SDS) solubility of polyglutamine aggregates. These results suggest the potential of celastrol as a therapeutic agent in the treatment of human polyglutamine expansion diseases.
Collapse
Affiliation(s)
- Yu-Qian Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone Street, Lexington, KY 40536 USA
| | - Kevin D. Sarge
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone Street, Lexington, KY 40536 USA
| |
Collapse
|
159
|
Skaggs HS, Xing H, Wilkerson DC, Murphy LA, Hong Y, Mayhew CN, Sarge KD. HSF1-TPR interaction facilitates export of stress-induced HSP70 mRNA. J Biol Chem 2007; 282:33902-7. [PMID: 17897941 PMCID: PMC2266631 DOI: 10.1074/jbc.m704054200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Stress conditions inhibit mRNA export, but mRNAs encoding heat shock proteins continue to be efficiently exported from the nucleus during stress. How HSP mRNAs bypass this stress-associated export inhibition was not known. Here, we show that HSF1, the transcription factor that binds HSP promoters after stress to induce their transcription, interacts with the nuclear pore-associating TPR protein in a stress-responsive manner. TPR is brought into proximity of the HSP70 promoter after stress and preferentially associates with mRNAs transcribed from this promoter. Disruption of the HSF1-TPR interaction inhibits the export of mRNAs expressed from the HSP70 promoter, both endogenous HSP70 mRNA and a luciferase reporter mRNA. These results suggest that HSP mRNA export escapes stress inhibition via HSF1-mediated recruitment of the nuclear pore-associating protein TPR to HSP genes, thereby functionally connecting the first and last nuclear steps of the gene expression pathway, transcription and mRNA export.
Collapse
Affiliation(s)
- Hollie S Skaggs
- Department of Molecular and Cellular Biochemistry, Chandler Medical Center, University of Kentucky, 741 S. Limestone Street, Lexington, KY 40536-0084, USA
| | | | | | | | | | | | | |
Collapse
|
160
|
Wilkerson DC, Skaggs HS, Sarge KD. HSF2 binds to the Hsp90, Hsp27, and c-Fos promoters constitutively and modulates their expression. Cell Stress Chaperones 2007; 12:283-90. [PMID: 17915561 PMCID: PMC1971238 DOI: 10.1379/csc-250.1] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Although the vast majority of genomic DNA is tightly compacted during mitosis, the promoter regions of a number of genes remain in a less compacted state throughout this stage of the cell cycle. The decreased compaction of these promoter regions, which is referred to as gene bookmarking, is thought to be important for the ability of cells to express these genes during the following interphase. Previously, we reported a role for the DNA-binding protein heat shock factor (HSF2) in bookmarking the stress-inducible 70,000-Da heat shock protein (hsp70) gene. In this report, we have extended those studies and found that during mitosis, HSF2 is bound to the HSE promoter elements of other heat shock genes, including hsp90 and hsp27, as well as the proto-oncogene c-fos. The presence of HSF2 is important for expression of these genes because blocking HSF2 levels by RNA interference techniques leads to decreased levels of these proteins. These results suggest that HSF2 is important for constitutive as well as stress-inducible expression of HSE-containing genes.
Collapse
Affiliation(s)
- Donald C Wilkerson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | | | | |
Collapse
|
161
|
Xing H, Hong Y, Sarge KD. Identification of the PP2A-interacting region of heat shock transcription factor 2. Cell Stress Chaperones 2007; 12:192-7. [PMID: 17688198 PMCID: PMC1949333 DOI: 10.1379/csc-249r.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Previous work in our laboratory demonstrated the existence of an association between heat shock transcription factor 2 (HSF2) and the serine/threonine phosphatase 2A, which is mediated by interaction between HSF2 and the A subunit (also called PR65) of this protein phosphatase. In light of the importance of HSF2-PP2A association for HSF2 cellular function, in this study, we have sought to dissect the sequences within HSF2 that are important for interaction with the A subunit of PP2A. The results of these experiments indicate that the HSF2 region comprising amino acids 343-363 is important for A subunit interaction. This region includes part of the C-terminal leucine zipper motif of HSF2 called heptad repeat C (HR-C). The results of transfection/immunoprecipitation experiments also show that deletion of the 6 amino acids from 343 to 348 from HSF2 (HSF2 (delta343-348)), is sufficient to prevent HSF2 from interacting with PP2A. These data provide insight into a new functional domain of HSF2, the PP2A A subunit-interacting region.
Collapse
Affiliation(s)
- Hongyan Xing
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S Limestone Streeet, Lexington, KY 40536, USA
| | | | | |
Collapse
|
162
|
Nagy E, Balogi Z, Gombos I, Åkerfelt M, Björkbom A, Balogh G, Török Z, Maslyanko A, Fiszer-Kierzkowska A, Lisowska K, Slotte PJ, Sistonen L, Horváth I, Vígh L. Hyperfluidization-coupled membrane microdomain reorganization is linked to activation of the heat shock response in a murine melanoma cell line. Proc Natl Acad Sci U S A 2007; 104:7945-50. [PMID: 17470815 PMCID: PMC1876552 DOI: 10.1073/pnas.0702557104] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Indexed: 12/21/2022] Open
Abstract
Targeting of the Hsp function in tumor cells is currently being assessed as potential anticancer therapy. An improved understanding of the molecular signals that trigger or attenuate the stress protein response is essential for advances to be made in this field. The present study provides evidence that the membrane fluidizer benzyl alcohol (BA), a documented nondenaturant, acts as a chaperone inducer in B16(F10) melanoma cells. It is demonstrated that this effect relies basically on heat shock transcription factor 1 (HSF1) activation. Under the conditions tested, the BA-induced Hsp response involves the up-regulation of a subset of hsp genes. It is shown that the same level of membrane fluidization (estimated in the core membrane region) attained with the closely analogous phenethyl alcohol (PhA) does not generate a stress protein signal. BA, at a concentration that activates heat shock genes, exerts a profound effect on the melting of raft-like cholesterol-sphingomyelin domains in vitro, whereas PhA, at a concentration equipotent with BA in membrane fluidization, has no such effect. Furthermore, through the in vivo labeling of melanoma cells with a fluorescein labeled probe that inserts into the cholesterol-rich membrane domains [fluorescein ester of polyethylene glycol-derivatized cholesterol (fPEG-Chol)], we found that, similarly to heat stress per se, BA, but not PhA, initiates profound alterations in the plasma membrane microdomain structure. We suggest that, apart from membrane hyperfluidization in the deep hydrophobic region, a distinct reorganization of cholesterol-rich microdomains may also be required for the generation and transmission of stress signals to activate hsp genes.
Collapse
Affiliation(s)
- Enikő Nagy
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Zsolt Balogi
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Imre Gombos
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Malin Åkerfelt
- Department of Biology, Turku Center for Biotechnology, and
| | - Anders Björkbom
- Department of Biochemistry and Pharmacy, Abo Akademi University, FI-20500, Turku, Finland; and
| | - Gábor Balogh
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Zsolt Török
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Andriy Maslyanko
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - Anna Fiszer-Kierzkowska
- Department of Tumor Biology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, 02-781, Gliwice, Poland
| | - Katarzyna Lisowska
- Department of Tumor Biology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, 02-781, Gliwice, Poland
| | - Peter J. Slotte
- Department of Biochemistry and Pharmacy, Abo Akademi University, FI-20500, Turku, Finland; and
| | - Lea Sistonen
- Department of Biology, Turku Center for Biotechnology, and
| | - Ibolya Horváth
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| | - László Vígh
- *Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, P.O. Box 521, H-6701, Szeged, Hungary
| |
Collapse
|
163
|
Wheeler DS, Wong HR. Heat shock response and acute lung injury. Free Radic Biol Med 2007; 42:1-14. [PMID: 17157189 PMCID: PMC1790871 DOI: 10.1016/j.freeradbiomed.2006.08.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 08/23/2006] [Accepted: 08/29/2006] [Indexed: 11/19/2022]
Abstract
All cells respond to stress through the activation of primitive, evolutionarily conserved genetic programs that maintain homeostasis and assure cell survival. Stress adaptation, which is known in the literature by a myriad of terms, including tolerance, desensitization, conditioning, and reprogramming, is a common paradigm found throughout nature, in which a primary exposure of a cell or organism to a stressful stimulus (e.g., heat) results in an adaptive response by which a second exposure to the same stimulus produces a minimal response. More interesting is the phenomenon of cross-tolerance, by which a primary exposure to a stressful stimulus results in an adaptive response whereby the cell or organism is resistant to a subsequent stress that is different from the initial stress (i.e., exposure to heat stress leading to resistance to oxidant stress). The heat shock response is one of the more commonly described examples of stress adaptation and is characterized by the rapid expression of a unique group of proteins collectively known as heat shock proteins (also commonly referred to as stress proteins). The expression of heat shock proteins is well described in both whole lungs and in specific lung cells from a variety of species and in response to a variety of stressors. More importantly, in vitro data, as well as data from various animal models of acute lung injury, demonstrate that heat shock proteins, especially Hsp27, Hsp32, Hsp60, and Hsp70 have an important cytoprotective role during lung inflammation and injury.
Collapse
Affiliation(s)
- Derek S. Wheeler
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center; Kindervelt Laboratory for Critical Care Medicine Research, Children’s Hospital Research Foundation;]Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Hector R. Wong
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center; Kindervelt Laboratory for Critical Care Medicine Research, Children’s Hospital Research Foundation;]Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
164
|
Jolly C, Lakhotia SC. Human sat III and Drosophila hsr omega transcripts: a common paradigm for regulation of nuclear RNA processing in stressed cells. Nucleic Acids Res 2006; 34:5508-14. [PMID: 17020918 PMCID: PMC1636489 DOI: 10.1093/nar/gkl711] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Exposure of cells to stressful conditions elicits a highly conserved defense mechanism termed the heat shock response, resulting in the production of specialized proteins which protect the cells against the deleterious effects of stress. The heat shock response involves not only a widespread inhibition of the ongoing transcription and activation of heat shock genes, but also important changes in post-transcriptional processing. In particular, a blockade in splicing and other post-transcriptional processing has been described following stress in different organisms, together with an altered spatial distribution of the proteins involved in these activities. However, the specific mechanisms that regulate these activities under conditions of stress are little understood. Non-coding RNA molecules are increasingly known to be involved in the regulation of various activities in the cell, ranging from chromatin structure to splicing and RNA degradation. In this review, we consider two non-coding RNAs, the hsrω transcripts in Drosophila and the sat III transcripts in human cells, that seem to be involved in the dynamics of RNA-processing factors in normal and/or stressed cells, and thus provide new paradigms for understanding transcriptional and post-transcriptional regulations in normal and stressed cells.
Collapse
|
165
|
Chang Y, Ostling P, Akerfelt M, Trouillet D, Rallu M, Gitton Y, El Fatimy R, Fardeau V, Le Crom S, Morange M, Sistonen L, Mezger V. Role of heat-shock factor 2 in cerebral cortex formation and as a regulator of p35 expression. Genes Dev 2006; 20:836-47. [PMID: 16600913 PMCID: PMC1472286 DOI: 10.1101/gad.366906] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Heat-shock factors (HSFs) are associated with multiple developmental processes, but their mechanisms of action in these processes remain largely enigmatic. Hsf2-null mice display gametogenesis defects and brain abnormalities characterized by enlarged ventricles. Here, we show that Hsf2-/- cerebral cortex displays mispositioning of neurons of superficial layers. HSF2 deficiency resulted in a reduced number of radial glia fibers, the architectural guides for migrating neurons, and of Cajal-Retzius cells, which secrete the positioning signal Reelin. Therefore, we focused on the radial migration signaling pathways. The levels of Reelin and Dab1 tyrosine phosphorylation were reduced, suggesting that the Reelin cascade is affected in Hsf2-/- cortices. The expression of p35, an activator of cyclin-dependent kinase 5 (Cdk5), essential for radial migration, was dependent on the amount of HSF2 in gain- and loss-of-function systems. p39, another Cdk5 activator, displayed reduced mRNA levels in Hsf2-/- cortices, which, together with the lowered p35 levels, decreased Cdk5 activity. We demonstrate in vivo binding of HSF2 to the p35 promoter and thereby identify p35 as the first target gene for HSF2 in cortical development. In conclusion, HSF2 affects cellular populations that assist in radial migration and directly regulates the expression of p35, a crucial actor of radial neuronal migration.
Collapse
Affiliation(s)
- Yunhua Chang
- Biologie Moléculaire du Stress, Centre National de la Recherche Scientifique (CNRS) UMR8541, Ecole Normale Supérieure, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Stanhill A, Levin V, Hendel A, Shachar I, Kazanov D, Arber N, Kaminski N, Engelberg D. Ha-ras(val12) induces HSP70b transcription via the HSE/HSF1 system, but HSP70b expression is suppressed in Ha-ras(val12)-transformed cells. Oncogene 2006; 25:1485-95. [PMID: 16278678 DOI: 10.1038/sj.onc.1209193] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heat shock proteins (Hsps) are overexpressed in many tumors, but are downregulated in some tumors. To check for a direct effect of Ha-Ras(val12) on HSP70 transcription, we transiently expressed the oncoprotein in Rat1 fibroblasts and monitored its effect on HSP70b promoter-driven reporter gene. We show that expression of Ha-Ras(val12) induced this promoter. Promoter analysis via systematic deletions and point mutations revealed that Ha-Ras(val12) induces HSP70b transcription via heat shock elements (HSEs). Also, Ha-Ras(val12) induction of HSE-mediated transcription was dramatically reduced in HSF1-/- cells. Yet, residual effect of Ha-Ras(val12) that was still measured in HSF1-/- cells suggests that some of the Ha-Ras(val12) effect is Hsf1-independent. When HSF1-/- cells, stably expressing Ha-Ras(val12), were grown on soft agar only small colonies were formed suggesting a role for heat shock factor 1 (Hsf1) in Ha-Ras(val12)-mediated transformation. Although Ha-ras(Val12) seems to be an inducer of HSP70's expression, we found that in Ha-ras(Val12-)transformed fibroblasts expression of this gene is suppressed. This suppression is correlated with higher sensitivity of Ha-ras(val12)-transformed cells to heat shock. We suggest that Ha-ras(Val12) is involved in Hsf1 activation, thereby inducing the cellular protective response. Cells that repress this response are perhaps those that acquire the capability to further proliferate and become transformed clones.
Collapse
Affiliation(s)
- A Stanhill
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
167
|
Shilova VY, Garbuz DG, Myasyankina EN, Chen B, Evgen'ev MB, Feder ME, Zatsepina OG. Remarkable site specificity of local transposition into the Hsp70 promoter of Drosophila melanogaster. Genetics 2006; 173:809-20. [PMID: 16582443 PMCID: PMC1526513 DOI: 10.1534/genetics.105.053959] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heat-shock genes have numerous features that ought to predispose them to insertional mutagenesis via transposition. To elucidate the evolvability of heat-shock genes via transposition, we have exploited a local transposition technique and Drosophila melanogaster strains with EPgy2 insertions near the Hsp70 gene cluster at 87A7 to produce numerous novel EPgy2 insertions into these Hsp70 genes. More than 50% of 45 independent insertions were made into two adjacent nucleotides in the proximal promoter at positions -96 and -97, and no insertions were into a coding or 3'-flanking sequence. All inserted transposons were in inverse orientation to the starting transposon. The frequent insertion into nucleotides -96 and -97 is consistent with the DNase hypersensitivity, absence of nucleosomes, flanking GAGA-factor-binding sites, and nucleotide sequence of this region. These experimental insertions recapitulated many of the phenotypes of natural transposition into Hsp70: reduced mRNA expression, less Hsp70 protein, and decreased inducible thermotolerance. The results suggest that the distinctive features of heat-shock promoters, which underlie the massive and rapid expression of heat-shock genes upon heat shock, also are a source of evolutionary variation on which natural selection can act.
Collapse
Affiliation(s)
- Victoria Y Shilova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
168
|
Singh H, Erkine AM, Kremer SB, Duttweiler HM, Davis DA, Iqbal J, Gross RR, Gross DS. A functional module of yeast mediator that governs the dynamic range of heat-shock gene expression. Genetics 2006; 172:2169-84. [PMID: 16452140 PMCID: PMC1456402 DOI: 10.1534/genetics.105.052738] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 01/20/2006] [Indexed: 11/18/2022] Open
Abstract
We report the results of a genetic screen designed to identify transcriptional coregulators of yeast heat-shock factor (HSF). This sequence-specific activator is required to stimulate both basal and induced transcription; however, the identity of factors that collaborate with HSF in governing noninduced heat-shock gene expression is unknown. In an effort to identify these factors, we isolated spontaneous extragenic suppressors of hsp82-deltaHSE1, an allele of HSP82 that bears a 32-bp deletion of its high-affinity HSF-binding site, yet retains its two low-affinity HSF sites. Nearly 200 suppressors of the null phenotype of hsp82-deltaHSE1 were isolated and characterized, and they sorted into six expression without heat-shock element (EWE) complementation groups. Strikingly, all six groups contain alleles of genes that encode subunits of Mediator. Three of the six subunits, Med7, Med10/Nut2, and Med21/Srb7, map to Mediator's middle domain; two subunits, Med14/Rgr1 and Med16/Sin4, to its tail domain; and one subunit, Med19/Rox3, to its head domain. Mutations in genes encoding these factors enhance not only the basal transcription of hsp82-deltaHSE1, but also that of wild-type heat-shock genes. In contrast to their effect on basal transcription, the more severe ewe mutations strongly reduce activated transcription, drastically diminishing the dynamic range of heat-shock gene expression. Notably, targeted deletion of other Mediator subunits, including the negative regulators Cdk8/Srb10, Med5/Nut1, and Med15/Gal11 fail to derepress hsp82-deltaHSE1. Taken together, our data suggest that the Ewe subunits constitute a distinct functional module within Mediator that modulates both basal and induced heat-shock gene transcription.
Collapse
Affiliation(s)
- Harpreet Singh
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Anckar J, Hietakangas V, Denessiouk K, Thiele DJ, Johnson MS, Sistonen L. Inhibition of DNA binding by differential sumoylation of heat shock factors. Mol Cell Biol 2006; 26:955-64. [PMID: 16428449 PMCID: PMC1347039 DOI: 10.1128/mcb.26.3.955-964.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Covalent modification of proteins by the small ubiquitin-related modifier SUMO regulates diverse biological functions. Sumoylation usually requires a consensus tetrapeptide, through which the binding of the SUMO-conjugating enzyme Ubc9 to the target protein is directed. However, additional specificity determinants are in many cases required. To gain insights into SUMO substrate selection, we have utilized the differential sumoylation of highly similar loop structures within the DNA-binding domains of heat shock transcription factor 1 (HSF1) and HSF2. Site-specific mutagenesis in combination with molecular modeling revealed that the sumoylation specificity is determined by several amino acids near the consensus site, which are likely to present the SUMO consensus motif to Ubc9. Importantly, we also demonstrate that sumoylation of the HSF2 loop impedes HSF2 DNA-binding activity, without affecting its oligomerization. Hence, SUMO modification of the HSF2 loop contributes to HSF-specific regulation of DNA binding and broadens the concept of sumoylation in the negative regulation of gene expression.
Collapse
Affiliation(s)
- Julius Anckar
- Turku Centre for Biotechnology, P.O. Box 123, FI-20521 Turku, Finland
| | | | | | | | | | | |
Collapse
|
170
|
Ahn SG, Kim SA, Yoon JH, Vacratsis P. Heat-shock cognate 70 is required for the activation of heat-shock factor 1 in mammalian cells. Biochem J 2006; 392:145-52. [PMID: 16050811 PMCID: PMC1317673 DOI: 10.1042/bj20050412] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
HSF1 (heat-shock factor 1) plays an essential role in mediating the appropriate cellular response to diverse forms of physiological stresses. However, it is not clear how HSF1 is regulated by interacting proteins under normal and stressful conditions. In the present study, Hsc70 (heat-shock cognate 70) was identified as a HSF1-interacting protein using the TAP (tandem affinity purification) system and MS. HSF1 can interact with Hsc70 in vivo and directly in vitro. Interestingly, Hsc70 is required for the regulation of HSF1 during heat stress and subsequent target gene expression in mammalian cells. Moreover, cells transfected with siRNAs (small interfering RNAs) targeted to Hsc70 showed greatly decreased HSF1 activation with expression of HSF1 target genes being dramatically reduced. Finally, loss of Hsc70 expression in cells resulted in an increase in stress-induced apoptosis. These results indicate that Hsc70 is a necessary and critical regulator of HSF1 activities.
Collapse
Affiliation(s)
- Sang-Gun Ahn
- Department of Pathology, Chosun University College of Dentistry, Gwangju 501-759, South Korea.
| | | | | | | |
Collapse
|
171
|
Tchénio T, Havard M, Martinez LA, Dautry F. Heat shock-independent induction of multidrug resistance by heat shock factor 1. Mol Cell Biol 2006; 26:580-91. [PMID: 16382149 PMCID: PMC1346900 DOI: 10.1128/mcb.26.2.580-591.2006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The screening of two different retroviral cDNA expression libraries to select genes that confer constitutive doxorubicin resistance has in both cases resulted in the isolation of the heat shock factor 1 (HSF1) transcription factor. We show that HSF1 induces a multidrug resistance phenotype that occurs in the absence of heat shock or cellular stress and is mediated at least in part through the constitutive activation of the multidrug resistance gene 1 (MDR-1). This drug resistance phenotype does not correlate with an increased expression of heat shock-responsive genes (heat shock protein genes, or HSPs). In addition, HSF1 mutants lacking HSP gene activation are also capable of conferring multidrug resistance, and only hypophosphorylated HSF1 complexes accumulate in transduced cells. Our results indicate that HSF1 can activate MDR-1 expression in a stress-independent manner that differs from the canonical heat shock-activated mechanism involved in HSP induction. We further provide evidence that the induction of MDR-1 expression occurs at a posttranscriptional level, revealing a novel undocumented role for hypophosphorylated HSF1 in posttranscriptional gene regulation.
Collapse
Affiliation(s)
- Thierry Tchénio
- Unité de Génétique Moléculaire et Intégrations des Fonctions Cellulaires, CNRS-UPR1983, 7 rue Guy Moquet, BP8, 94801 Villejuif Cedex, France.
| | | | | | | |
Collapse
|
172
|
Papaconstantinou M, Wu Y, Pretorius HN, Singh N, Gianfelice G, Tanguay RM, Campos AR, Bédard PA. Menin is a regulator of the stress response in Drosophila melanogaster. Mol Cell Biol 2005; 25:9960-72. [PMID: 16260610 PMCID: PMC1280255 DOI: 10.1128/mcb.25.22.9960-9972.2005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Menin, the product of the multiple endocrine neoplasia type I gene, has been implicated in several biological processes, including the control of gene expression and apoptosis, the modulation of mitogen-activated protein kinase pathways, and DNA damage sensing or repair. In this study, we have investigated the function of menin in the model organism Drosophila melanogaster. We show that Drosophila lines overexpressing menin or an RNA interference for this gene develop normally but are impaired in their response to several stresses, including heat shock, hypoxia, hyperosmolarity and oxidative stress. In the embryo subjected to heat shock, this impairment was characterized by a high degree of developmental arrest and lethality. The overexpression of menin enhanced the expression of HSP70 in embryos and interfered with its down-regulation during recovery at the normal temperature. In contrast, the inhibition of menin with RNA interference reduced the induction of HSP70 and blocked the activation of HSP23 upon heat shock, Menin was recruited to the Hsp70 promoter upon heat shock and menin overexpression stimulated the activity of this promoter in embryos. A 70-kDa inducible form of menin was expressed in response to heat shock, indicating that menin is also regulated in conditions of stress. The induction of HSP70 and HSP23 was markedly reduced or absent in mutant embryos harboring a deletion of the menin gene. These embryos, which did not express the heat shock-inducible form of menin, were also hypersensitive to various conditions of stress. These results suggest a novel role for menin in the control of the stress response and in processes associated with the maintenance of protein integrity.
Collapse
Affiliation(s)
- Maria Papaconstantinou
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4K1
| | | | | | | | | | | | | | | |
Collapse
|
173
|
Batulan Z, Nalbantoglu J, Durham HD. Nonsteroidal anti-inflammatory drugs differentially affect the heat shock response in cultured spinal cord cells. Cell Stress Chaperones 2005; 10:185-96. [PMID: 16184763 PMCID: PMC1226016 DOI: 10.1379/csc-30r.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to amplify the heat shock response in cell lines by increasing the binding of heat shock transcription factor-1 to heat shock elements within heat shock gene promoters. Because overexpression of the inducible heat shock protein 70 (Hsp70) was neuroprotective in a culture model of motor neuron disease, this study investigated whether NSAIDs induce Hsp70 and confer cytoprotection in motor neurons of dissociated spinal cord cultures exposed to various stresses. Two NSAIDs, sodium salicylate and niflumic acid, lowered the temperature threshold for induction of Hsp70 in glia but failed to do so in motor neurons. At concentrations that increased Hsp70 in heat shocked glial cells, sodium salicylate failed to delay death of motor neurons exposed to hyperthermia, paraquat-mediated oxidative stress, and glutamate excitotoxicity. Neither sodium salicylate nor the cyclooxygenase-2 inhibitor, niflumic acid, protected motor neurons from the toxicity of mutated Cu/Zn-superoxide dismutase (SOD-1) linked to a familial form of the motor neuron disease, amyotrophic lateral sclerosis. Thus, treatment with 2 types of NSAIDs failed to overcome the high threshold for the activation of heat shock response in motor neurons.
Collapse
Affiliation(s)
- Zarah Batulan
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | | | |
Collapse
|
174
|
Khaleque MA, Bharti A, Sawyer D, Gong J, Benjamin IJ, Stevenson MA, Calderwood SK. Induction of heat shock proteins by heregulin beta1 leads to protection from apoptosis and anchorage-independent growth. Oncogene 2005; 24:6564-73. [PMID: 16007186 DOI: 10.1038/sj.onc.1208798] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Elevation of heat shock protein (HSP) levels is widespread in cancer and predicts a poor prognosis and resistance to therapy. We show that HSP elevation in tumor cells can be induced by the highly malignant factor heregulin beta1 (HRGbeta1), which induces HSP expression through heat shock transcription factor 1 (HSF1). Inactivation of the hsf1 gene prevents HSP induction by HRGbeta1. HSP expression is induced through a cascade response initiated by HRGbeta1 binding to c-erbB receptors on the cell surface and which leads to the inhibition of intracellular HSF1 antagonist glycogen synthase kinase 3. HSF1 activated by this pathway plays a key role in the protection of cells from apoptosis and the mediation of anchorage independent growth by HRGbeta1, indicating a role for HSF1 in this tumorigenic pathway.
Collapse
Affiliation(s)
- Md Abdul Khaleque
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | |
Collapse
|
175
|
Yan D, Saito K, Ohmi Y, Fujie N, Ohtsuka K. Paeoniflorin, a novel heat shock protein-inducing compound. Cell Stress Chaperones 2005; 9:378-89. [PMID: 15633296 PMCID: PMC1065277 DOI: 10.1379/csc-51r.1] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Heat shock proteins (HSPs) are induced by various physical, chemical, and biological stresses. HSPs are known to function as molecular chaperones, and they not only regulate various processes of protein biogenesis but also function as lifeguards against proteotoxic stresses. Because it is very useful to discover nontoxic chaperone-inducing compounds, we searched for them in herbal medicines. Some herbal medicines had positive effects on the induction of HSPs (Hsp70, Hsp40, and Hsp27) in cultured mammalian cells. We next examined 2 major constituents of these herbal medicines, glycyrrhizin and paeoniflorin, with previously defined chemical structures. Glycyrrhizin had an enhancing effect on the HSP induction by heat shock but could not induce HSPs by itself. In contrast, paeoniflorin had not only an enhancing effect but also an inducing effect by itself on HSP expression. Thus, paeoniflorin might be termed a chaperone inducer and glycyrrhizin a chaperone coinducer. Treatment of cells with paeoniflorin but not glycyrrhizin resulted in enhanced phosphorylation and acquisition of the deoxyribonucleic acid-binding ability of heat shock transcription factor 1 (HSF1), as well as the formation of characteristic HSF1 granules in the nucleus, suggesting that the induction of HSPs by paeoniflorin is mediated by the activation of HSF1. Also, thermotolerance was induced by treatment with paeoniflorin but not glycyrrhizin. Paeoniflorin had no toxic effect at concentrations as high as 80 microg/ mL (166.4 microM). To our knowledge, this is the first report on the induction of HSPs by herbal medicines.
Collapse
Affiliation(s)
- Dai Yan
- Laboratory of Cell and Stress Biology, Department of Environmental Biology, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | | | | | | | | |
Collapse
|
176
|
Analysis of phosphorylation of human heat shock factor 1 in cells experiencing a stress. BMC BIOCHEMISTRY 2005; 6:4. [PMID: 15760475 PMCID: PMC1079796 DOI: 10.1186/1471-2091-6-4] [Citation(s) in RCA: 244] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2004] [Accepted: 03/11/2005] [Indexed: 11/25/2022]
Abstract
Background Heat shock factor (HSF/HSF1) not only is the transcription factor primarily responsible for the transcriptional response of cells to physical and chemical stress but also coregulates other important signaling pathways. The factor mediates the stress-induced expression of heat shock or stress proteins (HSPs). HSF/HSF1 is inactive in unstressed cells and is activated during stress. Activation is accompanied by hyperphosphorylation of the factor. The regulatory importance of this phosphorylation has remained incompletely understood. Several previous studies on human HSF1 were concerned with phosphorylation on Ser303, Ser307 and Ser363, which phosphorylation appears to be related to factor deactivation subsequent to stress, and one study reported stress-induced phosphorylation of Ser230 contributing to factor activation. However, no previous study attempted to fully describe the phosphorylation status of an HSF/HSF1 in stressed cells and to systematically identify phosphoresidues involved in factor activation. The present study reports such an analysis for human HSF1 in heat-stressed cells. Results An alanine scan of all Ser, Thr and Tyr residues of human HSF1 was carried out using a validated transactivation assay, and residues phosphorylated in HSF1 were identified by mass spectrometry and sequencing. HSF1 activated by heat treatment was phosphorylated on Ser121, Ser230, Ser292, Ser303, Ser307, Ser314, Ser319, Ser326, Ser344, Ser363, Ser419, and Ser444. Phosphorylation of Ser326 but none of the other Ser residues was found to contribute significantly to activation of the factor by heat stress. Phosphorylation on Ser326 increased rapidly during heat stress as shown by experiments using a pSer326 phosphopeptide antibody. Heat stress-induced DNA binding and nuclear translocation of a S326A substitution mutant was not impaired in HSF1-negative cells, but the mutant stimulated HSP70 expression several times less well than wild type factor. Conclusion Twelve Ser residues but no Thr or Tyr residues were identified that were phosphorylated in heat-activated HSF1. Mutagenesis experiments and functional studies suggested that phosphorylation of HSF1 residue Ser326 plays a critical role in the induction of the factor's transcriptional competence by heat stress. PhosphoSer326 also contributes to activation of HSF1 by chemical stress. To date, no functional role could be ascribed to any of the other newly identified phosphoSer residues.
Collapse
|
177
|
Trinklein ND, Chen WC, Kingston RE, Myers RM. Transcriptional regulation and binding of heat shock factor 1 and heat shock factor 2 to 32 human heat shock genes during thermal stress and differentiation. Cell Stress Chaperones 2005; 9:21-8. [PMID: 15270074 PMCID: PMC1065302 DOI: 10.1379/481.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transcription of mammalian heat shock genes can be regulated by heat shock factors (HSF) 1 and 2. Although it has been shown previously that these factors respond to distinct stimuli, a broad analysis of the induction and function of these factors in living cells has not been performed. In our study, we assayed binding of human HSF1 and HSF2 at the promoters of 32 genes identified through LocusLink as heat shock genes in response to elevated temperature and hemin-induced differentiation in human K562 erythroleukemic cells using the chromatin immunoprecipitation technique. We also measured the induced expression of these genes under these 2 conditions. We found that 17 of the 32 genes were transcriptionally induced during heat shock, and HSF1 binding was detected at 15 of the 17 promoters. Nearly all the genes induced by heat shock were also induced to a lesser degree during hemin treatment. However, some genes were induced significantly more during hemin treatment than during heat shock. A new finding is that HSF1 and HSF2 bind to the same targets, but HSF1 binding is activated more by heat than by hemin treatment, and HSF2 binding is only activated by hemin treatment and not by heat. This technology also identified previously unknown HSF1 binding sites near genes that were previously shown to be heat inducible that may contribute to gene-specific regulation.
Collapse
Affiliation(s)
- Nathan D Trinklein
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | | | | | | |
Collapse
|
178
|
Rieger TR, Morimoto RI, Hatzimanikatis V. Mathematical modeling of the eukaryotic heat-shock response: dynamics of the hsp70 promoter. Biophys J 2004; 88:1646-58. [PMID: 15626701 PMCID: PMC1305221 DOI: 10.1529/biophysj.104.055301] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The heat-shock response in humans and other eukaryotes is a highly conserved genetic network that coordinates the cellular response to protein damage and is essential for adaptation and survival of the stressed cell. It involves an immediate and transient activation of heat-shock transcription factor-1 (HSF1) which results in the elevated expression of genes encoding proteins important for protein homeostasis including molecular chaperones and components of the protein degradative machinery. We have developed a mathematical model of the critical steps in the regulation of HSF1 activity to understand how chronic exposure to a stress signal is converted into specific molecular events for activation and feedback regulated attenuation of HSF1. The model is utilized to identify the most sensitive steps in HSF1 activation and to evaluate how these steps affect the expression of molecular chaperones. This analysis allows the formulation of hypotheses about the differences between the heat-shock responses in yeast and humans and generates a model with predictive abilities relevant to diseases associated with the accumulation of damaged and aggregated proteins including cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Theodore R Rieger
- Department of Chemical and Biological Engineering, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, USA
| | | | | |
Collapse
|
179
|
Tchounwou PB, Yedjou CG, Foxx DN, Ishaque AB, Shen E. Lead-induced cytotoxicity and transcriptional activation of stress genes in human liver carcinoma (HepG2) cells. Mol Cell Biochem 2004; 255:161-70. [PMID: 14971657 DOI: 10.1023/b:mcbi.0000007272.46923.12] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Lead is a non-essential element that exhibits a high degree of toxicity, especially in children. Most research on lead has focused on its effects on organ systems such as the nervous system, the red blood cells, and the kidneys which are considered to be the primary targets of lead toxicity. However, the molecular mechanisms by which it induces toxicity, and carcinogenesis remain to be elucidated. In this research, we performed the MTT assay to assess the cytotoxicity, and the CAT-Tox assay to assess the transcriptional responses associated with lead exposure to thirteen different recombinant cell lines generated from human liver carcinoma cells (HepG2), by creating stable transfectants of mammalian promoter chloramphenicol (CAT) gene fusions. Study results indicated that lead nitrate is cytotoxic to HepG2 cells, showing LD50 values of 49.0 +/- 18.0 microg/mL, 37.5 +/- 9.2 microg/mL, and 3.5 +/- 0.7 microg/mL for cell mortality upon 24, 48 and 72 h of exposure, respectively; indicating a dose- and time-dependent response with regard to the cytotoxic effect of lead nitrate. A dose-response relationship was also recorded with respect to the induction of stress genes in HepG2 cells exposed to lead nitrate. Overall, six out of the thirteen recombinant cell lines tested showed inductions to statistically significant levels (p < 0.05). At 50 microg/mL of lead nitrate, the average fold inductions were: 2.1 +/- 1.0, 5.4 +/- 0.4, 12.1 +/- 6.2, 5.0 +/- 1.7, 2.5 +/- 1.3, and 4.8 +/- 4.5 for XRE, HSP70, CRE, GADD153, and GRP78, respectively. These results indicate the potential for lead nitrate to undergo biotransformation in the liver (XRE), to cause cell proliferation (c-fos), protein damage (HSP70, GRP78), metabolic perturbation (CRE), and growth arrest and DNA damage (GADD153). Marginal but not significant inductions were also obtained with the GSTYa (1.5 +/- 0.8), and GADD45 (5.7 +/- 8.1) promoters, and the NF-KB (2.0 +/- 1.7) response element, indicating the potential for oxidative stress. No significant inductions (p > 0.05) were recorded for CYP1A1, HMTIIA, p53RE, and RARE.
Collapse
Affiliation(s)
- Paul B Tchounwou
- Molecular Toxicology Research Laboratory, NIH-Center for Environmental Health, School of Science and Technology, Jackson State University, Jackson, MS 39217, USA.
| | | | | | | | | |
Collapse
|
180
|
Abstract
Nuclear stress granules are subnuclear compartments that form in response to heat shock and other stress stimuli. Although many components of nuclear stress granules have been identified, including HSF1 and pre-mRNA processing factors, their function remains a mystery. A paper in this issue describes the stress-induced transcriptional activation of one of the nuclear stress granule target sites, a heterochromatic region that has been considered silent (Jolly et al., 2004). These intriguing findings will certainly give the research of these structures a new twist.
Collapse
Affiliation(s)
- Anton Sandqvist
- Turku Centre for Biotechnology, BioCity, P.O. Box 123, FIN-20521 Turku, Finland
| | | |
Collapse
|
181
|
Hashikawa N, Sakurai H. Phosphorylation of the yeast heat shock transcription factor is implicated in gene-specific activation dependent on the architecture of the heat shock element. Mol Cell Biol 2004; 24:3648-59. [PMID: 15082761 PMCID: PMC387759 DOI: 10.1128/mcb.24.9.3648-3659.2004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Heat shock transcription factor (HSF) binds to the heat shock element (HSE) and regulates transcription, where the divergence of HSE architecture provides gene- and stress-specific responses. The phosphorylation state of HSF, regulated by stress, is involved in the activation and inactivation of the transcription activation function. A domain designated as CTM (C-terminal modulator) of the Saccharomyces cerevisiae HSF is required for the activation of genes containing atypical HSE but not typical HSE. Here, we demonstrate that CTM function is conserved among yeast HSFs and is necessary not only for HSE-specific activation but also for the hyperphosphorylation of HSF upon heat shock. Moreover, both transcription and phosphorylation defects due to CTM mutations were restored concomitantly by a set of intragenic suppressor mutations. Therefore, the hyperphosphorylation of HSF is correlated with the activation of genes with atypical HSE but is not involved in that of genes with typical HSE. The function of CTM was circumvented in an HSF derivative lacking CE2, a yeast-specific repression domain. Taken together, we suggest that CTM alleviates repression by CE2, which allows HSF to be heat-inducibly phosphorylated and presume that phosphorylation is a prerequisite for the activator function of HSF when it binds to an atypical HSE.
Collapse
Affiliation(s)
- Naoya Hashikawa
- School of Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-0942, Japan
| | | |
Collapse
|
182
|
Boellmann F, Guettouche T, Guo Y, Fenna M, Mnayer L, Voellmy R. DAXX interacts with heat shock factor 1 during stress activation and enhances its transcriptional activity. Proc Natl Acad Sci U S A 2004; 101:4100-5. [PMID: 15016915 PMCID: PMC384701 DOI: 10.1073/pnas.0304768101] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
DAXX, a modulator of apoptosis and a repressor of basal transcription, was identified in a two-hybrid screen as a protein capable of interacting with a trimeric form of human heat shock factor 1 (HSF1). In human cells, DAXX interacted with HSF1 essentially only during stress, i.e., when factor trimerization occurred. Several lines of experimentation suggested that DAXX is an important mediator of HSF1 activation: (i) overexpression of DAXX enhanced basal transactivation competence of HSF1 in the absence of a stress; (ii) a DAXX fragment exerted dominant-negative effects on HSF1 activation by different types of stress; (iii) induction of heat shock or stress protein (HSP)70 by heat stress was defective in a cell line lacking functional DAXX; and (iv) RNA interference depletion of DAXX also substantially reduced heat induction of HSF1 activity and HSP70 expression. HSF1 transactivation competence is repressed by an HSP90-containing multichaperone complex that interacts with trimeric factor. Overexpressed HSF1, known to be largely trimeric, only marginally increased HSF1 activity on its own but potentiated the activating effect of DAXX overexpression. Expression of a nonnative protein capable of competing for multichaperone complex also synergistically enhanced activation of HSF1 by DAXX. These observations suggest a model in which DAXX released from its nuclear stores during stress opposes repression of HSF1 transactivation competence by multichaperone complex through its interaction with trimerized HSF1. Our identification of DAXX as a mediator of HSF1 activation raises the question whether DAXX produces some of its pleiotropic effects through modulation of HSP levels.
Collapse
Affiliation(s)
- Frank Boellmann
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | |
Collapse
|
183
|
Voellmy R. On mechanisms that control heat shock transcription factor activity in metazoan cells. Cell Stress Chaperones 2004; 9:122-33. [PMID: 15497499 PMCID: PMC1065292 DOI: 10.1379/csc-14r.1] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Accepted: 03/29/2004] [Indexed: 12/31/2022] Open
Abstract
Heat shock factor Hsf in nonvertebrate animals and homologous heat shock factor Hsf1 in vertebrate animals are key transcriptional regulators of the stress protein response. Hsf/Hsf1 is constitutively present in cells but is, typically, only active during periods during which cells are experiencing a physical or chemical proteotoxic stress. It has become increasingly clear that regulation of Hsf/Hsf1 activity occurs at multiple levels: the oligomeric status of Hsf/Hsf1, its DNA-binding ability, posttranslational modification, transcriptional competence, nuclear/ subnuclear localization, as well as its interactions with regulatory cofactors or other transcription factors all appear to be carefully controlled. This review emphasizes work reported over the past several years suggesting that regulation at several of these levels is mediated by repressive interactions of Hsp90-containing multichaperone complexes and/or individual chaperones and Hsf/Hsf1.
Collapse
Affiliation(s)
- Richard Voellmy
- Department of Biochemistry and Molecular Biology, University of Miami, Gautier Building, Room 403, 1011 NW 15th Street, Miami, FL 33136, USA.
| |
Collapse
|
184
|
Hilgarth RS, Murphy LA, O'Connor CM, Clark JA, Park-Sarge OK, Sarge KD. Identification of Xenopus heat shock transcription factor-2: conserved role of sumoylation in regulating deoxyribonucleic acid-binding activity of heat shock transcription factor-2 proteins. Cell Stress Chaperones 2004; 9:214-20. [PMID: 15497507 PMCID: PMC1065300 DOI: 10.1379/csc-8r.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Revised: 03/16/2004] [Accepted: 04/15/2004] [Indexed: 11/24/2022] Open
Abstract
Heat shock transcription factor (Hsf)-1 and Hsf2 are members of the heat shock factor (HSF) protein family involved in heat shock protein (hsp) gene regulation, a regulation that is critical for the ability of cells to survive exposure to stress conditions. Although the role of Hsf1 in binding and activating transcription of hsp gene promoters in response to cell stress is well established, how Hsf2 enhances stress-induced hsp expression is not understood. To gain an insight into the critical conserved features of the regulation and function of Hsf2, we have identified and characterized the Hsf2 protein from Xenopus laevis. We found that, similar to its human counterpart, Xenopus Hsf2 is sumoylated at lysine 82 and that, as it does in human Hsf2, the modification event of the small ubiquitin-related modifier 1 functions to increase the deoxyribonucleic acid-binding activity of this transcription factor in Xenopus. These results indicate that sumoylation is an evolutionarily conserved modification of Hsf2 proteins, supporting the position of this modification as a critical regulator of Hsf2 function.
Collapse
Affiliation(s)
- Roland S Hilgarth
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA
| | | | | | | | | | | |
Collapse
|
185
|
Jolly C, Metz A, Govin J, Vigneron M, Turner BM, Khochbin S, Vourc'h C. Stress-induced transcription of satellite III repeats. ACTA ACUST UNITED AC 2003; 164:25-33. [PMID: 14699086 PMCID: PMC2171959 DOI: 10.1083/jcb.200306104] [Citation(s) in RCA: 240] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Exposure of mammalian cells to stress induces the activation of heat shock transcription factor 1 (HSF1) and the subsequent transcription of heat shock genes. Activation of the heat shock response also correlates with a rapid relocalization of HSF1 within a few nuclear structures termed nuclear stress granules. These stress-induced structures, which form primarily on the 9q12 region in humans through direct binding of HSF1 to satellite III repeats, do not colocalize with transcription sites of known hsp genes. In this paper, we show that nuclear stress granules correspond to RNA polymerase II transcription factories where satellite III repeats are transcribed into large and stable RNAs that remain associated with the 9q12 region, even throughout mitosis. This work not only reveals the existence of a new major heat-induced transcript in human cells that may play a role in chromatin structure, but also provides evidence for a transcriptional activity within a locus considered so far as heterochromatic and silent.
Collapse
Affiliation(s)
- Caroline Jolly
- INSERM U309, Institut A. Bonniot, 38706 La Tronche cedex, France.
| | | | | | | | | | | | | |
Collapse
|
186
|
Trinklein ND, Murray JI, Hartman SJ, Botstein D, Myers RM. The role of heat shock transcription factor 1 in the genome-wide regulation of the mammalian heat shock response. Mol Biol Cell 2003; 15:1254-61. [PMID: 14668476 PMCID: PMC363119 DOI: 10.1091/mbc.e03-10-0738] [Citation(s) in RCA: 256] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Previous work has implicated heat shock transcription factor 1 (HSF1) as the primary transcription factor responsible for the transcriptional response to heat stress in mammalian cells. We characterized the heat shock response of mammalian cells by measuring changes in transcript levels and assaying binding of HSF1 to promoter regions for candidate heat shock genes chosen by a combination of genome-wide computational and experimental methods. We found that many heat-inducible genes have HSF1 binding sites (heat shock elements, HSEs) in their promoters that are bound by HSF1. Surprisingly, for 24 heat-inducible genes, we detected no HSEs and no HSF1 binding. Furthermore, of 182 promoters with likely HSE sequences, we detected HSF1 binding at only 94 of these promoters. Also unexpectedly, we found 48 genes with HSEs in their promoters that are bound by HSF1 but that nevertheless did not show induction after heat shock in the cell types we examined. We also studied the transcriptional response to heat shock in fibroblasts from mice lacking the HSF1 gene. We found 36 genes in these cells that are induced by heat as well as they are in wild-type cells. These results provide evidence that HSF1 does not regulate the induction of every transcript that accumulates after heat shock, and our results suggest that an independent posttranscriptional mechanism regulates the accumulation of a significant number of transcripts.
Collapse
Affiliation(s)
- Nathan D Trinklein
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305-5120, USA
| | | | | | | | | |
Collapse
|
187
|
Wang X, Grammatikakis N, Siganou A, Calderwood SK. Regulation of molecular chaperone gene transcription involves the serine phosphorylation, 14-3-3 epsilon binding, and cytoplasmic sequestration of heat shock factor 1. Mol Cell Biol 2003; 23:6013-26. [PMID: 12917326 PMCID: PMC180972 DOI: 10.1128/mcb.23.17.6013-6026.2003] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2002] [Revised: 02/04/2003] [Accepted: 05/13/2003] [Indexed: 11/20/2022] Open
Abstract
Heat shock factor 1 (HSF1) regulates the transcription of molecular chaperone hsp genes. However, the cellular control mechanisms that regulate HSF1 activity are not well understood. In this study, we have demonstrated for the first time that human HSF1 binds to the essential cell signaling protein 14-3-3 epsilon. Binding of HSF1 to 14-3-3 epsilon occurs in cells in which extracellular signal regulated kinase (ERK) is activated and blockade of the ERK pathway by treatment with the specific ERK pathway inhibitor PD98059 in vivo strongly suppresses the binding. We previously showed that ERK1 phosphorylates HSF1 on serine 307 and leads to secondary phosphorylation by glycogen synthase kinase 3 (GSK3) on serine 303 within the regulatory domain and that these phosphorylation events repress HSF1. We show here that HSF1 binding to 14-3-3 epsilon requires HSF1 phosphorylation on serines 303 and 307. Furthermore, the serine phosphorylation-dependent binding of HSF1 to 14-3-3 epsilon results in the transcriptional repression of HSF1 and its sequestration in the cytoplasm. Leptomycin B, a specific inhibitor of nuclear export receptor CRM1, was found to reverse the cytoplasmic sequestration of HSF1 mediated by 14-3-3 epsilon, suggesting that CRM1/14-3-3 epsilon directed nuclear export plays a major role in repression of HSF1 by the ERK/GSK3/14-3-3 epsilon pathway. Our experiments indicate a novel pathway for HSF1 regulation and suggest a mechanism for suppression of its activity during cellular proliferation.
Collapse
Affiliation(s)
- XiaoZhe Wang
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
188
|
Abstract
Amongst the families of intracellular molecules that chaperone and assist with the trafficking of other proteins, notably during conditions of cellular stress, heat shock protein (hsp) 70 is one of the most studied. Although its name suggests that expression is exclusively induced during cellular hyperthermia, members of the hsp70 family of proteins can be constitutively expressed and/or induced by a range of other cellular insults. The ubiquitous presence of hsp70 in eukaryotic and prokaryotic cells, combined with its high degree of sequence homology and intrinsic immunogenicity, have prompted the suggestion that inappropriate immune reactivity to hsp70 might lead to pro-inflammatory responses and the development of autoimmune disease. Indeed, hsp70 has been shown to be a potent activator of innate immunity and aberrant expression of hsp70 in certain organs promotes immunopathology. However, studies also suggest that hsp70 might have immunotherapeutic potential, as hsp70 purified from malignant and virally infected cells can transfer and deliver antigenic peptides to antigen-presenting cells to elicit peptide-specific immunity and, in contrast to its reported pro-inflammatory effects, the administration of recombinant hsp70 can attenuate experimental autoimmune disease. This review focuses on the immunoregulatory capacity of hsp70 and its potential therapeutic value.
Collapse
Affiliation(s)
- Stephen M Todryk
- Immune Regulation Research Group, Department of Biochemistry, Trinity College, Dublin, Ireland.
| | | | | |
Collapse
|
189
|
Inouye S, Katsuki K, Izu H, Fujimoto M, Sugahara K, Yamada SI, Shinkai Y, Oka Y, Katoh Y, Nakai A. Activation of heat shock genes is not necessary for protection by heat shock transcription factor 1 against cell death due to a single exposure to high temperatures. Mol Cell Biol 2003; 23:5882-95. [PMID: 12897157 PMCID: PMC166333 DOI: 10.1128/mcb.23.16.5882-5895.2003] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Heat shock response, which is characterized by the induction of a set of heat shock proteins, is essential for induced thermotolerance and is regulated by heat shock transcription factors (HSFs). Curiously, HSF1 is essential for heat shock response in mammals, whereas in avian HSF3, an avian-specific factor is required for the burst activation of heat shock genes. Amino acid sequences of chicken HSF1 are highly conserved with human HSF1, but those of HSF3 diverge significantly. Here, we demonstrated that chicken HSF1 lost the ability to activate heat shock genes through the amino-terminal domain containing an alanine-rich sequence and a DNA-binding domain. Surprisingly, chicken and human HSF1 but not HSF3 possess a novel function that protects against a single exposure to mild heat shock, which is not mediated through the activation of heat shock genes. Overexpression of HSF1 mutants that could not bind to DNA did not restore the susceptibility to cell death in HSF1-null cells, suggesting that the new protective role of HSF1 is mediated through regulation of unknown target genes other than heat shock genes. These results uncover a novel role of vertebrate HSF1, which has been masked under the roles of heat shock proteins.
Collapse
Affiliation(s)
- Sachiye Inouye
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube 755-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Metzler B, Abia R, Ahmad M, Wernig F, Pachinger O, Hu Y, Xu Q. Activation of heat shock transcription factor 1 in atherosclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:1669-76. [PMID: 12707051 PMCID: PMC1851193 DOI: 10.1016/s0002-9440(10)64301-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Previous work established that increased expression of heat shock proteins (HSPs) in the vessel wall might evoke proinflammatory and autoimmune reactions in the pathogenesis of atherosclerosis. The present study was designed to further scrutinize the molecular mechanisms of HSP expression involving activation of heat shock transcription factors (HSFs) in atherosclerotic lesions in animal models. Severe atherosclerotic lesions developed in the aortas of rabbits 16 weeks after feeding a 0.2% cholesterol diet. When protein extracts from the aortas were subjected to Western blot analysis, the level of HSF1 in proteins from atherosclerotic lesions of hypercholesterolemic rabbits were significantly higher than those of normal vessels. Gel mobility shift assays revealed the formation of protein-heat shock element complexes containing HSF1 in protein extracts from atherosclerotic lesion. Furthermore, triglyceride-rich lipoprotein, oxidized-triglyceride-rich lipoprotein, low-density lipoprotein, and oxidized low-density lipoprotein did not activate HSF1 in cultured smooth muscle cells, whereas HSF1 was highly activated in cells treated with tumor necrosis factor-alpha. Interestingly, mechanical stretching of smooth muscle cells resulted in HSF1 translocation from the cytoplasm to the nucleus and hyperphosphorylation followed by increased HSP70 expression. Thus, our findings provide the first evidence that HSF1 is activated and highly expressed in atherosclerotic lesions and that cytokine stimulation and disturbed mechanical stress to the vessel wall may be responsible for such activation.
Collapse
Affiliation(s)
- Bernhard Metzler
- Department of Internal Medicine, Division of Cardiology, University Hospital of Innsbruck, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
191
|
Hietakangas V, Ahlskog JK, Jakobsson AM, Hellesuo M, Sahlberg NM, Holmberg CI, Mikhailov A, Palvimo JJ, Pirkkala L, Sistonen L. Phosphorylation of serine 303 is a prerequisite for the stress-inducible SUMO modification of heat shock factor 1. Mol Cell Biol 2003; 23:2953-68. [PMID: 12665592 PMCID: PMC152542 DOI: 10.1128/mcb.23.8.2953-2968.2003] [Citation(s) in RCA: 231] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2002] [Revised: 08/29/2002] [Accepted: 01/27/2003] [Indexed: 01/04/2023] Open
Abstract
The heat shock response, which is accompanied by a rapid and robust upregulation of heat shock proteins (Hsps), is a highly conserved protection mechanism against protein-damaging stress. Hsp induction is mainly regulated at transcriptional level by stress-inducible heat shock factor 1 (HSF1). Upon activation, HSF1 trimerizes, binds to DNA, concentrates in the nuclear stress granules, and undergoes a marked multisite phosphorylation, which correlates with its transcriptional activity. In this study, we show that HSF1 is modified by SUMO-1 and SUMO-2 in a stress-inducible manner. Sumoylation is rapidly and transiently enhanced on lysine 298, located in the regulatory domain of HSF1, adjacent to several critical phosphorylation sites. Sumoylation analyses of HSF1 phosphorylation site mutants reveal that specifically the phosphorylation-deficient S303 mutant remains devoid of SUMO modification in vivo and the mutant mimicking phosphorylation of S303 promotes HSF1 sumoylation in vitro, indicating that S303 phosphorylation is required for K298 sumoylation. This finding is further supported by phosphopeptide mapping and analysis with S303/7 phosphospecific antibodies, which demonstrate that serine 303 is a target for strong heat-inducible phosphorylation, corresponding to the inducible HSF1 sumoylation. A transient phosphorylation-dependent colocalization of HSF1 and SUMO-1 in nuclear stress granules provides evidence for a strictly regulated subnuclear interplay between HSF1 and SUMO.
Collapse
Affiliation(s)
- Ville Hietakangas
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University and Department of Biochemistry and Food Chemistry, University of Turku, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Ahn SG, Thiele DJ. Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress. Genes Dev 2003; 17:516-28. [PMID: 12600944 PMCID: PMC195992 DOI: 10.1101/gad.1044503] [Citation(s) in RCA: 328] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The activation of eukaryotic heat shock protein (Hsp) gene expression occurs in response to a wide variety of cellular stresses including heat shock, hydrogen peroxide, uncoupled oxidative phosphorylation, infection, and inflammation. Biochemical and genetic studies have clearly demonstrated critical roles for mammalian heat shock factor 1 (HSF1) in stress-inducible Hsp gene expression, resistance to stress-induced programmed cell death, extra-embryonic development, and other biological functions. Activation of mammalian Hsp gene expression involves the stress-inducible conversion of HSF1 from the inactive monomer to the DNA-binding competent homotrimer. Although Hsp activation is a central conserved process in biology, the precise mechanisms for stress sensing and signaling to activate HSF1, and the mechanisms by which many distinct stresses activate HSF1, are poorly understood. In this report we demonstrate that recombinant mammalian HSF1 directly senses both heat and hydrogen peroxide to assemble into a homotrimer in a reversible and redox-regulated manner. The sensing of both stresses requires two cysteine residues within the HSF1 DNA-binding domain that are engaged in redox-sensitive disulfide bonds. HSF1 derivatives in which either or both cysteines were mutated are defective in stress-inducible trimerization and DNA binding, stress-inducible nuclear translocation and Hsp gene trans-activation, and in the protection of mouse cells from stress-induced apoptosis. This redox-dependent activation of HSF1 by heat and hydrogen peroxide establishes a common mechanism in the stress activation of Hsp gene expression by mammalian HSF1.
Collapse
Affiliation(s)
- Sang-Gun Ahn
- Department of Biological Chemistry, The University of Michigan Medical School, Ann Arbor, Michigan 48109-0606, USA
| | | |
Collapse
|
193
|
Kallio M, Chang Y, Manuel M, Alastalo TP, Rallu M, Gitton Y, Pirkkala L, Loones MT, Paslaru L, Larney S, Hiard S, Morange M, Sistonen L, Mezger V. Brain abnormalities, defective meiotic chromosome synapsis and female subfertility in HSF2 null mice. EMBO J 2002; 21:2591-601. [PMID: 12032072 PMCID: PMC125382 DOI: 10.1093/emboj/21.11.2591] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Heat shock factor 2, one of the four vertebrate HSFs, transcriptional regulators of heat shock gene expression, is active during embryogenesis and spermatogenesis, with unknown functions and targets. By disrupting the Hsf2 gene, we show that, although the lack of HSF2 is not embryonic lethal, Hsf2(-/-) mice suffer from brain abnormalities, and meiotic and gameto genesis defects in both genders. The disturbances in brain are characterized by the enlargement of lateral and third ventricles and the reduction of hippocampus and striatum, in correlation with HSF2 expression in proliferative cells of the neuroepithelium and in some ependymal cells in adults. Many developing spermatocytes are eliminated via apoptosis in a stage-specific manner in Hsf2(-/-) males, and pachytene spermatocytes also display structural defects in the synaptonemal complexes between homologous chromosomes. Hsf2(-/-) females suffer from multiple fertility defects: the production of abnormal eggs, the reduction in ovarian follicle number and the presence of hemorrhagic cystic follicles are consistent with meiotic defects. Hsf2(-/-) females also display hormone response defects, that can be rescued by superovulation treatment, and exhibit abnormal rates of luteinizing hormone receptor mRNAs.
Collapse
Affiliation(s)
- Marko Kallio
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Yunhua Chang
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Martine Manuel
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Tero-Pekka Alastalo
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Murielle Rallu
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Yorick Gitton
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Lila Pirkkala
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Marie-Thérèse Loones
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Liliana Paslaru
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Severine Larney
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Sophie Hiard
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Michel Morange
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Lea Sistonen
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| | - Valérie Mezger
- Turku Centre for Biotechnology, University of Turku, Abo Akademi University, FIN-20520 Turku, Department of Biology, Abo Akademi University, Turku, Finland, UMR8541 and Animal Facilities, Ecole Normale Supérieure, F-75230 Paris cedex 05, France Present address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA Present address: Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK Present address: Developmental Genetics Program, Skirball Institute for Biomolecular Medicine, NYU Medical Center, New York,NY 10016, USA Corresponding author e-mail: M.Kallio, Y.Chang and M.Manuel contributed equally to this work
| |
Collapse
|
194
|
Jolly C, Konecny L, Grady DL, Kutskova YA, Cotto JJ, Morimoto RI, Vourc'h C. In vivo binding of active heat shock transcription factor 1 to human chromosome 9 heterochromatin during stress. J Cell Biol 2002; 156:775-81. [PMID: 11877455 PMCID: PMC2173303 DOI: 10.1083/jcb.200109018] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Activation of the mammalian heat shock transcription factor (HSF)1 by stress is a multistep process resulting in the transcription of heat shock genes. Coincident with these events is the rapid and reversible redistribution of HSF1 to discrete nuclear structures termed HSF1 granules, whose function is still unknown. Key features are that the number of granules correlates with cell ploidy, suggesting the existence of a chromosomal target. Here we show that in humans, HSF1 granules localize to the 9q11-q12 heterochromatic region. Within this locus, HSF1 binds through direct DNA-protein interaction with a nucleosome-containing subclass of satellite III repeats. HSF1 granule formation only requires the DNA binding competence and the trimerization of the factor. This is the first example of a transcriptional activator that accumulates transiently and reversibly on a chromosome-specific heterochromatic locus.
Collapse
Affiliation(s)
- Caroline Jolly
- DyOGen, INSERM U309, Institut A. Bonniot, Domaine de la Merci, 38706 La Tronche cedex, France.
| | | | | | | | | | | | | |
Collapse
|
195
|
Mathew A, Mathur SK, Jolly C, Fox SG, Kim S, Morimoto RI. Stress-specific activation and repression of heat shock factors 1 and 2. Mol Cell Biol 2001; 21:7163-71. [PMID: 11585899 PMCID: PMC99891 DOI: 10.1128/mcb.21.21.7163-7171.2001] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vertebrate cells express a family of heat shock transcription factors (HSF1 to HSF4) that coordinate the inducible regulation of heat shock genes in response to diverse signals. HSF1 is potent and activated rapidly though transiently by heat shock, whereas HSF2 is a less active transcriptional regulator but can retain its DNA binding properties for extended periods. Consequently, the differential activation of HSF1 and HSF2 by various stresses may be critical for cells to survive repeated and diverse stress challenges and to provide a mechanism for more precise regulation of heat shock gene expression. Here we show, using a novel DNA binding and detection assay, that HSF1 and HSF2 are coactivated to different levels in response to a range of conditions that cause cell stress. Above a low basal activity of both HSFs, heat shock preferentially activates HSF1, whereas the amino acid analogue azetidine or the proteasome inhibitor MG132 coactivates both HSFs to different levels and hemin preferentially induces HSF2. Unexpectedly, we also found that heat shock has dramatic adverse effects on HSF2 that lead to its reversible inactivation coincident with relocalization from the nucleus. The reversible inactivation of HSF2 is specific to heat shock and does not occur with other stressors or in cells expressing high levels of heat shock proteins. These results reveal that HSF2 activity is negatively regulated by heat and suggest a role for heat shock proteins in the positive regulation of HSF2.
Collapse
Affiliation(s)
- A Mathew
- Department of Biochemistry, Molecular Biology, and Cell Biology, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | | | | | |
Collapse
|
196
|
Molina A, Di Martino E, Martial JA, Muller M. Heat shock stimulation of a tilapia heat shock protein 70 promoter is mediated by a distal element. Biochem J 2001; 356:353-9. [PMID: 11368761 PMCID: PMC1221845 DOI: 10.1042/0264-6021:3560353] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We reported previously that a tilapia (Oreochromis mossambicus) heat shock protein 70 (HSP70) promoter is able to confer heat shock response on a reporter gene after transient expression both in cell culture and in microinjected zebrafish embryos. Here we present the first functional analysis of a fish HSP70 promoter, the tiHSP70 promoter. Using transient expression experiments in carp EPC (epithelioma papulosum cyprini) cells and in microinjected zebrafish embryos, we show that a distal heat shock response element (HSE1) at approx. -800 is predominantly responsible for the heat shock response of the tiHSP70 promoter. This element specifically binds an inducible transcription factor, most probably heat shock factor, and a constitutive factor. The constitutive complex is not observed with the non-functional, proximal HSE3 sequence, suggesting that both factors are required for the heat shock response mediated by HSE1.
Collapse
Affiliation(s)
- A Molina
- Laboratoire de Biologie Moléculaire et Génie Génétique, Université de Liège, Institut de Chimie B6, B-4000 Sart-Tilman, Belgium
| | | | | | | |
Collapse
|
197
|
Holmberg CI, Hietakangas V, Mikhailov A, Rantanen JO, Kallio M, Meinander A, Hellman J, Morrice N, MacKintosh C, Morimoto RI, Eriksson JE, Sistonen L. Phosphorylation of serine 230 promotes inducible transcriptional activity of heat shock factor 1. EMBO J 2001; 20:3800-10. [PMID: 11447121 PMCID: PMC125548 DOI: 10.1093/emboj/20.14.3800] [Citation(s) in RCA: 231] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Heat shock factor 1 (HSF1) is a serine-rich constitutively phosphorylated mediator of the stress response. Upon stress, HSF1 forms DNA-binding trimers, relocalizes to nuclear granules, undergoes inducible phosphorylation and acquires the properties of a transactivator. HSF1 is phosphorylated on multiple sites, but the sites and their function have remained an enigma. Here, we have analyzed sites of endogenous phosphorylation on human HSF1 and developed a phosphopeptide antibody to identify Ser230 as a novel in vivo phosphorylation site. Ser230 is located in the regulatory domain of HSF1, and promotes the magnitude of the inducible transcriptional activity. Ser230 lies within a consensus site for calcium/calmodulin-dependent protein kinase II (CaMKII), and CaMKII overexpression enhances both the level of in vivo Ser230 phosphorylation and transactivation of HSF1. The importance of Ser230 was further established by the S230A HSF1 mutant showing markedly reduced activity relative to wild-type HSF1 when expressed in hsf1(-/-) cells. Our study provides the first evidence that phosphorylation is essential for the transcriptional activity of HSF1, and hence for induction of the heat shock response.
Collapse
Affiliation(s)
- Carina I. Holmberg
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Ville Hietakangas
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Andrey Mikhailov
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Jouni O. Rantanen
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Marko Kallio
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Annika Meinander
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Jukka Hellman
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Nick Morrice
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Carol MacKintosh
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Richard I. Morimoto
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - John E. Eriksson
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| | - Lea Sistonen
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Department of Biochemistry and Food Chemistry and Department of Biology, University of Turku, Department of Biology, Åbo Akademi University, Finland, MRC Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee, UK and Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, IL, USA Corresponding author at Turku Centre for Biotechnology, PO Box 123, FIN-20521 Turku, Finland e-mail:
| |
Collapse
|
198
|
Nakai A, Ishikawa T. Cell cycle transition under stress conditions controlled by vertebrate heat shock factors. EMBO J 2001; 20:2885-95. [PMID: 11387221 PMCID: PMC125499 DOI: 10.1093/emboj/20.11.2885] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The roles of heat shock transcription factors (HSFs) under physiological conditions have recently become the focus of intense study. We generated avian cells lacking two heat-inducible HSFs, HSF1 and HSF3. In addition to complete loss of activation of heat shock genes under stress conditions, these cells exhibited a marked reduction in Hsp90alpha expression under normal growth conditions. Reduction in Hsp90alpha expression caused instability of a cyclin-dependent kinase, Cdc2, and cell cycle progression was blocked mainly at the G2 phase, but also at G1 phase even at mild heat shock temperatures. Restoration of Hsp90alpha expression rescued the temperature sensitivity without induction of HSPS: We demonstrated for the first time a molecular target affected by heat shock in vivo that causes cell cycle arrest in vertebrates and a novel mechanism of stress resistance controlled by vertebrate HSFS:
Collapse
Affiliation(s)
- A Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minamikogushi 1-1-1, Ube 775-8505, Japan.
| | | |
Collapse
|
199
|
Hasday JD, Singh IS. Fever and the heat shock response: distinct, partially overlapping processes. Cell Stress Chaperones 2001. [PMID: 11189454 DOI: 10.1379/1466-1268(2000)005<0471:fathsr>2.0.co;2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The heat shock response is an ancient and highly conserved process that is essential for surviving 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 studies showing that fever is beneficial in the infected host. We show that core temperatures achieved during fever can activate the heat shock response and discuss some of the biochemical consequences of such an effect. We present data suggesting 4 possible mechanisms by which fever might confer protection: (1) directly killing or inhibiting growth of pathogens; (2) inducing cytoprotective heat shock proteins (Hsps) in host cells; (3) inducing expression of pathogen Hsps, an activator of host defenses; and (4) modifying and orchestrating host defenses. Two of these mechanisms directly involve the heat shock response. We describe how heat shock factor-1, the predominant heat-induced transcriptional enhancer not only activates transcription of Hsps but also regulates expression of pivotal cytokines and early response genes. The relationship between fever and the heat shock response is an illuminating example of how a more recently evolved response might exploit preexisting biochemical pathways for a new function.
Collapse
Affiliation(s)
- J D Hasday
- Department of Medicine, University of Maryland School of Medicine and the Medicine and Research Services of the Baltimore VA Medical Center, 21201, USA.
| | | |
Collapse
|
200
|
Abstract
The heat shock response is an ancient and highly conserved process that is essential for surviving 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 studies showing that fever is beneficial in the infected host. We show that core temperatures achieved during fever can activate the heat shock response and discuss some of the biochemical consequences of such an effect. We present data suggesting 4 possible mechanisms by which fever might confer protection: (1) directly killing or inhibiting growth of pathogens; (2) inducing cytoprotective heat shock proteins (Hsps) in host cells; (3) inducing expression of pathogen Hsps, an activator of host defenses; and (4) modifying and orchestrating host defenses. Two of these mechanisms directly involve the heat shock response. We describe how heat shock factor-1, the predominant heat-induced transcriptional enhancer not only activates transcription of Hsps but also regulates expression of pivotal cytokines and early response genes. The relationship between fever and the heat shock response is an illuminating example of how a more recently evolved response might exploit preexisting biochemical pathways for a new function.
Collapse
Affiliation(s)
- J D Hasday
- Department of Medicine, University of Maryland School of Medicine and the Medicine and Research Services of the Baltimore VA Medical Center, 21201, USA.
| | | |
Collapse
|