251
|
Shamovsky I, Ivannikov M, Kandel ES, Gershon D, Nudler E. RNA-mediated response to heat shock in mammalian cells. Nature 2006; 440:556-60. [PMID: 16554823 DOI: 10.1038/nature04518] [Citation(s) in RCA: 271] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 12/15/2005] [Indexed: 12/16/2022]
Abstract
The heat-shock transcription factor 1 (HSF1) has an important role in the heat-shock response in vertebrates by inducing the expression of heat-shock proteins (HSPs) and other cytoprotective proteins. HSF1 is present in unstressed cells in an inactive monomeric form and becomes activated by heat and other stress stimuli. HSF1 activation involves trimerization and acquisition of a site-specific DNA-binding activity, which is negatively regulated by interaction with certain HSPs. Here we show that HSF1 activation by heat shock is an active process that is mediated by a ribonucleoprotein complex containing translation elongation factor eEF1A and a previously unknown non-coding RNA that we term HSR1 (heat shock RNA-1). HSR1 is constitutively expressed in human and rodent cells and its homologues are functionally interchangeable. Both HSR1 and eEF1A are required for HSF1 activation in vitro; antisense oligonucleotides or short interfering (si)RNA against HSR1 impair the heat-shock response in vivo, rendering cells thermosensitive. The central role of HSR1 during heat shock implies that targeting this RNA could serve as a new therapeutic model for cancer, inflammation and other conditions associated with HSF1 deregulation.
Collapse
Affiliation(s)
- Ilya Shamovsky
- Department of Biochemistry, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
252
|
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
|
253
|
Hashikawa N, Mizukami Y, Imazu H, Sakurai H. Mutated Yeast Heat Shock Transcription Factor Activates Transcription Independently of Hyperphosphorylation. J Biol Chem 2006; 281:3936-42. [PMID: 16361698 DOI: 10.1074/jbc.m510827200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The homotrimeric heat shock transcription factor (HSF) binds to the heat shock element of target genes and regulates transcription in response to various stresses. The Hsf1 protein of Saccharomyces cerevisiae is extensively phosphorylated upon heat shock; a modification that is under positive regulation by its C-terminal regulatory domain (CTM). Hyperphosphorylation has been implicated in gene-specific transcriptional activation. Here, we surveyed genes whose heat shock response is reduced by a CTM mutation. The CTM is indispensable for transcription via heat shock elements bound by a single Hsf1 trimer but is dispensable for transcription via heat shock elements bound by Hsf1 trimers in a cooperative manner. Intragenic mutations located within or near the wing region of the winged helix-turn-helix DNA-binding domain suppress the temperature-sensitive growth phenotype associated with the CTM mutation and enable Hsf1 to activate transcription independently of hyperphosphorylation. Deletion of the wing partially restores the transcriptional defects of the unphosphorylated Hsf1. These results demonstrate a functional link between hyperphosphorylation and the wing region and suggest that this modification is involved in a conformational change of a single Hsf1 trimer to an active form.
Collapse
Affiliation(s)
- Naoya Hashikawa
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | | | | | | |
Collapse
|
254
|
Balogh G, Horváth I, Nagy E, Hoyk Z, Benkõ S, Bensaude O, Vígh L. The hyperfluidization of mammalian cell membranes acts as a signal to initiate the heat shock protein response. FEBS J 2006; 272:6077-86. [PMID: 16302971 DOI: 10.1111/j.1742-4658.2005.04999.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The concentrations of two structurally distinct membrane fluidizers, the local anesthetic benzyl alcohol (BA) and heptanol (HE), were used at concentrations so that their addition to K562 cells caused identical increases in the level of plasma membrane fluidity as tested by 1,6-diphenyl-1,3,5-hexatriene (DPH) anisotropy. The level of membrane fluidization induced by the chemical agents on isolated membranes at such concentrations corresponded to the membrane fluidity increase seen during a thermal shift up to 42 degrees C. The formation of isofluid membrane states in response to the administration of BA or HE resulted in almost identical downshifts in the temperature thresholds of the heat shock response, accompanied by increases in the expression of genes for stress proteins such as heat shock protein (HSP)-70 at the physiological temperature. Similarly to thermal stress, the exposure of the cells to these membrane fluidizers elicited nearly identical increases of cytosolic Ca2+ concentration in both Ca2+-containing and Ca2+-free media and also closely similar extents of increase in mitochondrial hyperpolarization. We obtained no evidence that the activation of heat shock protein expression by membrane fluidizers is induced by a protein-unfolding signal. We suggest, that the increase of fluidity in specific membrane domains, together with subsequent alterations in key cellular events are converted into signal(s) leading to activation of heat shock genes.
Collapse
Affiliation(s)
- Gábor Balogh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
255
|
Abstract
The heat shock response is triggered primarily by nonnative proteins accumulating in a stressed cell and results in increased expression of heat shock proteins (Hsps), i.e., of chaperones capable of participating in the refolding or elimination of nonnative proteins. Best known is the transcriptional part of this response that is mediated predominantly by heat shock factor 1 (HSF1). HSF1 activity is regulated at different levels by Hsps and co-chaperones and is modulated further by a number of mechanisms involving other stress-regulated aspects of cell metabolism.
Collapse
Affiliation(s)
- R Voellmy
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, FL 33136, USA.
| |
Collapse
|
256
|
Abstract
This chapter focuses on immunological effects of eukaryotic and microbial heat shock proteins (HSPs), with molecular weights of about 60, 70, and 90 kDa. The search for tumor-specific antigens resulted in the identification of HSPs. They have been found to elicit a potent anti-cancer immune response mediated by the adoptive and innate immune system. Following receptor-mediated uptake of HSP (HSP70 and gp96) peptide complexes by antigen-presenting cells and representation of HSP-chaperoned peptides by MHC class I molecules, a CD8-specific T cell response is induced. Apart from chaperoning immunogenic peptides derived from tumors, bacterial and virally infected cells, they by themselves provide activatory signals for antigen-presenting cells and natural killer (NK) cells. After binding of peptide-free HSP70 to Toll-like receptors, the secretion of pro-inflammatory cytokines is initiated by antigen-presenting cells and thus results in a nonspecific stimulation of the immune system. Moreover, soluble as well as cell membrane-bound HSP70 on tumor cells can directly activate the cytolytic and migratory capacity of NK cells. Apart form cancer, HSPs of different origins, with a molecular weight of about 60, 70, and 90 kDa, also play a pivotal role in viral infections, including human and simian immunodeficiency virus (HIV, SIV), measles, and choriomeningitis. Moreover, HSPs have been found to induce tolerance against autoimmune diseases. In summary, depending on their mode of induction, intracellular/extracellular location, cellular origin (eukaryote/prokaryote), peptide loading status, intracellular ADP/ATP content, concentration, and route of application, HSPs either exert immune activation as danger signals in cancer immunity and mediate protection against infectious diseases or exhibit regulatory activities in controlling and preventing autoimmunity.
Collapse
Affiliation(s)
- G Multhoff
- Department of Hematology and Oncology, University Hospital Regensburg, Germany.
| |
Collapse
|
257
|
Jurivich DA, Choo M, Welk J, Qiu L, Han K, Zhou X. Human aging alters the first phase of the molecular response to stress in T-cells. Exp Gerontol 2005; 40:948-58. [PMID: 16168601 DOI: 10.1016/j.exger.2005.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 08/01/2005] [Accepted: 08/05/2005] [Indexed: 10/25/2022]
Abstract
This study examines how age affects the first phase of the heat shock response in human T-cells. To understand how age alters transcriptional regulation of the heat shock genes, a cross-sectional study was conducted utilizing human T-cells enriched from peripheral blood lymphocytes of healthy young (20-40 years old) and old (>70 years old) donors. Nuclear run-on analysis revealed a 66% reduction in hsp70 transcription rates in old compared to young nuclei harvested from T-cells exposed to a brief 42 degrees C heat shock. To determine if one or more protein transactivators of the proximal and distal promoter regions of the hsp70 gene were affected by age, gel shift analysis was performed. Both HSF1 and SP1 DNA-binding were reduced with age but no reduction was noted in CCAAT-DNA binding. Western blot analysis indicated that HSF1 but not HSF2 protein levels were reduced in aged donor samples. These data suggest that human T-cell senescence involves a multi-factorial mechanism that diminishes an important transcriptional response to thermal stress. The results are discussed relative to recent studies that support a multi-factorial mechanism for age-dependent attenuation of the heat shock transcription factor.
Collapse
Affiliation(s)
- Donald A Jurivich
- Department of Medicine, Section of Geriatric Medicine (m/c 717), University of Illinois at Chicago & Jesse Brown VA Medical Center, 840 S. Wood St Chicago, IL 60612, USA.
| | | | | | | | | | | |
Collapse
|
258
|
Kim HP, Wang X, Zhang J, Suh GY, Benjamin IJ, Ryter SW, Choi AMK. Heat shock protein-70 mediates the cytoprotective effect of carbon monoxide: involvement of p38 beta MAPK and heat shock factor-1. THE JOURNAL OF IMMUNOLOGY 2005; 175:2622-9. [PMID: 16081837 DOI: 10.4049/jimmunol.175.4.2622] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Carbon monoxide (CO), a product of heme oxygenase activity, exerts antiapoptotic and anti-inflammatory effects in vitro and in vivo. The anti-inflammatory effects of CO involve the inhibition of TNF-alpha expression and the enhancement of IL-10 production, resulting in reduced mortality after endotoxin challenge. In this study we demonstrate for the first time that the protective effects of CO involve the increased expression of the 70-kDa inducible heat shock protein (Hsp70) in murine lung endothelial cells and fibroblasts. The p38beta MAPK mediated the effects of CO on cytoprotection and Hsp70 regulation. Suppression of Hsp70 expression and/or genetic deletion of heat shock factor-1, the principle transcriptional regulator of Hsp70, attenuated the cytoprotective and immunomodulatory effects of CO in mouse lung cells and in vivo. These data provide a novel mechanism for the protective effects of CO and underscore a potential application of this gaseous molecule in anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hong Pyo Kim
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
259
|
Corry GN, Underhill DA. Subnuclear compartmentalization of sequence-specific transcription factors and regulation of eukaryotic gene expression. Biochem Cell Biol 2005; 83:535-47. [PMID: 16094457 DOI: 10.1139/o05-062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To date, the majority of the research regarding eukaryotic transcription factors has focused on characterizing their function primarily through in vitro methods. These studies have revealed that transcription factors are essentially modular structures, containing separate regions that participate in such activities as DNA binding, protein-protein interaction, and transcriptional activation or repression. To fully comprehend the behavior of a given transcription factor, however, these domains must be analyzed in the context of the entire protein, and in certain cases the context of a multiprotein complex. Furthermore, it must be appreciated that transcription factors function in the nucleus, where they must contend with a variety of factors, including the nuclear architecture, chromatin domains, chromosome territories, and cell-cycle-associated processes. Recent examinations of transcription factors in the nucleus have clarified the behavior of these proteins in vivo and have increased our understanding of how gene expression is regulated in eukaryotes. Here, we review the current knowledge regarding sequence-specific transcription factor compartmentalization within the nucleus and discuss its impact on the regulation of such processes as activation or repression of gene expression and interaction with coregulatory factors.
Collapse
Affiliation(s)
- Gareth N Corry
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
260
|
Li D, Sánchez ER. Glucocorticoid receptor and heat shock factor 1: novel mechanism of reciprocal regulation. VITAMINS AND HORMONES 2005; 71:239-62. [PMID: 16112270 DOI: 10.1016/s0083-6729(05)71008-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Glucocorticoids control a host of bodily responses, ranging from carbohydrate metabolism in the liver to immunity and inflammation in the lymph system. In response to stress, glucocorticoid levels are known to rise-a response thought to provide a protective function against the stress event. It is now understood that the major function of glucocorticoids under stress is to protect not against the stress event itself but against overstimulation by host defenses (e.g., inflammation). Control of these responses is achieved by the glucocorticoid receptor, a member of the steroid receptor transcription factor family. The oldest, most conserved, and most ubiquitous of the stress responses is induced expression of heat shock proteins that act as chaperones against stress-induced denaturation of protein. Expression of heat shock protein genes is controlled by heat shock transcription factor 1. In this work, we review our observations and those of other laboratories demonstrating a relationship between the glucocorticoid and heat shock responses. We show that complex but reciprocal mechanisms of regulation occur between glucocorticoid receptor and heat shock transcription factor 1 and present a model of coordinated action that likely serves to fully reestablish homeostasis following stress.
Collapse
Affiliation(s)
- Dapei Li
- Department of Pharmacology, Medical College of Ohio, Toledo, Ohio 43614, USA
| | | |
Collapse
|
261
|
Kim SA, Yoon JH, Kim DK, Kim SG, Ahn SG. CHIP interacts with heat shock factor 1 during heat stress. FEBS Lett 2005; 579:6559-63. [PMID: 16293251 DOI: 10.1016/j.febslet.2005.10.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 10/21/2005] [Accepted: 10/21/2005] [Indexed: 11/16/2022]
Abstract
Heat shock factor 1 (HSF1) is a major transactivator of heat shock genes in response to stress and mediates cell protection against various harmful conditions. In this study, we identified the interaction of CHIP (carboxyl terminus of the heat shock cognate protein 70-interacting protein) with the N-terminus of HSF1. Using GST full-down assay, we found that CHIP directly interacts with C-terminal deleted HSF1 (a.a. 1-290) but not with full-length HSF1 under non-stressed conditions. Interestingly, interaction of CHIP with full-length HSF1 was induced by heat shock treatment. The structural change of HSF1 was observed under heat stressed conditions by CD spectra. These observations demonstrate the direct interaction between HSF1 and CHIP and this interaction requires conformational change of HSF1 by heat stress.
Collapse
Affiliation(s)
- Soo-A Kim
- Oral Biology Research Institute, Chosun University College of Dentistry, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, Republic of Korea
| | | | | | | | | |
Collapse
|
262
|
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
|
263
|
Saito K, Dai Y, Ohtsuka K. Enhanced expression of heat shock proteins in gradually dying cells and their release from necrotically dead cells. Exp Cell Res 2005; 310:229-36. [PMID: 16129430 DOI: 10.1016/j.yexcr.2005.07.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2005] [Revised: 07/19/2005] [Accepted: 07/28/2005] [Indexed: 10/25/2022]
Abstract
Heat shock proteins (HSPs) have molecular chaperone functions in protein biogenesis as well as cytoprotective functions against deleterious environmental stresses, and they work mainly inside of the cells. HSPs are usually induced in living cells that have been exposed to mild stresses or have recovered from severe stresses. Here, we show the enhanced synthesis of HSPs in gradually and necrotically dying cells that were treated with a high concentration of acrylamide (10 mM). This treatment caused irreversible cell death. The synthesis of HSPs, which was enhanced before cell death, was mediated by the activation of heat shock transcription factor 1 (HSF1); that is, the treatment led to the phosphorylation of HSF1, formation of characteristic HSF1 granules in the nucleus, and acquisition of DNA binding ability of HSF1. The induction of HSPs by acrylamide treatment was dependent on the consensus sequence of heat shock element (HSE) as demonstrated by a reporter assay. Also, several HSPs (Hsp90, Hsc70, Hsp70, Hsp60, Hsp47, Hsp40, and Hsp27) were detected outside of the cells after the treatment with acrylamide, indicating that these HSPs are released from necrotically dead cells. These results suggest that when cells are slowly and irreversibly dying, they augment the expression of HSPs and release them outside of the cells as a danger signal or dying messages.
Collapse
Affiliation(s)
- Kiyoto Saito
- Laboratory of Cell and Stress Biology, Department of Environmental Biology, Chubu University,1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | | | | |
Collapse
|
264
|
Vydra N, Malusecka E, Jarzab M, Lisowska K, Glowala-Kosinska M, Benedyk K, Widlak P, Krawczyk Z, Widlak W. Spermatocyte-specific expression of constitutively active heat shock factor 1 induces HSP70i-resistant apoptosis in male germ cells. Cell Death Differ 2005; 13:212-22. [PMID: 16151457 DOI: 10.1038/sj.cdd.4401758] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Spermatocytes, the most sensitive male germ cells to heat-induced apoptosis, do not respond to hyperthermia by inducing heat shock proteins (HSPs), including HSP70i, which has been previously shown to confer resistance to apoptosis in somatic cells. To dissect the mechanism of heat-induced apoptosis and to determine if we could protect spermatocytes by expressing HSP70i, we engineered transgenic mice that express in spermatocytes constitutively active heat shock transcription factor (HSF)1. Such HSF1 expression did not lead to transcription of inducible Hsp70 genes, but instead induced caspase-dependent apoptosis that mimicked heat shock-induced death of spermatogenic cells. Both mitochondria-dependent and death receptor-dependent pathways appear to be involved in such HSF1-induced apoptosis: the levels of Bcl-2 family proteins became increased, p53 protein accumulated and expression levels of caspase-8 and death-receptor-interacting proteins (including Fas-associated death domain protein and TNF receptor associated death domain protein) became elevated. Surprisingly, the constitutive spermatocyte-specific expression of HSP70i in double-transgenic males did not protect against such HSF1-induced apoptosis.
Collapse
Affiliation(s)
- N Vydra
- Department of Tumor Biology, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, 44-101 Gliwice, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Kumar R, Singh PK, Arora R, Sharma A, Prasad J, Sagar R, Singh S, Sharma RK. Radioprotection by Podophyllum hexandrum in the liver of mice: A mechanistic approach. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2005; 20:326-334. [PMID: 21783608 DOI: 10.1016/j.etap.2005.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Accepted: 03/11/2005] [Indexed: 05/31/2023]
Abstract
To evaluate radiation protection offered by the extract of Podophyllum hexandrum, expression of various cytoprotective proteins was studied using liver of Swiss albino Strain 'A' male mouse by immunoblotting. Induction of heat-shock factor-1 (HSF-1), led to up-regulation of heat-shock protein-70 (HSP-70) upon P. hexandrum (200mg/kg b. wt.; i.p. 50% ethanolic extract) treatment 2h before irradiation (10Gy) as compared to sham-irradiated control. Translocation of free nuclear factor kappa B (NFκB) from cytoplasm to nucleus was found to be inhibited upon P. hexandrum treatment. An increase in Bcl-2, proliferating cell nuclear antigen (PCNA) along with a decrease in p53, caspase-3, apoptosis inducing factor (AIF) expression was observed in the mice treated with P. hexandrum. The present study indicated that P. hexandrum extract provides protection from radiation by modulation of expression of the proteins associated with apoptosis.
Collapse
Affiliation(s)
- Raj Kumar
- Division of Radiopharmaceuticals and Radiation Biology, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Marg, Delhi 110054, India
| | | | | | | | | | | | | | | |
Collapse
|
266
|
Gaikwad A, Poblenz A, Haridas V, Zhang C, Duvic M, Gutterman J. Triterpenoid electrophiles (avicins) suppress heat shock protein-70 and x-linked inhibitor of apoptosis proteins in malignant cells by activation of ubiquitin machinery: implications for proapoptotic activity. Clin Cancer Res 2005; 11:1953-62. [PMID: 15756021 DOI: 10.1158/1078-0432.ccr-04-1704] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Avicins are plant-derived triterpenoid stress metabolites that have both proapoptotic and cytoprotective properties. Avicins induce apoptosis in Jurkat T leukemia cells by targeting mitochondria and release of cytochrome c that occurs in a p53-independent manner. However, postmitochondrial antiapoptotic barriers, such as increased expression of heat shock proteins (Hsp) and X-linked inhibitor of apoptosis proteins (XIAP), frequently exist in cancer cells and often account for resistance to chemotherapy and a poor prognosis. In this article, we show the role of avicins in the activation of stress-regulated ubiquitination and degradation of Hsp70 and XIAP. This is the first report showing the regulation of Hsp70 via the ubiquitin/proteasome pathway. We also show the induction of E3alpha ubiquitin ligase in avicin-treated Jurkat T leukemia cells, and its involvement in the degradation of XIAP. Avicin-mediated suppression of Hsp70 and XIAP was further confirmed in other leukemic/lymphoma cell lines and freshly isolated peripheral blood lymphocytes from Sezary syndrome patients. No change in the Hsp70 and XIAP proteins was observed in peripheral blood lymphocytes from normal donors. We propose that the ability of avicins to induce ubiquitination and regulate the degradation of Hsp70 and XIAP in leukemia cells could have important implications in the treatment of drug-resistant neoplasia and inflammatory disorders.
Collapse
Affiliation(s)
- Amos Gaikwad
- Department of Molecular Therapeutics, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
267
|
Vujanac M, Fenaroli A, Zimarino V. Constitutive nuclear import and stress-regulated nucleocytoplasmic shuttling of mammalian heat-shock factor 1. Traffic 2005; 6:214-29. [PMID: 15702990 DOI: 10.1111/j.1600-0854.2005.00266.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inducible expression of major cytosolic and nuclear chaperone proteins is mediated by the heat-shock transcription factor HSF1 that is activated by derepressive mechanisms triggered by transient heat stress and sustained proteotoxicity. Despite progress in defining essential aspects of HSF1 regulation, little is known about the cellular dynamics enabling this factor to mediate gene responses to cytosolic stress signals. We report that the inactive, stress-responsive form of HSF1 accumulates in the nucleus due to a relatively potent import signal, which can be recognized by importin-alpha/beta, and simultaneously undergoes continuous nucleocytoplasmic shuttling due to a comparatively weak, nonetheless efficient, export activity not involving the classical exportin-1 pathway. Strikingly, experimental stresses at physiological or elevated temperature reversibly inactivate the export competence of HSF1. Likewise, mutations mimicking stress-induced derepression impair export but not import. These findings are consistent with a dynamic process whereby exported molecules that are derepressed in an inductive cytosolic environment are recollected and pause in the nucleoplasm, replacing progressively the inactive pool. While steady-state nuclear distribution of the bulk of HSF1 ensures a rapid gene response to acute heat stress, our results suggest that the capture in the nucleus of molecules primed for activation in the cytosol may underlie responses to sustained proteotoxicity.
Collapse
Affiliation(s)
- Milos Vujanac
- DIBIT--San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy
| | | | | |
Collapse
|
268
|
Fujikake N, Nagai Y, Popiel HA, Kano H, Yamaguchi M, Toda T. Alternative splicing regulates the transcriptional activity ofDrosophilaheat shock transcription factor in response to heat/cold stress. FEBS Lett 2005; 579:3842-8. [PMID: 15978579 DOI: 10.1016/j.febslet.2005.05.074] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 05/18/2005] [Accepted: 05/27/2005] [Indexed: 11/25/2022]
Abstract
Heat shock transcription factor 1 (HSF1) mediates the induction of heat shock proteins in response to various types of stress. Although HSF1 activity is regulated by its post-translational modifications, alterations in mRNA expression have also been suggested. We here identified three new alternatively spliced isoforms of Drosophila HSF (dHSF) mRNA, named dHSFb, dHSFc, and dHSFd. We found that the ratio of dHSFb increases upon heat exposure, while that of dHSFd increases upon cold exposure. The dHSFc and dHSFd isoforms showed greater transcriptional activity than the other isoforms. Our findings suggest that alternative splicing regulates the transcriptional activity of dHSF.
Collapse
Affiliation(s)
- Nobuhiro Fujikake
- Division of Clinical Genetics, Department of Medical Genetics, Osaka University Graduate School of Medicine, 2-2-B9 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
269
|
Hägg U, Johansson ME, Grönros J, Naylor AS, Jonsdottir IH, Bergström G, Svensson PA, Gan LM. Gene expression profile and aortic vessel distensibility in voluntarily exercised spontaneously hypertensive rats: potential role of heat shock proteins. Physiol Genomics 2005; 22:319-26. [PMID: 15914578 DOI: 10.1152/physiolgenomics.00073.2005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Physical exercise is considered to be beneficial for cardiovascular health. Nevertheless, the underlying specific molecular mechanisms still remain unexplored. In this study, we aimed to investigate the effects of voluntary exercise on vascular mechanical properties and gene regulation patterns in spontaneously hypertensive rats. By using ultrasound biomicroscopy in an ex vivo perfusion chamber, we studied the distensibility of the thoracic aorta. Furthermore, exercise-induced gene regulation was studied in aortae, using microarray analysis and validated with real-time PCR. We found that distensibility was significantly improved in aortas from exercising compared with control rats (P < 0.0001). Exercising rats demonstrated a striking pattern of coordinated downregulation of genes belonging to the heat shock protein family. In conclusion, voluntary exercise leads to improved vessel wall distensibility and reduced gene expression of heat shock protein 60 and 70, which may indicate decreased oxidative stress in the aortic vascular wall.
Collapse
Affiliation(s)
- Ulrika Hägg
- Department of Physiology, Institute of Physiology and Pharmacology, Göteborg University, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
270
|
Shamovsky I, Gershon D. Novel regulatory factors of HSF-1 activation: facts and perspectives regarding their involvement in the age-associated attenuation of the heat shock response. Mech Ageing Dev 2005; 125:767-75. [PMID: 15541771 DOI: 10.1016/j.mad.2004.07.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
An attenuated response to stress is characteristic of senescence. Heat shock (HS), a significant form of stress, is delayed and reduced in aging organisms. In the response to heat shock, heat shock factor 1 (HSF-1) is activated by trimerization of its monomeric subunits. This then initiates the transcription of a series of heat shock genes (hsp genes) that encode chaperone proteins protective against heat stress. Using a promoter binding electromobility shift assay (EMSA), we have found no activation of this transcription factor in the brains of old (36 months) rats in response to exposure to 41 degrees C for 1h while strong activation is elicited in young (6 months) animals. Since brains of young and old rats had approximately the same amount of HSF-1 subunits, we anticipated the presence of auxiliary regulatory factors essential for the activation of HSF-1 and the initiation of heat shock gene transcription. We describe three novel auxiliary factors--the proteins I-HSF [HSF inhibitor] and elongation factor-1 alpha (EF-1alpha) and a large non-coding RNA (HSR)--that participate in regulation and activation of HSF-1 in early stages of heat shock gene transcription. I-HSF inhibits trimerization of HSF-1 at normal temperatures. HSR and EF-1alpha form a complex with HSF-1 and facilitate its trimerization and binding to heat shock element (HSE) in the promoters of hsps. It is proposed that structural changes in any one or a combination of these factors in response to heat shock may contribute to the age-associated attenuation in the response to stress.
Collapse
Affiliation(s)
- Ilya Shamovsky
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | |
Collapse
|
271
|
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
|
272
|
Kim SA, Yoon JH, Lee SH, Ahn SG. Polo-like kinase 1 phosphorylates heat shock transcription factor 1 and mediates its nuclear translocation during heat stress. J Biol Chem 2005; 280:12653-7. [PMID: 15661742 DOI: 10.1074/jbc.m411908200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heat shock transcription factor 1 (HSF1) is activated by pathophysiologic stresses and activation leads to an increased cellular level of heat shock proteins (Hsp(s)). Although the activation of HSF1 occurs via multiple stress-induced processes such as hyperphosphorylation, the exact cellular mechanism of HSF1 activation is still unclear. Here we show polo-like kinase 1 (PLK1) and HSF1 interact in vivo using the tandem affinity purification system. Although the interaction between HSF1 and PLK1 is increased by thermal stress, overexpression of PLK1 did not affect HSF1 trimerization or DNA binding activity. This interaction results in the phosphorylation of HSF1 on serine 419 by PLK1. Interestingly, mutation of serine 419 to alanine inhibited heat-stress induced HSF1 nuclear translocation. Our results suggest that the phosphorylation of HSF1 by PLK1 is an essential step for HSF1 nuclear translocation by heat stress.
Collapse
Affiliation(s)
- Soo-A Kim
- Oral Biology Research Institute, Chosun University College of Dentistry, Gwangju 501-759, Korea
| | | | | | | |
Collapse
|
273
|
Metz A, Soret J, Vourc'h C, Tazi J, Jolly C. A key role for stress-induced satellite III transcripts in the relocalization of splicing factors into nuclear stress granules. J Cell Sci 2005; 117:4551-8. [PMID: 15331664 DOI: 10.1242/jcs.01329] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Exposure of cells to stressful conditions results in the rapid synthesis of a subset of specialized proteins termed heat shock proteins (HSPs) which function in protecting the cell against damage. The stress-induced activation of hsp genes is controlled by the heat shock transcription factor 1 (HSF1). At the cellular level, one of the most striking effects of stress is the rapid and reversible redistribution of HSF1 into a few nuclear structures termed nuclear stress granules which form primarily on the 9q12 locus in humans. Within these structures, HSF1 binds to satellite III repeated elements and drives the RNA polymerase II-dependent transcription of these sequences into stable RNAs which remain associated with the 9q12 locus for a certain time after synthesis. Other proteins, in particular splicing factors, were also shown to relocalize to the granules upon stress. Here, we investigated the role of stress-induced satellite III transcripts in the relocalization of splicing factors to the granules. We show that the recruitment of the two serine/arginine-rich (SR) proteins SF2/ASF and SRp30c requires the presence of stress-induced satellite III transcripts. In agreement with these findings, we identified the second RNA-recognition motif (RRM2) of hSF2/ASF as the motif required for the targeting to the granules, and we showed by immunoprecipitation that the endogenous hSF2/ASF protein is present in a complex with satellite III transcripts in stressed cells in vivo. Interestingly, satellite III transcripts also immunoprecipitate together with small nuclear ribonucleoproteins (snRNPs) in vivo whereas the intronless hsp70 transcripts do not, supporting the proposal that these transcripts are subject to splicing. Altogether, these data highlight the central role for satellite III transcripts in the targeting and/or retention of splicing factors into the granules upon stress.
Collapse
Affiliation(s)
- Alexandra Metz
- INSERM U309, Institut Albert Bonniot, Domaine de la Merci, 38706 La Tronche CEDEX, France
| | | | | | | | | |
Collapse
|
274
|
Elo MA, Kaarniranta K, Helminen HJ, Lammi MJ. Hsp90 inhibitor geldanamycin increases hsp70 mRNA stabilisation but fails to activate HSF1 in cells exposed to hydrostatic pressure. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1743:115-9. [PMID: 15777846 DOI: 10.1016/j.bbamcr.2004.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Revised: 08/30/2004] [Accepted: 09/08/2004] [Indexed: 10/26/2022]
Abstract
High hydrostatic pressure (HP) increases Hsp70 protein and mRNA levels by increasing the mRNA half-life without activation of HSF1 transcription factor. We investigated whether this change in gene expression requires Hsp90, previously shown to regulate hsp70 genes via HSF1. In HeLa cells, both HP and Hsp90 inhibitor geldanamycin (GA) up-regulated Hsp70 expression through mRNA stabilisation. GA, unlike HP, increased HSF1 activation. However, when exposures were used together a marked Hsp70 response was observed with mRNA stabilisation without coincidence of HSF1 activation. Our data suggests that Hsp90 is involved in hsp70 mRNA stabilisation and the HSF1 activation can be suppressed by high HP.
Collapse
Affiliation(s)
- Mika A Elo
- Department of Anatomy, University of Kuopio, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | | | |
Collapse
|
275
|
Jiang HY, Wek RC. Phosphorylation of the alpha-subunit of the eukaryotic initiation factor-2 (eIF2alpha) reduces protein synthesis and enhances apoptosis in response to proteasome inhibition. J Biol Chem 2005; 280:14189-202. [PMID: 15684420 DOI: 10.1074/jbc.m413660200] [Citation(s) in RCA: 253] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein ubiquitination and subsequent degradation by the proteasome are important mechanisms regulating cell cycle, growth and differentiation, and apoptosis. Recent studies in cancer therapy suggest that drugs that disrupt the ubiquitin/proteasome pathway induce apoptosis and sensitize malignant cells and tumors to conventional chemotherapy. In this study we addressed the role of phosphorylation of the alpha-subunit eukaryotic initiation factor-2 (eIF2), and its attendant regulation of gene expression, in the cellular stress response to proteasome inhibition. Phosphorylation of eIF2alpha in mouse embryo fibroblast (MEF) cells subjected to proteasome inhibition leads to a significant reduction in protein synthesis, concomitant with induced expression of the bZIP transcription regulator, ATF4, and its target gene CHOP/GADD153. The primary eIF2alpha kinase activated by exposure of these fibroblast cells to proteasome inhibition is GCN2 (EIF2AK4), which has a central role in the recognition of cytoplasmic stress signals. Endoplasmic reticulum (ER) stress is not effectively induced in MEF cells subjected to proteasome inhibition, with minimal activation of the ER stress sensory proteins, eIF2alpha kinase PEK (PERK/EIF2AK3), IRE1 protein kinase and the transcription regulator ATF6 following up to 6 h of proteasome inhibitor treatment. Loss of eIF2alpha phosphorylation thwarts caspase activation and delays apoptosis. Central to this pro-apoptotic function of eIF2alpha kinases during proteasome inhibition is the transcriptional regulator CHOP, as deletion of CHOP in MEF cells impedes apoptosis. We conclude that eIF2alpha kinases are integral to cellular stress pathways induced by proteasome inhibitors, and may be central to the efficacy of anticancer drugs that target the ubiquitin/proteasome pathway.
Collapse
Affiliation(s)
- Hao-Yuan Jiang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | |
Collapse
|
276
|
Uenishi R, Suzuki K, Koizumi S. Overexpression of Heat Shock Factor 1 Masks the Heavy Metal Response of the Heat Shock Protein 70 (hsp70) Gene Promoter. ACTA ACUST UNITED AC 2005. [DOI: 10.1248/jhs.51.242] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Rie Uenishi
- Department of Health Effects Research, National Institute of Industrial Health
| | - Kaoru Suzuki
- Department of Health Effects Research, National Institute of Industrial Health
| | - Shinji Koizumi
- Department of Health Effects Research, National Institute of Industrial Health
| |
Collapse
|
277
|
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
|
278
|
Freitas FZ, Bertolini MC. Genomic organization of the Neurospora crassa gsn gene: possible involvement of the STRE and HSE elements in the modulation of transcription during heat shock. Mol Genet Genomics 2004; 272:550-61. [PMID: 15558319 DOI: 10.1007/s00438-004-1086-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2004] [Accepted: 10/26/2004] [Indexed: 10/26/2022]
Abstract
Glycogen synthase, an enzyme involved in glycogen biosynthesis, is regulated by phosphorylation and by the allosteric ligand glucose-6-phosphate (G6P). In addition, enzyme levels can be regulated by changes in gene expression. We recently cloned a cDNA for glycogen synthase ( gsn) from Neurospora crassa, and showed that gsn transcription decreased when cells were exposed to heat shock (shifted from 30 degrees C to 45 degrees C). In order to understand the mechanisms that control gsn expression, we isolated the gene, including its 5' and 3' flanking regions, from the genome of N. crassa. An ORF of approximately 2.4 kb was identified, which is interrupted by four small introns (II-V). Intron I (482 bp) is located in the 5'UTR region. Three putative Transcription Initiation Sites (TISs) were mapped, one of which lies downstream of a canonical TATA-box sequence (5'-TGTATAAA-3'). Analysis of the 5'-flanking region revealed the presence of putative transcription factor-binding sites, including Heat Shock Elements (HSEs) and STress Responsive Elements (STREs). The possible involvement of these motifs in the negative regulation of gsn transcription was investigated using Electrophoretic Mobility Shift Assays (EMSA) with nuclear extracts of N. crassa mycelium obtained before and after heat shock, and DNA fragments encompassing HSE and STRE elements from the 5'-flanking region. While elements within the promoter region are involved in transcription under heat shock, elements in the 5'UTR intron may participate in transcription during vegetative growth. The results thus suggest that N. crassa possesses trans -acting elements that interact with the 5'-flanking region to regulate gsn transcription during heat shock and vegetative growth.
Collapse
Affiliation(s)
- F Zanolli Freitas
- Departamento de Bioquímica e Tecnologia Química, Instituto de Química, UNESP, 14800-900 Araraquara, SP, Brazil
| | | |
Collapse
|
279
|
Westerheide SD, Bosman JD, Mbadugha BNA, Kawahara TLA, Matsumoto G, Kim S, Gu W, Devlin JP, Silverman RB, Morimoto RI. Celastrols as inducers of the heat shock response and cytoprotection. J Biol Chem 2004; 279:56053-60. [PMID: 15509580 DOI: 10.1074/jbc.m409267200] [Citation(s) in RCA: 297] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Alterations in protein folding and the regulation of conformational states have become increasingly important to the functionality of key molecules in signaling, cell growth, and cell death. Molecular chaperones, because of their properties in protein quality control, afford conformational flexibility to proteins and serve to integrate stress-signaling events that influence aging and a range of diseases including cancer, cystic fibrosis, amyloidoses, and neurodegenerative diseases. We describe here characteristics of celastrol, a quinone methide triterpene and an active component from Chinese herbal medicine identified in a screen of bioactive small molecules that activates the human heat shock response. From a structure/function examination, the celastrol structure is remarkably specific and activates heat shock transcription factor 1 (HSF1) with kinetics similar to those of heat stress, as determined by the induction of HSF1 DNA binding, hyperphosphorylation of HSF1, and expression of chaperone genes. Celastrol can activate heat shock gene transcription synergistically with other stresses and exhibits cytoprotection against subsequent exposures to other forms of lethal cell stress. These results suggest that celastrols exhibit promise as a new class of pharmacologically active regulators of the heat shock response.
Collapse
Affiliation(s)
- Sandy D Westerheide
- Department of Biochemistry, Molecular Biology and Cell Biology, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
280
|
Ohnishi K, Takahashi A, Yokota S, Ohnishi T. Effects of a heat shock protein inhibitor KNK437 on heat sensitivity and heat tolerance in human squamous cell carcinoma cell lines differing in p53 status. Int J Radiat Biol 2004; 80:607-14. [PMID: 15370972 DOI: 10.1080/09553000412331283470] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE The effects of a heat shock protein (hsp) inhibitor KNK437 (N-formyl-3,4-methylenedioxy-benzylidene-gamma-butyrolactam) were examined on the heat sensitivity and heat tolerance of human cancer cells with special reference to p53 status. MATERIALS AND METHODS Human squamous cell carcinoma (SAS) and glioblastoma cell lines (A-172) transfected with mutant p53 (mp53) or control neo genes were used. KNK437 was added in culture medium at a final concentration of 50, 100 or 300 microM 1 h before heating (42 degrees C). Surviving fractions of cells were measured by use of a clonogenic assay. Effects of KNK437 on the accumulation of heat shock proteins and DNA binding activity of heat shock factor 1 were examined with Western blot analysis and gel mobility-shift assay, respectively. Heat-induced apoptotic bodies were detected by Hoechst 33342 staining. RESULTS The mp53-transfected SAS (SAS/mp53) and A-172 (A-172/mp53) cells were more resistant to heat than the neomycin (neo)-transfected SAS (SAS/neo) and A-172 (A-172/neo) cells. The constitutive amount of hsp27 was larger in SAS/mp53 than in SAS/neo cells. Clear differences in the constitutive amounts of hsp40, hsp72 and hsp90 were not observed between SAS/mp53 and SAS/neo cells. KNK437 enhanced the heat sensitivity in SAS/mp53 and A-172/mp53 cells more effectively than in neo control cells. Heat tolerance was suppressed by KNK437 in SAS/mp53 and SAS/neo cells and also in A-172/mp53 and A-172/neo cells. Along with suppression of heat tolerance, KNK437 suppressed heat-induced accumulation of both hsp27 and hsp72. Heat-induced apoptotic bodies were enhanced by KNK437 in SAS/mp53 and SAS/neo cells. CONCLUSION The results suggest a possible mechanism for the heat sensitivity of SAS cells. Heat sensitivity depends on p53 status regulating the amount of hsp27. Heat tolerance is suppressed by KNK437 through the suppression of heat-induced accumulations of hsp27 and hsp72 and the induction of p53-independent apoptosis.
Collapse
Affiliation(s)
- K Ohnishi
- Department of Biology, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | | | | | | |
Collapse
|
281
|
Wang G, Ying Z, Jin X, Tu N, Zhang Y, Phillips M, Moskophidis D, Mivechi NF. Essential requirement for both hsf1 and hsf2 transcriptional activity in spermatogenesis and male fertility. Genesis 2004; 38:66-80. [PMID: 14994269 DOI: 10.1002/gene.20005] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heat shock factors (Hsfs) are major transactivators of heat shock proteins but are also involved in regulation of other genes active in embryonic development. High expression levels of Hsfs in mouse testis during development suggest a role for these factors in spermatogenesis, a cyclic process of spermatogonia cell-differentiation into mature spermatozoa. In contrast to hsf1(-/-) mice, which exhibit normal spermatogenesis, targeted disruption of hsf2 results in reduced testicular size but only a small impairment in male fertility. We show here that disruption of both hsf1 and hsf2 results in a more severe phenotype associated with male sterility due to severe defects in spermatogenesis. Earliest defects observed are the reduced number of germ cells in juvenile mice and germ cells that enter the meiotic prophase fail to progress beyond the pachytene stage. This was associated with a reduction or absence of transcription of genes critically involved in spermatogenesis. The findings suggest that additive or synergistic transcriptional activity of both hsf1 and hsf2 is required for normal mammalian spermatogenesis and male fertility.
Collapse
Affiliation(s)
- Guanghu Wang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia
| | | | | | | | | | | | | | | |
Collapse
|
282
|
Wang X, Grammatikakis N, Siganou A, Stevenson MA, Calderwood SK. Interactions between extracellular signal-regulated protein kinase 1, 14-3-3epsilon, and heat shock factor 1 during stress. J Biol Chem 2004; 279:49460-9. [PMID: 15364926 DOI: 10.1074/jbc.m406059200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytoprotection during the heat shock response is a complex phenomenon involving multiple inducible mechanisms. We have examined the interaction of two key molecular components in the response, heat shock transcription factor 1 (HSF1) and extracellular signal regulated protein kinase (ERK). Whereas both HSF1 and ERK are required to protect cells against apoptosis, ERK activation is paradoxically antagonistic to trans-activation of hsp promoters by HSF1 and HSP accumulation during heat shock. We have found that the two pathways interact directly and that heat shock causes the physical association of ERK1 with HSF1, an interaction that promotes the kinase activity of ERK in heat-shocked cells. ERK activation results in the recruitment of the phosphoserine binding protein 14-3-3epsilon in a manner dependent on previous HSF1 phosphorylation by ERK. The effects of 14-3-3epsilon binding on HSF1 were complex, however, depending on extracellular conditions, in that HSF1-14-3-3 binding at 37 degrees C led to the cytoplasmic sequestration and repression of HSF1, whereas heat shock overrode these effects and caused quantitative nuclear localization of HSF1. Although the effects of 14-3-3epsilon binding to HSF1 were overridden acutely by stress, during recovery from heat shock, 14-3-3epsilon association again led to enhanced cytoplasmic localization of HSF1, implicating a role for ERK/14-3-3epsilon in HSF1 deactivation in recovering cells. Association of HSF1 with ERK and 14-3-3epsilon during heat shock may thus modulate the amplitude of the response and lead to efficient termination of HSP expression on resumption of growth conditions.
Collapse
Affiliation(s)
- Xiaozhe Wang
- Dana-Farber Cancer Institute and Division of Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
283
|
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
|
284
|
DeFranco DB, Ho L, Falke E, Callaway CW. Small molecule activators of the heat shock response and neuroprotection from stroke. Curr Atheroscler Rep 2004; 6:295-300. [PMID: 15191704 DOI: 10.1007/s11883-004-0061-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Various cellular defense pathways are mobilized in response to stress that serve to limit potential damage to organelles and biochemical pathways that would disrupt normal cellular function or trigger cell death. Strategies utilized by cells subjected to various forms of stress include the activation of detoxification systems that act to eliminate the primary damaging molecules, remove damaged cellular macromolecules, or restore organelle and macromolecule function in cases where loss of activity is generated by reversible modifications or alterations in conformation (ie, misfolding). Central to many intracellular defense mechanisms that operate to limit damage to protein function are molecular chaperones of the heat shock protein (HSP) family. This review briefly discusses the molecular mechanisms that are thought to dictate the well-established neuroprotective effects of HSPs and highlight the recent attempts to use pharmacologic approaches to activate this important cellular defense pathway.
Collapse
Affiliation(s)
- Donald B DeFranco
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | | | | | | |
Collapse
|
285
|
Wang Y, Theriault JR, He H, Gong J, Calderwood SK. Expression of a Dominant Negative Heat Shock Factor-1 Construct Inhibits Aneuploidy in Prostate Carcinoma Cells*. J Biol Chem 2004; 279:32651-9. [PMID: 15152009 DOI: 10.1074/jbc.m401475200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent studies have implicated heat shock proteins (HSP) and heat shock transcription factor 1 (HSF1) in tumor progression. We have examined the role of HSF1 in the malignant phenotype of PC-3 prostate carcinoma cells. We have developed a dominant negative construct of HSF1 that antagonizes transcription from HSP promoters and results in the depletion of intracellular HSP 70. Our studies indicate that expression of DN-HSF1 dramatically alters the DNA content of PC-3 cells (derived from p53 null prostatic carcinoma) and inhibits aneuploidy in these cells. This effect is due to prolonged expression of DN-HSF1, and transient expression of the dominant negative factor from an inducible promoter failed to cause the effect. Inhibition of aneuploidy in p53 null PC-3 cells by DN-HSF1 expression was recapitulated by expression within the cells of wild type p53. Furthermore, cells expressing DN-HSF1 showed a profound inhibition in the development of aneuploidy when exposed to chemical agents that disrupt the mitotic spindle and prevent progression through metaphase. Inhibition of aneuploidy in PC-3 cells expressing DN-HSF1 was associated with delayed breakdown of cyclin B1 compared with controls, consistent with a role for wild type HSF1 in the regulation of cyclin B1 degradation, a key step in the control of mitosis. Our experiments therefore demonstrate that HSF1 plays a functional role in cancer cells under nonstress conditions and influences cell cycle behavior and progression through mitosis and promotes the development of the aneuploid state.
Collapse
MESH Headings
- Aneuploidy
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Cycle
- Cell Division
- Cell Line
- Cell Line, Tumor
- Cells, Cultured
- Cyclin B/metabolism
- Cyclin B1
- DNA/metabolism
- DNA-Binding Proteins/genetics
- Demecolcine/pharmacology
- Dose-Response Relationship, Drug
- Electrophoresis, Polyacrylamide Gel
- Genes, Dominant
- Genes, Reporter
- Genes, p53
- Genetic Vectors
- HSP72 Heat-Shock Proteins
- Heat Shock Transcription Factors
- Heat-Shock Proteins/metabolism
- Humans
- Immunoblotting
- Luciferases/metabolism
- Male
- Mitosis
- Mutation
- Phenotype
- Ploidies
- Promoter Regions, Genetic
- Prostatic Neoplasms/genetics
- Protein Structure, Tertiary
- Resting Phase, Cell Cycle
- Spectrometry, Fluorescence
- Transcription Factors
- Transfection
Collapse
Affiliation(s)
- Yiqun Wang
- Department of Adult Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
286
|
Affiliation(s)
- Giuseppe Biamonti
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Via Ferrata 1, 27100 Pavia, Italy.
| |
Collapse
|
287
|
Le Goff P, Le Dréan Y, Le Péron C, Le Jossic-Corcos C, Ainouche A, Michel D. Intracellular trafficking of heat shock factor 2. Exp Cell Res 2004; 294:480-93. [PMID: 15023536 DOI: 10.1016/j.yexcr.2003.11.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2003] [Revised: 10/23/2003] [Indexed: 12/14/2022]
Abstract
HSF2 is an enigmatic member of the heat shock factor family, identified in the homeotherm classes of birds and mammals. We report the characterization of HSF2 from an evolutionary ancient vertebrate, the fish rainbow trout (rtHSF2). rtHSF2 appears closely related to its mammalian counterparts at structural and functional levels. The conservation of the distinctive features of HSF2 from fish to human suggests that it should ensure important biological functions, not redundant with those of HSF1. Proteasome inhibition, reported as a potent stimulator of HSF2, leads to the stabilization and to a striking nuclear trafficking of rtHSF2-GFP fusion protein. Upon treatment with the proteasome inhibitor MG132, rtHSF2-GFP accumulates into PML nuclear bodies (NBs) independently of its sumoylation and, if expressed at moderate level, moves to nucleoli. The translocation of rtHSF2-GFP from NBs to nucleoli is greatly favored by overexpression of the heat shock protein Hsp70. The mammalian counterpart mouse HSF2 (mHSF2) also exhibited changes in intracellular distribution upon MG132 treatment. mHSF2 partitioned between a juxtanuclear area that we characterized as an aggresome and the nucleoli. These relocalizations are likely to reflect common structural changes of mouse and trout HSF2 upon activation.
Collapse
Affiliation(s)
- Pascale Le Goff
- UMR CNRS 6026 Information et Programmation Cellulaire, Université de Rennes1, 35042 Rennes Cedex, France.
| | | | | | | | | | | |
Collapse
|
288
|
Kieran D, Kalmar B, Dick JRT, Riddoch-Contreras J, Burnstock G, Greensmith L. Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice. Nat Med 2004; 10:402-5. [PMID: 15034571 DOI: 10.1038/nm1021] [Citation(s) in RCA: 384] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 03/02/2004] [Indexed: 11/10/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition in which motoneurons of the spinal cord and motor cortex die, resulting in progressive paralysis. This condition has no cure and results in eventual death, usually within 1-5 years of diagnosis. Although the specific etiology of ALS is unknown, 20% of familial cases of the disease carry mutations in the gene encoding Cu/Zn superoxide dismutase-1 (SOD1). Transgenic mice overexpressing human mutant SOD1 have a phenotype and pathology that are very similar to that seen in human ALS patients. Here we show that treatment with arimoclomol, a coinducer of heat shock proteins (HSPs), significantly delays disease progression in mice expressing a SOD1 mutant in which glycine is substituted with alanine at position 93 (SOD1(G93A)). Arimoclomol-treated SOD1(G93A) mice show marked improvement in hind limb muscle function and motoneuron survival in the later stages of the disease, resulting in a 22% increase in lifespan. Pharmacological activation of the heat shock response may therefore be a successful therapeutic approach to treating ALS, and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Dairin Kieran
- The Graham Watts Laboratory, Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, The National Hospital for Neurology and Neurosurgery, University College London, Queen Square, London WC1N 3BG, UK
| | | | | | | | | | | |
Collapse
|
289
|
Kim KY, Shin SM, Kim JK, Paik SG, Yang Y, Choi I. Heat shock factor regulates VDUP1 gene expression. Biochem Biophys Res Commun 2004; 315:369-75. [PMID: 14766217 DOI: 10.1016/j.bbrc.2004.01.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2003] [Indexed: 11/20/2022]
Abstract
The vitamin D3 up-regulated protein 1 (VDUP1) is identified as interacting protein with thioredoxin (TRX) and functions as a natural antagonist of TRX. Its expression is regulated by various stresses including ROS, UV, and heat shock. In the present study, we observed an inducible expression of VDUP1 in Bosc cells by high density and serum deprivation cultures. To determine transcription factors associated with the induction of VDUP1 by stresses, the promoter region of VDUP1 was cloned. Through reporter assays with plasmids having various deletion of its promoter region and analysis of putative cis-elements, heat shock factor element (HSE) was identified. The deletion of HSE abolished transcriptional activity of VDUP1 promoter by stresses and the binding of heat shock factor (HSF) to HSE was confirmed by gel-shift and supershift assays using nuclear extracts prepared from stressed Bosc cells. Also, the enforced expression of HSF or heat shock increased the transcription of endogenous VDUP1. These imply that HSF is an important transcription factor involved in up-regulation of VDUP1 expression by stresses such as high density and serum deprivation cultures.
Collapse
Affiliation(s)
- Kun-Yong Kim
- Laboratory of Immunology, Korea Research Institute of Bioscience and Biotechnology, Yusong, Taejon 305-333, Republic of Korea
| | | | | | | | | | | |
Collapse
|
290
|
Fiorenza MT, Bevilacqua A, Canterini S, Torcia S, Pontecorvi M, Mangia F. Early transcriptional activation of the hsp70.1 gene by osmotic stress in one-cell embryos of the mouse. Biol Reprod 2004; 70:1606-13. [PMID: 14766729 DOI: 10.1095/biolreprod.103.024877] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In fertilized mouse eggs, de novo transcription of embryonic genes is first observed during the S phase of the one-cell stage. This transcription, however, is mostly limited to the male pronucleus and possibly uncoupled from translation, making the functional meaning obscure. We found that one-cell mouse embryos respond to the osmotic shock of in vitro isolation with migration of HSF1, the canonical stress activator of mammalian heat shock genes, to pronuclei and by transient transcription of the hsp70.1, but not hsp70.3 and hsp90, heat shock genes. Isolated growing dictyate oocytes also display a nuclear HSF1 localization, but, in contrast with embryos, they transcribe both hsp70.1 and hsp70.3 genes only after heat shock. Intranuclear injection of double-stranded oligodeoxyribonucleotides containing HSE, GAGA box or GC box consensus sequences, and antibodies raised to transcription factors HSF1, HSF2, Drosophila melanogaster GAGA factor, or Sp1 demonstrated that hsp70.1 transcription depends on HSF1 in both oocytes and embryos and that Sp1 is dispensable in oocytes and inhibitory in the embryos. Hsp70.1 thus represents the first endogenous gene so far identified to be physiologically activated and tightly regulated after fertilization in mammals.
Collapse
Affiliation(s)
- Maria Teresa Fiorenza
- Istituto Pasteur-Fondazione Cenci Bolognetti and Department of Psychology, Section of Neuroscience, UniversityLa Sapienza of Rome, 00185 Rome, Italy
| | | | | | | | | | | |
Collapse
|
291
|
Hong S, Kim SH, Heo MA, Choi YH, Park MJ, Yoo MA, Kim HD, Kang HS, Cheong J. Coactivator ASC-2 mediates heat shock factor 1-mediated transactivation dependent on heat shock. FEBS Lett 2004; 559:165-70. [PMID: 14960326 DOI: 10.1016/s0014-5793(04)00028-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2003] [Revised: 10/07/2003] [Accepted: 10/17/2003] [Indexed: 11/27/2022]
Abstract
Upon exposure to elevated temperatures, mammalian cells increase the expression of the heat shock proteins (HSP) through activation of the heat shock factor 1 (HSF1). Since most transcription factors require coactivators for efficient transcriptional activity, we tried to identify the coactivator(s) that interacts with and modulates the activities of HSF1. In vitro glutathione S-transferase (GST) pull-down assay revealed that HSF1 strongly interacts with activating signal cointegrator (ASC)-2 and weakly with cyclic adenosine monophosphate responsive element binding protein (CBP). We also show that cotransfection of ASC-2, but not CBP, potentiates HSF1-mediated transactivation based on its cognate element (heat shock element, HSE) linked to luciferase reporter. The molecular interaction of HSF1 and ASC-2 was stimulated by heat shock in cells and the overexpression of HSF1-interacting domain of ASC-2 inhibited the specific induced protein association and HSF1-mediated transactivation. Taking these results together, we suggest that ASC-2 in a novel coactivator for HSF1 and heat shock stress may contribute the strong active transcription complex through sequential recruitment of HSF1 and ASC-2.
Collapse
Affiliation(s)
- SunHwa Hong
- Department of Molecular Biology, Pusan National University, Busan 609-735, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
292
|
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
|
293
|
Voellmy R. Transcriptional Regulation of the Metazoan Stress Protein Response. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2004; 78:143-85. [PMID: 15210330 DOI: 10.1016/s0079-6603(04)78004-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
This review provides an updated account of the regulation of the metazoan stress protein response. Where indicated, observations made with yeasts are also included. However, a discussion of the plant stress protein response is intentionally omitted (for a review, see 1). The stress protein response, as discussed hereafter, is understood to relate to the response by virtually all cells to heat and other stressors that results in the induced expression of so-called heat shock or stress genes. The protein products of these genes localize largely to the cytoplasm, nucleus, or organelles. An analogous response controls the expression of related genes, whose products reside in the endoplasmic reticulum. The response, termed ER stress response or unfolded protein response, is mediated by a separate regulation system that is not discussed in this review. Note, however, that recent work suggests the existence of commonalities between the regulatory systems controlling the stress protein and ER stress responses (2).
Collapse
Affiliation(s)
- Richard Voellmy
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
294
|
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: 24] [Impact Index Per Article: 1.1] [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
|
295
|
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 2004. [DOI: 10.1379/1466-1268(2004)009<0021:traboh>2.0.co;2] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
296
|
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
|
297
|
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
|
298
|
Nonaka T, Akimoto T, Mitsuhashi N, Tamaki Y, Nakano T. Changes in the number of HSF1 positive granules in the nucleus reflects heat shock semiquantitatively. Cancer Lett 2003; 202:89-100. [PMID: 14643030 DOI: 10.1016/s0304-3835(03)00481-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE The purpose of this study is to examine the changes in the number of HSF1 granules in the nucleus caused by different degree of heat stress. MATERIALS AND METHODS A human esophageal cancer cell line, TE-2, was used. HSF1 granules were examined in an immunofluorescence study, and the changes in the average number of HSF1 granules after heat alone or heat in combination with KNK437, Hsp inhibitor, were evaluated. A band shift of HSF1 was also determined by western blot. RESULTS HSF1 granules appeared soon after the start of heating at 43 degrees C and reached a peak at 60 min and gradually disappeared after discontinuation of heat. In the fractionated heat treatment, preheating (43 degrees C, 30 min) suppressed the increase in the number of the granules during the second heating, but suppression of Hsp72 by KNK437 resulted in increase in the number of granules. Continued heating at 43 degrees C with or without KNK437 maintained the number of the granules at the peak level during heat treatment. The band shift of HSF1 examined by western blot correlated with the changes in the number of granules. The number of granules also reflected the degree of stress according to different temperature. CONCLUSION The number of HSF1 granules in the nucleus well reflected heat stress, and was almost consistent with phosphorylation of HSF1. The number of HSF1 granules would be a useful tool for evaluating different degrees of heat stress semiquantitatively.
Collapse
Affiliation(s)
- Tetsuo Nonaka
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | | | | | | | | |
Collapse
|
299
|
Airaksinen S, Jokilehto T, Råbergh CMI, Nikinmaa M. Heat- and cold-inducible regulation of HSP70 expression in zebrafish ZF4 cells. Comp Biochem Physiol B Biochem Mol Biol 2003; 136:275-82. [PMID: 14529753 DOI: 10.1016/s1096-4959(03)00205-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Elevated temperature induces a rapid heat shock transcription factor (HSFs)-mediated expression of heat shock (hsp) genes. The effect of cold exposure on hsp gene expression has hardly been investigated, although ectothermic animals experience both cold and heat stress. We have previously shown in zebrafish that the expression of hsf1a and a unique isoform hsf1b vary in a tissue-specific manner upon heat stress. In the current study, using a zebrafish (Danio rerio) embryonic cell line (ZF4), we have compared the effects of heat shock (28-->37 degrees C) vs. cold shock (28-->20 degrees C) on the expression of ahsf1a, zhsf1b and hsp70. Concomitantly, the suitability of the ZF4 cells as a model system was verified. The expression pattern of HSP70 proteins following heat or cold exposure is distinct, and the total HSP70 level is upregulated or stable, respectively. Moreover, heat exposure specifically increases the ratio of zhsf1a/b expression (10-fold), whereas cold exposure decreases it to one half. These data suggest that the zhsf1a/zhsf1b ratio is regulated in a temperature-dependent manner, and the ratio may be indicative of the stressor-specific HSP70 expression. Furthermore, the response in ZF4 cells upon heat shock resembles the response observed in zebrafish liver and thus, supports the use of this cell line in stress response studies.
Collapse
Affiliation(s)
- Susanna Airaksinen
- Department of Biology, Laboratory of Animal Physiology, University of Turku, Turku FIN-20014, Finland.
| | | | | | | |
Collapse
|
300
|
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
|