Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Bioessays ; 45(7): e2300036, 2023 07.
Article in English | MEDLINE | ID: mdl-37092382

ABSTRACT

Environmental, physiological, and pathological stimuli induce the misfolding of proteins, which results in the formation of aggregates and amyloid fibrils. To cope with proteotoxic stress, cells are equipped with adaptive mechanisms that are accompanied by changes in gene expression. The evolutionarily conserved mechanism called the heat shock response is characterized by the induction of a set of heat shock proteins (HSPs), and is mainly regulated by heat shock transcription factor 1 (HSF1) in mammals. We herein introduce the mechanisms by which HSF1 tightly controls the transcription of HSP genes via the regulation of pre-initiation complex recruitment in their promoters under proteotoxic stress. These mechanisms involve the stress-induced regulation of HSF1-transcription complex formation with a number of coactivators, changes in chromatin states, and the formation of phase-separated condensates through post-translational modifications.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Animals , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Chromatin/genetics , Proteotoxic Stress , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Transcription, Genetic , Mammals/genetics
2.
EMBO J ; 38(24): e102566, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31657478

ABSTRACT

The recruitment of RNA polymerase II (Pol II) to core promoters is highly regulated during rapid induction of genes. In response to heat shock, heat shock transcription factor 1 (HSF1) is activated and occupies heat shock gene promoters. Promoter-bound HSF1 recruits general transcription factors and Mediator, which interact with Pol II, but stress-specific mechanisms of Pol II recruitment are unclear. Here, we show in comparative analyses of HSF1 paralogs and their mutants that HSF1 interacts with the pericentromeric adaptor protein shugoshin 2 (SGO2) during heat shock in mouse cells, in a manner dependent on inducible phosphorylation of HSF1 at serine 326, and recruits SGO2 to the HSP70 promoter. SGO2-mediated binding and recruitment of Pol II with a hypophosphorylated C-terminal domain promote expression of HSP70, implicating SGO2 as one of the coactivators that facilitate Pol II recruitment by HSF1. Furthermore, the HSF1-SGO2 complex supports cell survival and maintenance of proteostasis in heat shock conditions. These results exemplify a proteotoxic stress-specific mechanism of Pol II recruitment, which is triggered by phosphorylation of HSF1 during the heat shock response.


Subject(s)
Cell Cycle Proteins/metabolism , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/physiology , RNA Polymerase II/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Mice , Mice, Knockout , Phosphorylation , Protein Binding
3.
Biol Reprod ; 105(4): 976-986, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34007999

ABSTRACT

Activating transcription factor 1 (ATF1), belonging to the CREB/ATF family of transcription factors, is highly expressed in the testes. However, its role in spermatogenesis has not yet been established. Here, we aimed to elucidate the impact of ATF1 in spermatogenesis by examining the expression pattern of ATF1 in mice and the effect of ATF1 knockdown in the mouse testes. We found that ATF1 is expressed in various organs, with very high levels in the testes. Immunohistochemical staining showed that ATF1 was localized in the nuclei of spermatogonia and co-localized with proliferating cell nuclear antigen. In ATF1-deficient mice, the seminiferous tubules of the testis contained cells at all developmental stages; however, the number of spermatocytes was decreased. Proliferating cell nuclear antigen expression was decreased and apoptotic cells were rare in the seminiferous tubules. These results indicate that ATF1 plays a role in male germ cell proliferation and sperm production.


Subject(s)
Activating Transcription Factor 1/genetics , Gene Expression , Mice/genetics , Spermatogenesis/genetics , Testis/metabolism , Activating Transcription Factor 1/metabolism , Animals , Gene Expression Profiling , Male , Mice/metabolism
4.
J Cell Sci ; 131(9)2018 05 08.
Article in English | MEDLINE | ID: mdl-29632240

ABSTRACT

Heat shock transcription factor 1 (HSF1) regulates the expression of a wide array of genes, controls the expression of heat shock proteins (HSPs) as well as cell growth. Although acute depletion of HSF1 induces cellular senescence, the underlying mechanisms are poorly understood. Here, we report that HSF1 depletion-induced senescence (HDIS) of human diploid fibroblasts (HDFs) was independent of HSP-mediated proteostasis but dependent on activation of the p53-p21 pathway, partly because of the increased expression of dehydrogenase/reductase 2 (DHRS2), a putative MDM2 inhibitor. We observed that HDIS occurred without decreased levels of major HSPs or increased proteotoxic stress in HDFs. Additionally, VER155008, an inhibitor of HSP70 family proteins, increased proteotoxicity and suppressed cell growth but failed to induce senescence. Importantly, we found that activation of the p53-p21 pathway resulting from reduced MDM2-dependent p53 degradation was required for HDIS. Furthermore, we provide evidence that increased DHRS2 expression contributes to p53 stabilization and HDIS. Collectively, our observations uncovered a molecular pathway in which HSF1 depletion-induced DHRS2 expression leads to activation of the MDM2-p53-p21 pathway required for HDIS.


Subject(s)
Fibroblasts/metabolism , Heat Shock Transcription Factors/deficiency , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cell Proliferation , Cellular Senescence/physiology , Diploidy , Fibroblasts/cytology , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors/metabolism , Humans , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics
5.
Mol Cell ; 48(2): 182-94, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22940245

ABSTRACT

Transcription factor access to regulatory elements is prevented by the nucleosome. Heat shock factor 1 (HSF1) is a winged helix transcription factor that plays roles in control and stressed conditions by gaining access to target elements, but mechanisms of HSF1 access are not well known in mammalian cells. Here, we show the physical interaction between the wing motif of human HSF1 and replication protein A (RPA), which is involved in DNA metabolism. Depletion of RPA1 abolishes HSF1 access to the promoter of HSP70 in unstressed condition and delays its rapid activation in response to heat shock. The HSF1-RPA complex leads to preloading of RNA polymerase II and opens the chromatin structure by recruiting a histone chaperone, FACT. Furthermore, this interaction is required for melanoma cell proliferation. These results provide a mechanism of constitutive HSF1 access to nucleosomal DNA, which is important for both basal and inducible gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , High Mobility Group Proteins , Regulatory Elements, Transcriptional , Replication Protein A/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors , Amino Acid Sequence , Base Sequence , Chromatin/genetics , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , HEK293 Cells , Heat Shock Transcription Factors , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Molecular Sequence Data , Nucleosomes/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
6.
Carcinogenesis ; 35(2): 272-81, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24130164

ABSTRACT

Heat shock factor 1 (HSF1), a major transactivator of stress responses, has been implicated in carcinogenesis in various organs. However, little is known about the biological functions of HSF1 in the development of hepatocellular carcinoma (HCC). To clarify the functional role of HSF1 in HCC, we established HSF1-knockdown (HSF1 KD) KYN2 HCC cells by stably expressing either small hairpin RNA (shRNA) against HSF1 (i.e. HSF1 KD) or control shRNA (HSF1 control). Tumorigenicity was significantly reduced in orthotopic mice with HSF1 KD cells compared with those with HSF1 control cells. Reduced tumorigenesis in HSF1 KD cells appeared attributable to increased apoptosis and decreased proliferation. Tumor necrosis factor-α-induced apoptosis was increased in HSF1 KD cells and HSF1(-/-) mouse hepatocytes compared with controls. Decreased expression of IκB kinase γ, a positive regulator of nuclear factor-κB, was also observed in HSF1 KD cells and HSF1(-/-) mouse hepatocytes. Furthermore, expression of bcl-2-associated athanogene domain 3 (BAG3) was dramatically reduced in HSF1 KD cells and HSF1(-/-) mouse hepatocytes. We also found that epidermal growth factor-stimulated mitogen-activated protein kinase signaling was impaired in HSF1 KD cells. Clinicopathological analysis demonstrated frequent overexpression of HSF1 in human HCCs. Significant correlations between HSF1 and BAG3 protein levels and prognosis were also observed. In summary, these results identify a mechanistic link between HSF1 and liver tumorigenesis and may provide as a potential molecular target for the development of anti-HCC therapies.


Subject(s)
Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/metabolism , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Transcription Factors/metabolism , Adult , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Cell Transformation, Neoplastic , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Hepatocytes , Humans , Immunoenzyme Techniques , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
7.
EMBO J ; 29(20): 3459-69, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20834230

ABSTRACT

Heat shock transcription factor 1 (HSF1) is an important regulator of protein homeostasis (proteostasis) by controlling the expression of major heat shock proteins (Hsps) that facilitate protein folding. However, it is unclear whether other proteostasis pathways are mediated by HSF1. Here, we identified novel targets of HSF1 in mammalian cells, which suppress the aggregation of polyglutamine (polyQ) protein. Among them, we show that one of the nuclear factor of activated T cells (NFAT) proteins, NFATc2, significantly inhibits polyQ aggregation in cells and is required for HSF1-mediated suppression of polyQ aggregation. NFAT deficiency accelerated disease progression including aggregation of a mutant polyQ-huntingtin protein and shortening of lifespan in R6/2 Huntington's disease mice. Furthermore, we found that HSF1 and NFAT cooperatively induce the expression of the scaffold protein PDZK3 and αB-crystallin, which facilitate the degradation of polyQ protein. These results show the first mechanistic basis for the observation that HSF1 has a much more profound effect on proteostasis than individual Hsp or combination of different Hsps, and suggest a new pathway for ameliorating protein-misfolding diseases.


Subject(s)
DNA-Binding Proteins/metabolism , NFATC Transcription Factors/metabolism , Peptides/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , DNA-Binding Proteins/genetics , Gene Expression Regulation , HeLa Cells , Heat Shock Transcription Factors , Humans , Huntingtin Protein , Life Expectancy , Mice , Mice, Inbred Strains , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , NFATC Transcription Factors/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/genetics , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
8.
Proc Natl Acad Sci U S A ; 108(4): 1681-6, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21205885

ABSTRACT

Aberrant transcriptional regulation in the brain is thought to be one of the key components of the pathogenesis and pathophysiology of neuropsychiatric disorders. Heat shock factors (HSFs) modulate cellular homeostasis through the control of gene expression. However, the roles of HSFs in brain function have yet to be elucidated fully. In the present study, we attempted to clarify the role of HSF1-mediated gene regulation in neuronal and behavioral development using HSF1-deficient (HSF1(-/-)) mice. We found granule neurons of aberrant morphology and impaired neurogenesis in the dentate gyrus of HSF1(-/-) mice. In addition, HSF1(-/-) mice showed aberrant affective behavior, including reduced anxiety and sociability but increased depression-like behavior and aggression. Furthermore, HSF1 deficiency enhanced behavioral vulnerability to repeated exposure to restraint stress. Importantly, rescuing the HSF1 deficiency in the neonatal but not the adult hippocampus reversed the aberrant anxiety and depression-like behaviors. These results indicate a crucial role for hippocampal HSF1 in neuronal and behavioral development. Analysis of the molecular mechanisms revealed that HSF1 directly modulates the expression of polysialyltransferase genes, which then modulate polysialic acid-neural cell adhesion molecule (PSA-NCAM) levels in the hippocampus. Enzymatic removal of PSA from the neonatal hippocampus resulted in aberrant behavior during adulthood, similar to that observed in HSF1(-/-) mice. Thus, these results suggest that one role of HSF1 is to control hippocampal PSA-NCAM levels through the transcriptional regulation of polysialyltransferases, a process that might be involved in neuronal and behavioral development in mice.


Subject(s)
Behavior, Animal/physiology , DNA-Binding Proteins/metabolism , Hippocampus/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Anxiety/physiopathology , Base Sequence , Blotting, Western , DNA-Binding Proteins/genetics , Dendritic Spines/physiology , Feeding Behavior/physiology , Female , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors , Hippocampus/cytology , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Molecular Sequence Data , Motor Activity/physiology , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Neurogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sialic Acids/genetics , Sialic Acids/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Transcription Factors/genetics
9.
Pharmacology ; 91(1-2): 104-11, 2013.
Article in English | MEDLINE | ID: mdl-23328693

ABSTRACT

In this study, we investigated the protective effect of glutamine on barrier dysfunction induced by ethanol, by using human epithelial colorectal adenocarcinoma cells (Caco-2). Our results show that addition of glutamine to culture medium significantly improved the disruption of integrity caused by ethanol, which was associated with increased expression of heat shock protein 70 (Hsp70). Ethanol exposure moderately activates heat shock factor 1 (HSF1), which was characterized by increased DNA-binding activity and phosphorylation status of HSF1. Remarkably, glutamine treatment enhanced ethanol-mediated expression of Hsp70 and activation of HSF1. Up-regulation of Hsp70 by pretreatment with heat stress also promoted recovery from the ethanol-induced barrier dysfunction. Taken together, these observations indicate that glutamine protects the intestinal barrier function in Caco-2 cells, in part by modulating HSF1-mediated Hsp70 expression.


Subject(s)
DNA-Binding Proteins/metabolism , Epithelial Cells/drug effects , Glutamine/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Transcription Factors/metabolism , Caco-2 Cells , Colon , Epithelial Cells/metabolism , Ethanol , Heat Shock Transcription Factors , Humans , Inulin/metabolism , Zonula Occludens-1 Protein/metabolism
10.
FEBS Lett ; 597(13): 1702-1717, 2023 07.
Article in English | MEDLINE | ID: mdl-36971000

ABSTRACT

Upon heat shock, activated heat shock transcription factor 1 (HSF1) binds to the heat shock response elements (HSEs) in the promoters of mammalian heat shock protein (HSP)-encoding genes and recruits the preinitiation complex and coactivators, including Mediator. These transcriptional regulators may be concentrated in phase-separated condensates around the promoters, but they are too minute to be characterized in detail. We herein established HSF1-/- mouse embryonic fibroblasts harbouring HSP72-derived multiple HSE arrays and visualized the condensates of fluorescent protein-tagged HSF1 with liquid-like properties upon heat shock. Using this experimental system, we demonstrate that endogenous MED12, a subunit of Mediator, is concentrated in artificial HSF1 condensates upon heat shock. Furthermore, the knockdown of MED12 markedly reduces the size of condensates, suggesting an important role for MED12 in HSF1 condensate formation.


Subject(s)
DNA-Binding Proteins , Fibroblasts , Animals , Mice , Heat Shock Transcription Factors/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Transcription Factors/metabolism , Heat-Shock Response/genetics , Mammals/metabolism
11.
J Immunol ; 184(2): 1041-8, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018623

ABSTRACT

The febrile response is a complex physiological reaction to disease, including a cytokine-mediated increase in body temperature and the activation of inflammatory systems. Fever has beneficial roles in terms of disease prognosis, partly by suppressing the expression of inflammatory cytokines. However, the molecular mechanisms underlining the fever-mediated suppression of inflammatory gene expression have not been clarified. In this study, we showed that heat shock suppresses LPS-induced expression of IL-6, a major pyrogenic cytokine, in mouse embryonic fibroblasts and macrophages. Heat shock transcription factor 1 (HSF1) activated by heat shock induced the expression of activating transcription factor (ATF) 3, a negative regulator of IL-6, and ATF3 was necessary for heat-mediated suppression of IL-6, indicating a fever-mediated feedback loop consisting of HSF1 and ATF3. A comprehensive analysis of inflammatory gene expression revealed that heat pretreatment suppresses LPS-induced expression of most genes (86%), in part (67%) via ATF3. When HSF1-null and ATF3-null mice were injected with LPS, they expressed much higher levels of IL-6 than wild-type mice, resulting in an exaggerated febrile response. These results demonstrate a novel inhibitory pathway for inflammatory cytokines.


Subject(s)
Activating Transcription Factor 3/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation/immunology , Heat-Shock Response/immunology , Interleukin-6/antagonists & inhibitors , Transcription Factors/physiology , Activating Transcription Factor 3/genetics , Animals , Feedback, Physiological , Fever , Fibroblasts/immunology , Fibroblasts/metabolism , Heat Shock Transcription Factors , Interleukin-6/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Repressor Proteins
12.
Nat Commun ; 13(1): 4355, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906200

ABSTRACT

Transcriptional regulation by RNA polymerase II is associated with changes in chromatin structure. Activated and promoter-bound heat shock transcription factor 1 (HSF1) recruits transcriptional co-activators, including histone-modifying enzymes; however, the mechanisms underlying chromatin opening remain unclear. Here, we demonstrate that HSF1 recruits the TRRAP-TIP60 acetyltransferase complex in HSP72 promoter during heat shock in a manner dependent on phosphorylation of HSF1-S419. TRIM33, a bromodomain-containing ubiquitin ligase, is then recruited to the promoter by interactions with HSF1 and a TIP60-mediated acetylation mark, and cooperates with the related factor TRIM24 for mono-ubiquitination of histone H2B on K120. These changes in histone modifications are triggered by phosphorylation of HSF1-S419 via PLK1, and stabilize the HSF1-transcription complex in HSP72 promoter. Furthermore, HSF1-S419 phosphorylation is constitutively enhanced in and promotes proliferation of melanoma cells. Our results provide mechanisms for HSF1 phosphorylation-dependent establishment of an active chromatin status, which is important for tumorigenesis.


Subject(s)
Chromatin , Histones , Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/genetics , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Histones/metabolism , Humans , Lysine Acetyltransferase 5/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Transcription Factors/genetics
13.
FEBS Lett ; 595(14): 1933-1948, 2021 07.
Article in English | MEDLINE | ID: mdl-34056708

ABSTRACT

Activated and promoter-bound heat-shock transcription factor 1 (HSF1) induces RNA polymerase II recruitment upon heat shock, and this is facilitated by the core Mediator in Drosophila and yeast. Another Mediator module, CDK8 kinase module (CKM), consisting of four subunits including MED12 and CDK8, plays a negative or positive role in the regulation of transcription; however, its involvement in HSF1-mediated transcription remains unclear. We herein demonstrated that HSF1 interacted with MED12 and recruited MED12 and CDK8 to the HSP70 promoter during heat shock in mammalian cells. The kinase activity of CDK8 (and its paralog CDK19) promoted HSP70 expression partly by phosphorylating HSF1-S326 and maintained proteostasis capacity. These results indicate an important role for CKM in the protection of cells against proteotoxic stress.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Heat Shock Transcription Factors/genetics , Heat-Shock Response/genetics , Mediator Complex/genetics , Multiprotein Complexes/genetics , Proteostasis/genetics , Animals , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Fibroblasts , Gene Expression Regulation , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat Shock Transcription Factors/metabolism , Humans , Mediator Complex/metabolism , Mice , Multiprotein Complexes/metabolism , Neurons , Osteoblasts , Phosphorylation , Protein Binding , Signal Transduction , Transcription, Genetic
14.
FEBS Open Bio ; 10(6): 1135-1148, 2020 06.
Article in English | MEDLINE | ID: mdl-32302062

ABSTRACT

The mitochondrial unfolded protein response (UPRmt ) is characterized by the transcriptional induction of mitochondrial chaperone and protease genes in response to impaired mitochondrial proteostasis and is regulated by ATF5 and CHOP in mammalian cells. However, the detailed mechanisms underlying the UPRmt are currently unclear. Here, we show that HSF1 is required for activation of mitochondrial chaperone genes, including HSP60, HSP10, and mtHSP70, in mouse embryonic fibroblasts during inhibition of matrix chaperone TRAP1, protease Lon, or electron transfer complex 1 activity. HSF1 bound constitutively to mitochondrial chaperone gene promoters, and we observed that its occupancy was remarkably enhanced at different levels during the UPRmt . Furthermore, HSF1 supported the maintenance of mitochondrial function under the same conditions. These results demonstrate that HSF1 is required for induction of mitochondrial chaperones during the UPRmt , and thus, it may be one of the guardians of mitochondrial function under conditions of impaired mitochondrial proteostasis.


Subject(s)
Heat Shock Transcription Factors/metabolism , Mitochondria/metabolism , Molecular Chaperones/genetics , Unfolded Protein Response/genetics , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors/genetics , Humans , Membrane Potential, Mitochondrial/genetics , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Brain Res ; 1212: 9-17, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18445491

ABSTRACT

Mechanisms of age-related hearing loss (ARHL) have not been elucidated as aging processes are extremely complex. Although oxidative stress and apoptotic cell death are involved in progression of ARHL, number of trial to treat ARHL is limited. Heat shock response is characterized by induction of heat shock proteins (HSPs) in response to stresses such as heat shock, which diminishes during aging. HSPs act as molecular chaperones, and some HSPs also inhibit apoptotic pathways. Here, we examined age-related expression of HSPs in the cochlea of ARHL model DBA/2J mice and control CBA/N mice. Western blot assay revealed that CBA/N mice showed constant expression of Hsp70 and Hsp110 with age, but not in DBA/2J mice. The result suggests that pharmacological upregulation of HSPs might attenuate ARHL. We administered DBA/2J mice with food containing geranylgeranylacetone (GGA) that induces HSPs in the cochlea, and found that its administration suppresses ARHL examined by ABR test and histological examination though protection is specific for the apical part of the cochlea. These results demonstrate that dietary supplementation of GGA could be an effective therapeutic strategy for treatment of ARHL.


Subject(s)
Aging , Anti-Ulcer Agents/administration & dosage , Diterpenes/administration & dosage , Presbycusis/drug therapy , Animals , Brain/metabolism , Cell Count , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Gene Expression Regulation/drug effects , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Hair Cells, Auditory/pathology , Male , Mice , Mice, Inbred DBA , Presbycusis/pathology , Presbycusis/physiopathology , Psychophysics
16.
Mol Cell Biol ; 38(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29661921

ABSTRACT

The heat shock response (HSR) is characterized by the rapid and robust induction of heat shock proteins (HSPs), including HSP70, in response to heat shock and is regulated by heat shock transcription factor 1 (HSF1) in mammalian cells. Poly(ADP-ribose) polymerase 1 (PARP1), which can form a complex with HSF1 through the scaffold protein PARP13, has been suggested to be involved in the HSR. However, its effects on and the regulatory mechanisms of the HSR are not well understood. Here we show that prior to heat shock, the HSF1-PARP13-PARP1 complex binds to the HSP70 promoter. In response to heat shock, activated and auto-PARylated PARP1 dissociates from HSF1-PARP13 and is redistributed throughout the HSP70 locus. Remarkably, chromatin in the HSP70 promoter is initially PARylated at high levels and decondensed, whereas chromatin in the gene body is moderately PARylated afterwards. Activated HSF1 then binds to the promoter efficiently and promotes the HSR. Chromatin PARylation and HSF1 binding to the promoter are also facilitated by the phosphorylation-dependent dissociation of PARP13. Furthermore, the HSR and proteostasis capacity are reduced by pretreatment with genotoxic stresses, which disrupt the ternary complex. These results illuminate one of the priming mechanisms of the HSR that facilitates the binding of HSF1 to DNA during heat shock.


Subject(s)
DNA/metabolism , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/physiology , Poly (ADP-Ribose) Polymerase-1/metabolism , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA Damage , Gene Knockdown Techniques , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Humans , Mice , Models, Biological , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic , Protein Binding , Proteostasis , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Immunobiology ; 212(1): 11-8, 2007.
Article in English | MEDLINE | ID: mdl-17270705

ABSTRACT

Experimental autoimmune uveoretinitis (EAU) serves as a model of human endogeneous uveitis. In the present study we examined whether induction of heat shock protein (HSP) 70 by oral geranylgeranylacetone (GGA) administration had a therapeutic effect on murine EAU. When C57BL/6 mice that had received oral administration of GGA (500mg/kg) were immunized with interphotoreceptor retinoid-binding protein (IRBP)-derived peptide plus adjuvants, the expression levels of HSP70 mRNA and protein were rapidly and transiently upregulated in eyes of the GGA-treated mice, compared with those from vehicle-pretreated and IRBP-immunized mice. The antigen-specific T cell proliferation was partially suppressed in these mice treated with GGA. The mean EAU scores of the GGA-treated mice on day 21 and 28 (2.4+/-0.2 and 2.1+/-0.2, respectively) were significantly lower than those in the controls (3.0+/-0.1 and 2.6+/-0.2, respectively p<0.01). The histopathological severity of the GGA-treated mice (average 0.33) was markedly milder than that in the controls (average 1.63, p<0.05) at day 21. The present findings demonstrate that the pharmacological induction of HSP70 may be applicable to the amelioration of ocular autoimmune diseases.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Autoimmune Diseases/prevention & control , Diterpenes/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Uveitis/prevention & control , Animals , Autoimmune Diseases/metabolism , Cell Proliferation/drug effects , Eye/drug effects , Eye/pathology , Female , HSP70 Heat-Shock Proteins/drug effects , Immunohistochemistry , Mice , Mice, Inbred C57BL , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , Uveitis/metabolism
18.
Mol Cell Biol ; 23(16): 5882-95, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897157

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.


Subject(s)
Avian Proteins , Cell Death , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Heat-Shock Proteins , Trans-Activators/physiology , Adenoviridae/genetics , Alanine/chemistry , Amino Acid Sequence , Animals , Blotting, Northern , Cell Line , Chickens , Chromatography, Gel , DNA/metabolism , DNA, Complementary/metabolism , Gene Deletion , Heat Shock Transcription Factors , Humans , Models, Genetic , Molecular Sequence Data , Mutation , Phylogeny , Protein Binding , Protein Structure, Tertiary , Subcellular Fractions , Temperature , Time Factors , Trans-Activators/metabolism , Transcription Factors
19.
Nat Commun ; 8(1): 1638, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29158484

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) is involved in DNA repair, chromatin structure, and transcription. However, the mechanisms that regulate PARP1 distribution on DNA are poorly understood. Here, we show that heat shock transcription factor 1 (HSF1) recruits PARP1 through the scaffold protein PARP13. In response to DNA damage, activated and auto-poly-ADP-ribosylated PARP1 dissociates from HSF1-PARP13, and redistributes to DNA lesions and DNA damage-inducible gene loci. Histone deacetylase 1 maintains PARP1 in the ternary complex by inactivating PARP1 through deacetylation. Blocking ternary complex formation impairs redistribution of PARP1 during DNA damage, which reduces gene expression and DNA repair. Furthermore, ternary complex formation and PARP1 redistribution protect cells from DNA damage by promoting DNA repair, and support growth of BRCA1-null mammary tumors, which are sensitive to PARP inhibitors. Our findings identify HSF1 as a regulator of genome integrity and define this function as a guarding mechanism for a specific type of mammary tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/metabolism , DNA Repair , Heat Shock Transcription Factors/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA-Binding Proteins/metabolism , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , DNA Damage , Female , Genomic Instability , Heat Shock Transcription Factors/genetics , Humans , Mice , Poly (ADP-Ribose) Polymerase-1/genetics , Protein Binding , RNA-Binding Proteins/genetics
20.
Endocrinology ; 158(8): 2648-2658, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28575284

ABSTRACT

Testicular testosterone synthesis begins with cholesterol transport into mitochondria via steroidogenic acute regulatory (StAR) protein in Leydig cells. Acute heat stress is known to obstruct testicular steroidogenesis by transcriptional repression of StAR. In contrast, chronic heat stress such as cryptorchidism or varicocele generally does not affect testicular steroidogenesis, suggesting that Leydig cells adapt to heat stress and retain their steroid synthesis ability. However, the mechanisms of the stress response in steroid-producing cells are unclear. We examined the relationship between the heat stress response and heat shock factor 1 (HSF1), which protects cells from proteotoxic stress by inducing heat shock protein as a molecular chaperone. The influences of HSF1 deficiency on cholesterol transport by StAR and the expression of steroidogenic enzymes under chronic heat stress were studied in testes of HSF1-knockout (HSF1KO) mice with experimental cryptorchidism. StAR protein in wild-type-cryptorchid mice was transiently decreased after induction of cryptorchidism and then gradually returned to basal levels. In contrast, StAR protein in HSF1KO mice continued to decrease and failed to recover, resulting in impaired serum testosterone. StAR messenger RNA was not decreased with cryptorchidism, indicating that posttranslational modification of StAR, not its transcription, was obstructed in cryptorchidism. Other steroidogenic enzymes, including CYP11A1, 3ß-HSD, and CYP17A1, were not decreased. Lipid droplets were increased in the cytosol of HSF1KO-cryptorchid mice, suggesting dysfunctional cholesterol transportation. These findings provide insight into the role of HSF1 in Leydig cell steroidogenesis, suggesting that it maintains cholesterol transport by recovering StAR under chronic heat stress.


Subject(s)
Cholesterol/metabolism , DNA-Binding Proteins/metabolism , Leydig Cells/metabolism , Testosterone/biosynthesis , Transcription Factors/metabolism , Animals , Biological Transport , Cryptorchidism , DNA-Binding Proteins/genetics , Epithelium , Gene Expression Regulation/physiology , Heat Shock Transcription Factors , Hot Temperature , Male , Mice , Mice, Knockout , Stress, Physiological , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL