1
|
Khan S, Aggarwal S, Bhatia P, Yadav AK, Kumar Y, Veerapu NS. Glucose and glutamine drive hepatitis E virus replication. Arch Virol 2024; 169:233. [PMID: 39476184 DOI: 10.1007/s00705-024-06160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/03/2024] [Indexed: 11/30/2024]
Abstract
Viruses have undergone evolutionary adaptations to tune their utilization of carbon sources, enabling them to extract specific cellular substrates necessary for their replication. The lack of a reliable cell culture system and a small-animal model has hampered our understanding of the molecular mechanism of replication of hepatitis E virus (HEV) genotype 1. Our recent identification of a replicative ensemble of mutant HEV RNA libraries has allowed us to study the metabolic prerequisites for HEV replication. Initial assessments revealed increased glucose and glutamine utilization during HEV replication. Inhibition of glycolysis and glycolysis + glutaminolysis reduced the levels of HEV replication to similar levels. An integrated analysis of protein-metabolite pathways suggests that HEV replication markedly alters glycolysis, the TCA cycle, and glutamine-associated metabolic pathways. Cells supporting HEV replication showed a requirement for fructose-6-phosphate and glutamine utilization through the hexosamine biosynthetic pathway (HBP), stimulating HSP70 expression to facilitate virus replication. Observations of mannose utilization and glutamine dependence suggest a crucial role of the HBP in supporting HEV replication. Inhibition of glycolysis and HSP70 activity or knockdown of glutamine fructose-6-phosphate amidotransferase expression led to a substantial reduction in HEV RNA and ORF2 expression accompanied by a significant decrease in HSP70 levels. This study demonstrates that glucose and glutamine play critical roles in facilitating HEV replication.
Collapse
Affiliation(s)
- Shaheen Khan
- Virology Section, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, UP201314, India
| | - Suruchi Aggarwal
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad, Haryana, India
| | - Pooja Bhatia
- Virology Section, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, UP201314, India
| | - Amit Kumar Yadav
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad, Haryana, India
| | - Yashwant Kumar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad, Haryana, India
| | - Naga Suresh Veerapu
- Virology Section, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, UP201314, India.
| |
Collapse
|
2
|
Kose S, Ogawa Y, Imamoto N. Thermal Stress and Nuclear Transport. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:61-78. [PMID: 39289274 DOI: 10.1007/978-981-97-4584-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Nuclear transport is the basis for the biological reaction of eukaryotic cells, as it is essential to coordinate nuclear and cytoplasmic events separated by nuclear envelope. Although we currently understand the basic molecular mechanisms of nuclear transport in detail, many unexplored areas remain. For example, it is believed that the regulations and biological functions of the nuclear transport receptors (NTRs) highlights the significance of the transport pathways in physiological contexts. However, physiological significance of multiple parallel transport pathways consisting of more than 20 NTRs is still poorly understood, because our knowledge of each pathway, regarding their substrate information or how they are differently regulated, is still limited. In this report, we describe studies showing how nuclear transport systems in general are affected by temperature rises, namely, thermal stress or heat stress. We will then focus on Importin α family members and unique transport factor Hikeshi, because these two NTRs are affected in heat stress. Our present review will provide an additional view to point out the importance of diversity of the nuclear transport pathways in eukaryotic cells.
Collapse
Affiliation(s)
- Shingo Kose
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan.
| | - Yutaka Ogawa
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan.
| | - Naoko Imamoto
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan.
| |
Collapse
|
3
|
Waldhart AN, Lau KH, Dykstra H, Avequin T, Wu N. Optimal HSF1 activation in response to acute cold stress in BAT requires nuclear TXNIP. iScience 2023; 26:106538. [PMID: 37168572 PMCID: PMC10164894 DOI: 10.1016/j.isci.2023.106538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/04/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
While TXNIP (thioredoxin interacting protein) in the plasma membrane and vesicular location is known to negatively regulate cellular glucose uptake by facilitating glucose transporter endocytosis, the function of TXNIP in the nucleus is far less understood. Herein, we sought to determine the function of nuclear TXNIP in vivo, using a new HA-tagged TXNIP knock-in mouse model. We observed that TXNIP can be found in the nucleus of a variety of cells from different tissues including hepatocytes (liver), enterocytes (small intestine), exocrine cells (pancreas), and brown adipocytes (BAT). Further investigations into the role of nuclear TXNIP in BAT revealed that cold stress rapidly and transiently activated HSF1 (heat shock factor 1). HSF1 interaction with TXNIP during its activation is required for optimal HSF1 directed cold shock response in BAT.
Collapse
Affiliation(s)
| | - Kin H. Lau
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | | | - Ning Wu
- Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
4
|
Kose S, Imai K, Watanabe A, Nakai A, Suzuki Y, Imamoto N. Lack of Hikeshi activates HSF1 activity under normal conditions and disturbs the heat-shock response. Life Sci Alliance 2022; 5:5/9/e202101241. [PMID: 35580988 PMCID: PMC9113944 DOI: 10.26508/lsa.202101241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 01/09/2023] Open
Abstract
Hikeshi mediates the nuclear import of the molecular chaperone HSP70 under heat-shock (acute heat stress) conditions, which is crucial for recovery from cellular damage. The cytoplasmic function of HSP70 is well studied, but its nuclear roles, particularly under nonstressed conditions, remain obscure. Here, we show that Hikeshi regulates the nucleocytoplasmic distribution of HSP70 not only under heat-shock conditions but also under nonstressed conditions. Nuclear HSP70 affects the transcriptional activity of HSF1 and nuclear proteostasis under nonstressed conditions. Depletion of Hikeshi induces a reduction in nuclear HSP70 and up-regulation of the mRNA expression of genes regulated by HSF1 under nonstressed conditions. In addition, the heat-shock response is impaired in Hikeshi-knockout cells. Our results suggest that HSF1 transcriptional activity is tightly regulated by nuclear HSP70 because nuclear-localized Hsp70 effectively suppresses transcriptional activity in a dose-dependent manner. Furthermore, the cytotoxicity of nuclear pathologic polyglutamine proteins was increased by Hikeshi depletion. Thus, proper nucleocytoplasmic distribution of HSP70, mediated by Hikeshi, is required for nuclear proteostasis and adaptive response to heat shock.
Collapse
Affiliation(s)
- Shingo Kose
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Japan,Correspondence: ;
| | - Kenichiro Imai
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Ai Watanabe
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Naoko Imamoto
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Japan,Correspondence: ;
| |
Collapse
|
5
|
Reyes A, Navarro AJ, Diethelm-Varela B, Kalergis AM, González PA. Is there a role for HSF1 in viral infections? FEBS Open Bio 2022; 12:1112-1124. [PMID: 35485710 PMCID: PMC9157408 DOI: 10.1002/2211-5463.13419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/29/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022] Open
Abstract
Cells undergo numerous processes to adapt to new challenging conditions and stressors. Heat stress is regulated by a family of heat shock factors (HSFs) that initiate a heat shock response by upregulating the expression of heat shock proteins (HSPs) intended to counteract cellular damage elicited by increased environmental temperature. Heat shock factor 1 (HSF1) is known as the master regulator of the heat shock response and upon its activation induces the transcription of genes that encode for molecular chaperones, such as HSP40, HSP70, and HSP90. Importantly, an accumulating body of studies relates HSF1 with viral infections; the induction of fever during viral infection may activate HSF1 and trigger a consequent heat shock response. Here, we review the role of HSF1 in different viral infections and its impact on the health outcome for the host. Studying the relationship between HSF1 and viruses could open new potential therapeutic strategies given the availability of drugs that regulate the activation of this transcription factor.
Collapse
Affiliation(s)
- Antonia Reyes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile
| | - Areli J Navarro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina Pontificia, Universidad Católica de Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile
| |
Collapse
|
6
|
Kurop MK, Huyen CM, Kelly JH, Blagg BSJ. The heat shock response and small molecule regulators. Eur J Med Chem 2021; 226:113846. [PMID: 34563965 PMCID: PMC8608735 DOI: 10.1016/j.ejmech.2021.113846] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 01/09/2023]
Abstract
The heat shock response (HSR) is a highly conserved cellular pathway that is responsible for stress relief and the refolding of denatured proteins [1]. When a host cell is exposed to conditions such as heat shock, ischemia, or toxic substances, heat shock factor-1 (HSF-1), a transcription factor, activates the genes that encode for the heat shock proteins (Hsps), which are a family of proteins that work alongside other chaperones to relieve stress and refold proteins that have been denatured (Burdon, 1986) [2]. Along with the refolding of denatured proteins, Hsps facilitate the removal of misfolded proteins by escorting them to degradation pathways, thereby preventing the accumulation of misfolded proteins [3]. Research has indicated that many pathological conditions, such as diabetes, cancer, neuropathy, cardiovascular disease, and aging have a negative impact on HSR function and are commonly associated with misfolded protein aggregation [4,5]. Studies indicate an interplay between mitochondrial homeostasis and HSF-1 levels can impact stress resistance, proteostasis, and malignant cell growth, which further support the role of Hsps in pathological and metabolic functions [6]. On the other hand, Hsp activation by specific small molecules can induce the heat shock response, which can afford neuroprotection and other benefits [7]. This review will focus on the modulation of Hsps and the HSR as therapeutic options to treat these conditions.
Collapse
Affiliation(s)
- Margaret K Kurop
- Warren Center for Drug Discovery, Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Cormac M Huyen
- Warren Center for Drug Discovery, Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - John H Kelly
- Warren Center for Drug Discovery, Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Brian S J Blagg
- Warren Center for Drug Discovery, Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
7
|
Kerschbaum S, Wegrostek C, Riegel E, Czerny T. Senescence in a cell culture model for burn wounds. Exp Mol Pathol 2021; 122:104674. [PMID: 34437877 DOI: 10.1016/j.yexmp.2021.104674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/25/2021] [Accepted: 08/15/2021] [Indexed: 11/29/2022]
Abstract
Thermal injuries cause severe damage on the cellular and tissue level and are considered especially challenging in the clinical routine. Complex interactions of different cell types and pathways dictate the formation of burn wounds. Thus, complications like burn wound progression, where so far viable tissue becomes necrotic and the size and depth of the wound increases, are difficult to explain, mainly due to the lack of simple model systems. We tested the behavior of human fibroblasts after heat treatment. A prominent response of the cells is to activate the heat shock response (HSR), which is one of the primary emergency mechanisms of the cell to proteotoxic stress factors such as heat. However, after a powerful but not lethal heat shock we observed a delayed activation of the HSR. Extending this model system, we further investigated these static cells and observed the emergence of senescent cells. In particular, the cells became β-galactosidase positive, increased p16 levels and developed a senescence-associated secretory phenotype (SASP). The secretion of cytokines like IL-6 is reminiscent of burn wounds and generates a bystander effect in so far non-senescent cells. In agreement with burn wounds, a wave of cytokine secretion enhanced by invading immune cells could explain complications like burn wound progression. A simple cell culture model can thus be applied for the analysis of highly complex conditions in human tissues.
Collapse
Affiliation(s)
- Sarah Kerschbaum
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Christina Wegrostek
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Elisabeth Riegel
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Thomas Czerny
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria.
| |
Collapse
|
8
|
The ABL2 kinase regulates an HSF1-dependent transcriptional program required for lung adenocarcinoma brain metastasis. Proc Natl Acad Sci U S A 2020; 117:33486-33495. [PMID: 33318173 PMCID: PMC7777191 DOI: 10.1073/pnas.2007991117] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Among all cancer types, lung cancer patients exhibit the highest prevalence of brain metastasis, often associated with cognitive impairment, seizures, decline in quality of life, and decreased survival. Limited therapeutic options are currently available to treat brain metastasis. A comprehensive understanding of the signaling pathways and transcriptional networks required for survival and growth of brain-metastatic cancer cells is needed to develop effective strategies to treat this disease. Here, we report that the Heat Shock Transcription Factor 1 (HSF1) is upregulated in brain-metastatic lung cancer cells and is required for brain metastasis in mice. Importantly, we show that the HSF1-dependent expression of E2F target genes implicated in cell cycle progression and survival is decreased by blood–brain barrier-penetrant ABL allosteric inhibitors. Brain metastases are the most common intracranial tumors in adults and are associated with increased patient morbidity and mortality. Limited therapeutic options are currently available for the treatment of brain metastasis. Here, we report on the discovery of an actionable signaling pathway utilized by metastatic tumor cells whereby the transcriptional regulator Heat Shock Factor 1 (HSF1) drives a transcriptional program, divergent from its canonical role as the master regulator of the heat shock response, leading to enhanced expression of a subset of E2F transcription factor family gene targets. We find that HSF1 is required for survival and outgrowth by metastatic lung cancer cells in the brain parenchyma. Further, we identify the ABL2 tyrosine kinase as an upstream regulator of HSF1 protein expression and show that the Src-homology 3 (SH3) domain of ABL2 directly interacts with HSF1 protein at a noncanonical, proline-independent SH3 interaction motif. Pharmacologic inhibition of the ABL2 kinase using small molecule allosteric inhibitors, but not ATP-competitive inhibitors, disrupts this interaction. Importantly, knockdown as well as pharmacologic inhibition of ABL2 using allosteric inhibitors impairs expression of HSF1 protein and HSF1-E2F transcriptional gene targets. Collectively, these findings reveal a targetable ABL2-HSF1-E2F signaling pathway required for survival by brain-metastatic tumor cells.
Collapse
|
9
|
Carpenter RL, Gökmen-Polar Y. HSF1 as a Cancer Biomarker and Therapeutic Target. Curr Cancer Drug Targets 2020; 19:515-524. [PMID: 30338738 DOI: 10.2174/1568009618666181018162117] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/30/2018] [Accepted: 09/15/2018] [Indexed: 12/30/2022]
Abstract
Heat shock factor 1 (HSF1) was discovered in 1984 as the master regulator of the heat shock response. In this classical role, HSF1 is activated following cellular stresses such as heat shock that ultimately lead to HSF1-mediated expression of heat shock proteins to protect the proteome and survive these acute stresses. However, it is now becoming clear that HSF1 also plays a significant role in several diseases, perhaps none more prominent than cancer. HSF1 appears to have a pleiotropic role in cancer by supporting multiple facets of malignancy including migration, invasion, proliferation, and cancer cell metabolism among others. Because of these functions, and others, of HSF1, it has been investigated as a biomarker for patient outcomes in multiple cancer types. HSF1 expression alone was predictive for patient outcomes in multiple cancer types but in other instances, markers for HSF1 activity were more predictive. Clearly, further work is needed to tease out which markers are most representative of the tumor promoting effects of HSF1. Additionally, there have been several attempts at developing small molecule inhibitors to reduce HSF1 activity. All of these HSF1 inhibitors are still in preclinical models but have shown varying levels of efficacy at suppressing tumor growth. The growth of research related to HSF1 in cancer has been enormous over the last decade with many new functions of HSF1 discovered along the way. In order for these discoveries to reach clinical impact, further development of HSF1 as a biomarker or therapeutic target needs to be continued.
Collapse
Affiliation(s)
- Richard L Carpenter
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Bloomington, IN 47405, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, IN 47405, United States.,Department of Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, United States
| | - Yesim Gökmen-Polar
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Bloomington, IN 47405, United States.,Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
10
|
Agarwal S, Ganesh S. Perinuclear mitochondrial clustering, increased ROS levels, and HIF1 are required for the activation of HSF1 by heat stress. J Cell Sci 2020; 133:jcs245589. [PMID: 32503939 DOI: 10.1242/jcs.245589] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/19/2020] [Indexed: 08/31/2023] Open
Abstract
The heat shock response (HSR) is a conserved cellular defensive response against stresses such as temperature, oxidative stress and heavy metals. A significant group of players in the HSR is the set of molecular chaperones known as heat shock proteins (HSPs), which assist in the refolding of unfolded proteins and prevent the accumulation of damaged proteins. HSP genes are activated by the HSF1 transcription factor, a master regulator of the HSR pathway. A variety of stressors activate HSF1, but the key molecular players and the processes that directly contribute to HSF1 activation remain unclear. In this study, we show that heat shock induces perinuclear clustering of mitochondria in mammalian cells, and this clustering is essential for activation of the HSR. We also show that this perinuclear clustering of mitochondria results in increased levels of reactive oxygen species in the nucleus, leading to the activation of hypoxia-inducible factor-1α (HIF-1α). To conclude, we provide evidence to suggest that HIF-1α is one of the crucial regulators of HSF1 and that HIF-1α is essential for activation of the HSR during heat shock.
Collapse
Affiliation(s)
- Saloni Agarwal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur 208016, India
| |
Collapse
|
11
|
Guilbert M, Anquez F, Pruvost A, Thommen Q, Courtade E. Protein level variability determines phenotypic heterogeneity in proteotoxic stress response. FEBS J 2020; 287:5345-5361. [PMID: 32222033 DOI: 10.1111/febs.15297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/03/2020] [Accepted: 03/16/2020] [Indexed: 01/19/2023]
Abstract
Cell-to-cell variability in stress response is a bottleneck for the construction of accurate and predictive models which could guide clinical diagnosis and treatment of certain diseases, for example, cancer. Indeed, such phenotypic heterogeneity can lead to fractional killing and persistence of a subpopulation of cells which are resistant to a given treatment. The heat shock response network plays a major role in protecting the proteome against several types of injuries. Here, we combine high-throughput measurements and mathematical modeling to unveil the molecular origin of the phenotypic variability in the heat shock response network. Although the mean response coincides with known biochemical measurements, we found a surprisingly broad diversity in single-cell dynamics with a continuum of response amplitudes and temporal shapes for several stimulus strengths. We theoretically predict that the broad phenotypic heterogeneity is due to network ultrasensitivity together with variations in the expression level of chaperones controlled by the transcription factor heat shock factor 1. Furthermore, we experimentally confirm this prediction by mapping the response amplitude to chaperone and heat shock factor 1 expression levels.
Collapse
Affiliation(s)
- Marie Guilbert
- UMR 8523, PhLAM - Physique des Lasers Atomes et Molécules, CNRS, Université de Lille, France
| | - François Anquez
- UMR 8523, PhLAM - Physique des Lasers Atomes et Molécules, CNRS, Université de Lille, France
| | - Alexandra Pruvost
- UMR 8523, PhLAM - Physique des Lasers Atomes et Molécules, CNRS, Université de Lille, France
| | - Quentin Thommen
- UMR 8523, PhLAM - Physique des Lasers Atomes et Molécules, CNRS, Université de Lille, France
| | - Emmanuel Courtade
- UMR 8523, PhLAM - Physique des Lasers Atomes et Molécules, CNRS, Université de Lille, France
| |
Collapse
|
12
|
Robert HML, Savatier J, Vial S, Verghese J, Wattellier B, Rigneault H, Monneret S, Polleux J, Baffou G. Photothermal Control of Heat-Shock Protein Expression at the Single Cell Level. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1801910. [PMID: 29995322 DOI: 10.1002/smll.201801910] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/06/2018] [Indexed: 05/10/2023]
Abstract
Laser heating of individual cells in culture recently led to seminal studies in cell poration, fusion, migration, or nanosurgery, although measuring the local temperature increase in such experiments remains a challenge. Here, the laser-induced dynamical control of the heat-shock response is demonstrated at the single cell level, enabled by the use of light-absorbing gold nanoparticles as nanosources of heat and a temperature mapping technique based on quadriwave lateral shearing interferometry (QLSI) measurements. As it is label-free, this approach does not suffer from artifacts inherent to previously reported fluorescence-based temperature-mapping techniques and enables the use of any standard fluorescent labels to monitor in parallel the cell's response.
Collapse
Affiliation(s)
- Hadrien M L Robert
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
- PHASICS S.A., Parc technologique de Saint Aubin, Route de l'Orme des Merisiers, 91190, Saint Aubin, France
| | - Julien Savatier
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
| | - Stéphanie Vial
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
| | - Jacob Verghese
- Max Planck Institute of Biochemistry, Department of Cellular Biochemistry, 82152, Martinsried, Germany
| | - Benoit Wattellier
- PHASICS S.A., Parc technologique de Saint Aubin, Route de l'Orme des Merisiers, 91190, Saint Aubin, France
| | - Hervé Rigneault
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
| | - Serge Monneret
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
| | - Julien Polleux
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, 82152, Martinsried, Germany
- Center for NanoScience, Ludwig Maximilian University, 80799, Munich, Germany
| | - Guillaume Baffou
- Institut Fresnel, CNRS, Aix Marseille Univ, Centrale Marseille, Marseille, 13013, France
| |
Collapse
|
13
|
Zhao Z, Zhu J, Quan H, Wang G, Li B, Zhu W, Xie C, Lou L. X66, a novel N-terminal heat shock protein 90 inhibitor, exerts antitumor effects without induction of heat shock response. Oncotarget 2018; 7:29648-63. [PMID: 27105490 PMCID: PMC5045423 DOI: 10.18632/oncotarget.8818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/28/2016] [Indexed: 01/16/2023] Open
Abstract
Heat shock protein 90 (HSP90) is essential for cancer cells to assist the function of various oncoproteins, and it has been recognized as a promising target in cancer therapy. Although the HSP90 inhibitors in clinical trials have shown encouraging clinical efficacy, these agents induce heat shock response (HSR), which undermines their therapeutic effects. In this report, we detailed the pharmacologic properties of 4-(2-((1H-indol-3-yl)methylene)hydrazinyl)-N-(4-bromophenyl)-6-(3,5- dimethyl-1H -pyrazol-1-yl)-1,3,5-triazin-2-amine (X66), a novel and potent HSP90 inhibitor. X66 binds to the N-terminal domain in a different manner from the classic HSP90 inhibitors. Cellular study showed that X66 depleted HSP90 client proteins, resulted in cell cycle arrest and apoptosis, and inhibition of proliferation in cancer cell lines. X66 did not activate heat shock factor-1 (HSF-1) or stimulate transcription of HSPs. Moreover, the combination of X66 with HSP90 and proteasome inhibitors yielded synergistic cytotoxicity which was involved in X66-mediated abrogation of HSR through inhibition of HSF-1 activity. The intraperitoneal administration of X66 alone depleted client protein and inhibited tumor growth, and led to enhanced activity when combined with celastrol as compared to either agent alone in BT-474 xenograft model. Collectively, the HSP90 inhibitory action and the potent antitumor activity, with the anti-HSR action, promise X66 a novel HSP90-targeted agent, which merits further research and development.
Collapse
Affiliation(s)
- Zhixin Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianming Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haitian Quan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guimin Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bo Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiliang Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chengying Xie
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Liguang Lou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
14
|
Rashmi KC, Atreya HS, Harsha Raj M, Salimath BP, Aparna HS. A pyrrole-based natural small molecule mitigates HSP90 expression in MDA-MB-231 cells and inhibits tumor angiogenesis in mice by inactivating HSF-1. Cell Stress Chaperones 2017; 22:751-766. [PMID: 28717943 PMCID: PMC5573693 DOI: 10.1007/s12192-017-0802-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 12/15/2022] Open
Abstract
Heat shock proteins (HSPs), molecular chaperones, are crucial for the cancer cells to facilitate proper functioning of various oncoproteins involved in cell survival, proliferation, migration, and tumor angiogenesis. Tumor cells are said to be "addicted" to HSPs. HSPs are overexpressed in many cancers due to upregulation of transcription factor Heat-shock factor 1 (HSF-1), the multifaceted master regulator of heat shock response. Therefore, pharmacological targeting of HSPs via HSF-1 is an effective strategy to treat malignant cancers like triple negative breast cancer. In the current study, we evaluated the efficacy of a pyrrole derivative [bis(2-ethylhexyl)1H-pyrrole-3,4-dicarboxylate], TCCP, purified from leaves of Tinospora cordifolia for its ability to suppress heat shock response and angiogenesis using MDA-MB-231 cells and the murine mammary carcinoma: Ehrlich ascites tumor model. HSP90 was downregulated by TCCP by inactivation of HSF-1 resulting in inhibition of tumor cell proliferation, VEGF-induced cell migration, and concomitant decrease in tumor burden and neo-angiogenesis in vivo. The mechanism of suppression of HSPs involves inactivation of PI3K/Akt and phosphorylation on serine 307 of HSF-1 by the activation of ERK1. HSF-1 and HSP90 and 70 localization and expression were ascertained by immunolocalization, immunoblotting, and qPCR experiments. The anti-angiogenic effect of TCCP was studied in vivo in tumor-bearing mice and ex vivo using rat corneal micro-pocket assay. All the results thus corroborate the logic behind inactivating HSF-1 using TCCP as an alternative approach for cancer therapy.
Collapse
Affiliation(s)
- K C Rashmi
- Department of Studies in Biotechnology, University of Mysore, Mysuru, Karnataka, 570 006, India
| | - H S Atreya
- NMR Research Centre, Indian Institute of Science, Bengaluru, 560 012, India
| | - M Harsha Raj
- Department of Studies in Biotechnology, University of Mysore, Mysuru, Karnataka, 570 006, India
| | - Bharathi P Salimath
- Department of Studies in Biotechnology, University of Mysore, Mysuru, Karnataka, 570 006, India
| | - H S Aparna
- Department of Studies in Biotechnology, University of Mysore, Mysuru, Karnataka, 570 006, India.
| |
Collapse
|
15
|
Gomez-Pastor R, Burchfiel ET, Thiele DJ. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat Rev Mol Cell Biol 2017; 19:4-19. [PMID: 28852220 DOI: 10.1038/nrm.2017.73] [Citation(s) in RCA: 537] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The heat shock transcription factors (HSFs) were discovered over 30 years ago as direct transcriptional activators of genes regulated by thermal stress, encoding heat shock proteins. The accepted paradigm posited that HSFs exclusively activate the expression of protein chaperones in response to conditions that cause protein misfolding by recognizing a simple promoter binding site referred to as a heat shock element. However, we now realize that the mammalian family of HSFs comprises proteins that independently or in concert drive combinatorial gene regulation events that activate or repress transcription in different contexts. Advances in our understanding of HSF structure, post-translational modifications and the breadth of HSF-regulated target genes have revealed exciting new mechanisms that modulate HSFs and shed new light on their roles in physiology and pathology. For example, the ability of HSF1 to protect cells from proteotoxicity and cell death is impaired in neurodegenerative diseases but can be exploited by cancer cells to support their growth, survival and metastasis. These new insights into HSF structure, function and regulation should facilitate the development tof new disease therapeutics to manipulate this transcription factor family.
Collapse
Affiliation(s)
- Rocio Gomez-Pastor
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine
| | | | - Dennis J Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine.,Department of Biochemistry, Duke University School of Medicine.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
16
|
Schaefer S, Svenstrup TH, Guerra B. The small-molecule kinase inhibitor D11 counteracts 17-AAG-mediated up-regulation of HSP70 in brain cancer cells. PLoS One 2017; 12:e0177706. [PMID: 28542269 PMCID: PMC5436671 DOI: 10.1371/journal.pone.0177706] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/02/2017] [Indexed: 01/22/2023] Open
Abstract
Many types of cancer express high levels of heat shock proteins (HSPs) that are molecular chaperones regulating protein folding and stability ensuring protection of cells from potentially lethal stress. HSPs in cancer cells promote survival, growth and spreading even in situations of growth factors deprivation by associating with oncogenic proteins responsible for cell transformation. Hence, it is not surprising that the identification of potent inhibitors of HSPs, notably HSP90, has been the primary research focus, in recent years. Exposure of cancer cells to HSP90 inhibitors, including 17-AAG, has been shown to cause resistance to chemotherapeutic treatment mostly attributable to induction of the heat shock response and increased cellular levels of pro-survival chaperones. In this study, we show that treatment of glioblastoma cells with 17-AAG leads to HSP90 inhibition indicated by loss of stability of the EGFR client protein, and significant increase in HSP70 expression. Conversely, co-treatment with the small-molecule kinase inhibitor D11 leads to suppression of the heat shock response and inhibition of HSF1 transcriptional activity. Beside HSP70, Western blot and differential mRNA expression analysis reveal that combination treatment causes strong down-regulation of the small chaperone protein HSP27. Finally, we demonstrate that incubation of cells with both agents leads to enhanced cytotoxicity and significantly high levels of LC3-II suggesting autophagy induction. Taken together, results reported here support the notion that including D11 in future treatment regimens based on HSP90 inhibition can potentially overcome acquired resistance induced by the heat shock response in brain cancer cells.
Collapse
Affiliation(s)
- Susanne Schaefer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Tina H. Svenstrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Barbara Guerra
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- * E-mail:
| |
Collapse
|
17
|
Neudegger T, Verghese J, Hayer-Hartl M, Hartl FU, Bracher A. Structure of human heat-shock transcription factor 1 in complex with DNA. Nat Struct Mol Biol 2016; 23:140-6. [PMID: 26727489 DOI: 10.1038/nsmb.3149] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023]
Abstract
Heat-shock transcription factor 1 (HSF1) has a central role in mediating the protective response to protein conformational stresses in eukaryotes. HSF1 consists of an N-terminal DNA-binding domain (DBD), a coiled-coil oligomerization domain, a regulatory domain and a transactivation domain. Upon stress, HSF1 trimerizes via its coiled-coil domain and binds to the promoters of heat shock protein-encoding genes. Here, we present cocrystal structures of the human HSF1 DBD in complex with cognate DNA. A comparative analysis of the HSF1 paralog Skn7 from Chaetomium thermophilum showed that single amino acid changes in the DBD can switch DNA binding specificity, thus revealing the structural basis for the interaction of HSF1 with cognate DNA. We used a crystal structure of the coiled-coil domain of C. thermophilum Skn7 to develop a model of the active human HSF1 trimer in which HSF1 embraces the heat-shock-element DNA.
Collapse
Affiliation(s)
- Tobias Neudegger
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jacob Verghese
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Manajit Hayer-Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
18
|
The heat shock response restricts virus infection in Drosophila. Sci Rep 2015; 5:12758. [PMID: 26234525 PMCID: PMC4522674 DOI: 10.1038/srep12758] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/09/2015] [Indexed: 01/02/2023] Open
Abstract
Innate immunity is the first line of defence against pathogens and is essential for survival of the infected host. The fruit fly Drosophila melanogaster is an emerging model to study viral pathogenesis, yet antiviral defence responses remain poorly understood. Here, we describe the heat shock response, a cellular mechanism that prevents proteotoxicity, as a component of the antiviral immune response in Drosophila. Transcriptome analyses of Drosophila S2 cells and adult flies revealed strong induction of the heat shock response upon RNA virus infection. Dynamic induction patterns of heat shock pathway components were characterized in vitro and in vivo following infection with different classes of viruses. The heat shock transcription factor (Hsf), as well as active viral replication, were necessary for the induction of the response. Hsf-deficient adult flies were hypersensitive to virus infection, indicating a role of the heat shock response in antiviral defence. In accordance, transgenic activation of the heat shock response prolonged survival time after infection and enabled long-term control of virus replication to undetectable levels. Together, our results establish the heat shock response as an important constituent of innate antiviral immunity in Drosophila.
Collapse
|
19
|
Ortner V, Ludwig A, Riegel E, Dunzinger S, Czerny T. An artificial HSE promoter for efficient and selective detection of heat shock pathway activity. Cell Stress Chaperones 2015; 20:277-88. [PMID: 25168173 PMCID: PMC4326385 DOI: 10.1007/s12192-014-0540-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 08/14/2014] [Accepted: 08/15/2014] [Indexed: 11/26/2022] Open
Abstract
Detection of cellular stress is of major importance for the survival of cells. During evolution, a network of stress pathways developed, with the heat shock (HS) response playing a major role. The key transcription factor mediating HS signalling activity in mammalian cells is the HS factor HSF1. When activated it binds to the heat shock elements (HSE) in the promoters of target genes like heat shock protein (HSP) genes. They are induced by HSF1 but in addition they integrate multiple signals from different stress pathways. Here, we developed an artificial promoter consisting only of HSEs and therefore selectively reacting to HSF-mediated pathway activation. The promoter is highly inducible but has an extreme low basal level. Direct comparison with the HSPA1A promoter activity indicates that heat-dependent expression can be fully recapitulated by isolated HSEs in human cells. Using this sensitive reporter, we measured the HS response for different temperatures and exposure times. In particular, long heat induction times of 1 or 2 h were compared with short heat durations down to 1 min, conditions typical for burn injuries. We found similar responses to both long and short heat durations but at completely different temperatures. Exposure times of 2 h result in pathway activation at 41 to 44 °C, whereas heat pulses of 1 min lead to a maximum HS response between 47 and 50 °C. The results suggest that the HS response is initiated by a combination of temperature and exposure time but not by a certain threshold temperature.
Collapse
Affiliation(s)
- Viktoria Ortner
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | - Alfred Ludwig
- Department of Agrarian Production, Genetics and Microbiology Research Group Public, University of Navarre, Pamplona, Navarre Spain
| | - Elisabeth Riegel
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | - Sarah Dunzinger
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | - Thomas Czerny
- Department of Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| |
Collapse
|
20
|
Abstract
The heat shock response (HSR) is an ancient and highly conserved process that is essential for coping with environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review the phylogenetically conserved mechanisms that regulate fever and discuss the effects that febrile-range temperatures have on multiple biological processes involved in host defense and cell death and survival, including the HSR and its implications for patients with severe sepsis, trauma, and other acute systemic inflammatory states. Heat shock factor-1, a heat-induced transcriptional enhancer is not only the central regulator of the HSR but also regulates expression of pivotal cytokines and early response genes. Febrile-range temperatures exert additional immunomodulatory effects by activating mitogen-activated protein kinase cascades and accelerating apoptosis in some cell types. This results in accelerated pathogen clearance, but increased collateral tissue injury, thus the net effect of exposure to febrile range temperature depends in part on the site and nature of the pathologic process and the specific treatment provided.
Collapse
Affiliation(s)
- Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore V.A. Medical Center, Baltimore, Maryland
| | | | | |
Collapse
|
21
|
Raychaudhuri S, Loew C, Körner R, Pinkert S, Theis M, Hayer-Hartl M, Buchholz F, Hartl F. Interplay of Acetyltransferase EP300 and the Proteasome System in Regulating Heat Shock Transcription Factor 1. Cell 2014; 156:975-85. [DOI: 10.1016/j.cell.2014.01.055] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/20/2013] [Accepted: 01/23/2014] [Indexed: 12/22/2022]
|
22
|
Morton EA, Lamitina T. Caenorhabditis elegans HSF-1 is an essential nuclear protein that forms stress granule-like structures following heat shock. Aging Cell 2013; 12:112-20. [PMID: 23107491 DOI: 10.1111/acel.12024] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2012] [Indexed: 01/06/2023] Open
Abstract
The heat shock transcription factor (HSF) is a conserved regulator of heat shock-inducible gene expression. Organismal roles for HSF in physiological processes such as development, aging, and immunity have been defined largely through studies of the single Caenorhabditis elegans HSF homolog, hsf-1. However, the molecular and cell biological properties of hsf-1 in C. elegans are incompletely understood. We generated animals expressing physiological levels of an HSF-1::GFP fusion protein and examined its function, localization, and regulation in vivo. HSF-1::GFP was functional, as measured by its ability to rescue phenotypes associated with two hsf-1 mutant alleles. Rescue of hsf-1 development phenotypes was abolished in a DNA-binding-deficient mutant, demonstrating that the transcriptional targets of hsf-1 are critical to its function even in the absence of stress. Under nonstress conditions, HSF-1::GFP was found primarily in the nucleus. Following heat shock, HSF-1::GFP rapidly and reversibly redistributed into dynamic, subnuclear structures that share many properties with human nuclear stress granules, including colocalization with markers of active transcription. Rapid formation of HSF-1 stress granules required HSF-1 DNA-binding activity, and the threshold for stress granule formation was altered by growth temperature. HSF-1 stress granule formation was not induced by inhibition of IGF signaling, a pathway previously suggested to function upstream of hsf-1. Our findings suggest that development, stress, and aging pathways may regulate HSF-1 function in distinct ways, and that HSF-1 nuclear stress granule formation is an evolutionarily conserved aspect of HSF-1 regulation in vivo.
Collapse
Affiliation(s)
- Elizabeth A Morton
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
23
|
Chiang WC, Ching TT, Lee HC, Mousigian C, Hsu AL. HSF-1 regulators DDL-1/2 link insulin-like signaling to heat-shock responses and modulation of longevity. Cell 2012; 148:322-34. [PMID: 22265419 DOI: 10.1016/j.cell.2011.12.019] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 07/13/2011] [Accepted: 12/21/2011] [Indexed: 11/16/2022]
Abstract
Extended longevity is often correlated with increased resistance against various stressors. Insulin/IGF-1-like signaling (IIS) is known to have a conserved role in aging and cellular mechanisms against stress. In C. elegans, genetic studies suggest that heat-shock transcription factor HSF-1 is required for IIS to modulate longevity. Here, we report that the activity of HSF-1 is regulated by IIS. This regulation occurs at an early step of HSF-1 activation via two HSF-1 regulators, DDL-1 and DDL-2. Inhibition of DDL-1/2 increases longevity and thermotolerance in an hsf-1-dependent manner. Furthermore, biochemical analyses suggest that DDL-1/2 negatively regulate HSF-1 activity by forming a protein complex with HSF-1. The formation of this complex (DHIC) is affected by the phosphorylation status of DDL-1. Both the formation of DHIC and the phosphorylation of DDL-1 are controlled by IIS. Our findings point to DDL-1/2 as a link between IIS and the HSF-1 pathway.
Collapse
Affiliation(s)
- Wei-Chung Chiang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
24
|
Heilman JM, Burke TJ, McClain CJ, Watson WH. Transactivation of gene expression by NF-κB is dependent on thioredoxin reductase activity. Free Radic Biol Med 2011; 51:1533-42. [PMID: 21782934 PMCID: PMC3755477 DOI: 10.1016/j.freeradbiomed.2011.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 06/23/2011] [Accepted: 06/27/2011] [Indexed: 01/19/2023]
Abstract
The redox-sensitive transcription factor NF-κB mediates the expression of genes involved in inflammation and cell survival. Thioredoxin reductase-1 (TR1) and its substrate thioredoxin-1 act together to reduce oxidized cysteine residues within the DNA-binding domain of NF-κB and promote maximal DNA-binding activity in vitro. It is not clear, however, if NF-κB is regulated via this mechanism within living cells. The purpose of this study was to determine the mechanism of NF-κB modulation by TR1 in cells stimulated with the inflammatory cytokine tumor necrosis factor-α (TNF). In both control cells and cells depleted of TR1 activity through chemical inhibition or siRNA knockdown, TNF stimulation resulted in degradation of the cytoplasmic NF-κB inhibitor IκB-α and translocation of NF-κB to the nucleus. Similarly, the DNA-binding activity and redox state of NF-κB were unaffected by TR1 depletion. In contrast, NF-κB-mediated gene expression was markedly inhibited in cells lacking TR1 activity, suggesting that the transactivation potential of NF-κB is sensitive to changes in TR1 activity. Consistent with this concept, phosphorylation of the transactivation domain of NF-κB was inhibited in the presence of curcumin. Surprisingly, another TR1 inhibitor, 1-chloro-2,4-dinitrobenzene, had no effect, and siRNA knockdown of TR1 actually increased phosphorylation at this site. These results demonstrate that TR1 activity controls the transactivation potential of NF-κB and that more than one mechanism may mediate this effect.
Collapse
Affiliation(s)
- Jacqueline M. Heilman
- Division of Toxicology, Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Tom J. Burke
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40292
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40292
| | - Walter H. Watson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40292
| |
Collapse
|
25
|
Anckar J, Sistonen L. Regulation of HSF1 function in the heat stress response: implications in aging and disease. Annu Rev Biochem 2011; 80:1089-115. [PMID: 21417720 DOI: 10.1146/annurev-biochem-060809-095203] [Citation(s) in RCA: 558] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To dampen proteotoxic stresses and maintain protein homeostasis, organisms possess a stress-responsive molecular machinery that detects and neutralizes protein damage. A prominent feature of stressed cells is the increased synthesis of heat shock proteins (Hsps) that aid in the refolding of misfolded peptides and restrain protein aggregation. Transcriptional activation of the heat shock response is orchestrated by heat shock factor 1 (HSF1), which rapidly translocates to hsp genes and induces their expression. Although the role of HSF1 in protecting cells and organisms against severe stress insults is well established, many aspects of how HSF1 senses qualitatively and quantitatively different forms of stresses have remained poorly understood. Moreover, recent discoveries that HSF1 controls life span have prompted new ways of thinking about an old transcription factor. Here, we review the established role of HSF1 in counteracting cell stress and prospect the role of HSF1 as a regulator of disease states and aging.
Collapse
Affiliation(s)
- Julius Anckar
- Department of Biosciences, Åbo Akademi University, BioCity, 20520 Turku, Finland.
| | | |
Collapse
|
26
|
Satoh T, Rezaie T, Seki M, Sunico CR, Tabuchi T, Kitagawa T, Yanagitai M, Senzaki M, Kosegawa C, Taira H, McKercher SR, Hoffman JK, Roth GP, Lipton SA. Dual neuroprotective pathways of a pro-electrophilic compound via HSF-1-activated heat-shock proteins and Nrf2-activated phase 2 antioxidant response enzymes. J Neurochem 2011; 119:569-78. [PMID: 21883218 DOI: 10.1111/j.1471-4159.2011.07449.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activation of the Keap1/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and consequent induction of phase 2 antioxidant enzymes is known to afford neuroprotection. Here, we present a series of novel electrophilic compounds that protect neurons via this pathway. Natural products, such as carnosic acid (CA), are present in high amounts in the herbs rosemary and sage as ortho-dihydroquinones, and have attracted particular attention because they are converted by oxidative stress to their active form (ortho-quinone species) that stimulate the Keap1/Nrf2 transcriptional pathway. Once activated, this pathway leads to the production of a series of antioxidant phase 2 enzymes. Thus, such dihydroquinones function as redox-activated 'pro-electrophiles'. Here, we explored the concept that related para-dihydroquinones represent even more effective bioactive pro-electrophiles for the induction of phase 2 enzymes without producing toxic side effects. We synthesized several novel para-hydroquinone-type pro-electrophilic compounds (designated D1 and D2) to analyze their protective mechanism. DNA microarray, PCR, and western blot analyses showed that compound D1 induced expression of heat-shock proteins (HSPs), including HSP70, HSP27, and DnaJ, in addition to phase 2 enzymes such as hemeoxygenase-1 (HO-1), NADP(H) quinine-oxidoreductase1, and the Na(+)-independent cystine/glutamate exchanger (xCT). Treatment with D1 resulted in activation of Nrf2 and heat-shock transcription factor-1 (HSF-1) transcriptional elements, thus inducing phase 2 enzymes and HSPs, respectively. In this manner, D1 protected neuronal cells from both oxidative and endoplasmic reticulum (ER)-related stress. Additionally, D1 suppressed induction of 78 kDa glucose-regulated protein (GRP78), an ER chaperone protein, and inhibited hyperoxidation of peroxiredoxin 2 (PRX2), a molecule that is in its reduced state can protect from oxidative stress. These results suggest that D1 is a novel pro-electrophilic compound that activates both the Nrf2 and HSF-1 pathways, and may thus offer protection from oxidative and ER stress.
Collapse
Affiliation(s)
- Takumi Satoh
- Department of Welfare Engineering, Faculty of Engineering, Iwate University, Morioka, Iwate, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kremer SB, Gross DS. SAGA and Rpd3 chromatin modification complexes dynamically regulate heat shock gene structure and expression. J Biol Chem 2009; 284:32914-31. [PMID: 19759026 DOI: 10.1074/jbc.m109.058610] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The chromatin structure of heat shock protein (HSP)-encoding genes undergoes dramatic alterations upon transcriptional induction, including, in extreme cases, domain-wide nucleosome disassembly. Here, we use a combination of gene knock-out, in situ mutagenesis, chromatin immunoprecipitation, and expression assays to investigate the role of histone modification complexes in regulating heat shock gene structure and expression in Saccharomyces cerevisiae. Two histone acetyltransferases, Gcn5 and Esa1, were found to stimulate HSP gene transcription. A detailed chromatin immunoprecipitation analysis of the Gcn5-containing SAGA complex (signified by Spt3) revealed its presence within the promoter of every heat shock factor 1-regulated gene examined. The occupancy of SAGA increased substantially upon heat shock, peaking at several HSP promoters within 30-45 s of temperature upshift. SAGA was also efficiently recruited to the coding regions of certain HSP genes (where its presence mirrored that of pol II), although not at others. Robust and rapid recruitment of repressive, Rpd3-containing histone deacetylase complexes was also seen and at all HSP genes examined. A detailed analysis of HSP82 revealed that both Rpd3(L) and Rpd3(S) complexes (signified by Sap30 and Rco1, respectively) were recruited to the gene promoter, yet only Rpd3(S) was recruited to its open reading frame. A consensus URS1 cis-element facilitated the recruitment of each Rpd3 complex to the HSP82 promoter, and this correlated with targeted deacetylation of promoter nucleosomes. Collectively, our observations reveal that SAGA and Rpd3 complexes are rapidly and synchronously recruited to heat shock factor 1-activated genes and suggest that their opposing activities modulate heat shock gene chromatin structure and fine-tune transcriptional output.
Collapse
Affiliation(s)
- Selena B Kremer
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | |
Collapse
|
28
|
Demarco VG, Scumpia PO, Bosanquet JP, Skimming JW. α-Lipoic Acid Inhibits Endotoxin-stimulated Expression of iNOS and Nitric Oxide Independent of the Heat Shock Response in RAW 264.7 Cells. Free Radic Res 2009; 38:675-82. [PMID: 15453632 DOI: 10.1080/10715760410001702503] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The heat shock response protects against sepsis-induced mortality, organ injury, cardiovascular dysfunction, and apoptosis. Several inducers of the heat shock response, such as hyperthermia, sodium arsenite, and pyrollidine dithiocarbonate, inhibit NF-kappaB activation and nitric oxide formation. The antioxidant lipoic acid (LA) has recently been found to inhibit NF-kappaB activation and nitric oxide formation. We therefore tested the hypothesis that LA induces a heat shock response. To test this hypothesis, we determined whether exposure to LA affects expression of both heat shock protein 70 (HSP-70) and nuclear heat shock factor-1 (HSF-1) in lipopolysaccharide (LPS) stimulated macrophages. LA and hyperthermia attenuated LPS-induced increases in nuclear NF-kappaB, iNOS protein, and media nitrite concentrations. LPS and hyperthermia increased HSP-70 concentrations 8-fold and 20-fold, respectively. No effect of LA treatment alone on HSP-70 protein expression was detected. Likewise, no effect of LA on HSF-1 protein expression was detected. These data suggest that LA inhibits LPS-induced activation of iNOS in macrophages independent of the heat shock response.
Collapse
Affiliation(s)
- Vincent G Demarco
- Department of Child Health, University of Missouri, Columbia, MO 65211, USA
| | | | | | | |
Collapse
|
29
|
Singh IS, Shah NG, Almutairy E, Hasday JD. Role of HSF1 in Infectious Disease. HEAT SHOCK PROTEINS 2009. [DOI: 10.1007/978-90-481-2976-8_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Qiao JY, Shao W, Wei HJ, Sun YM, Zhao YC, Xing WL, Zhang L, Mitchelson K, Cheng J. Novel high-throughput profiling of human transcription factors and its use for systematic pathway mapping. J Proteome Res 2008; 7:2769-79. [PMID: 18537283 DOI: 10.1021/pr700883t] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Transcription factors (TFs) are crucial components of regulatory networks that control gene transcription. Current TF assays are limited to the analysis of a single TF or require TF-specific antibodies. Here we report the Single Primer Amplification assisted Oligonucleotide Array-based Transcription Factor Assay (SPA-OATFA) which can directly analyze the binding activities of 240 human TFs simultaneously. Examining early events during serum-stimulation of HeLa cells as a model, we demonstrated the utility of SPA-OATFA combined with whole genome gene expression to systematically map the temporal activation of signaling pathways. Both TFs known to function in this stimulation response such as EGR1 and AP1 and new TFs such as HSF1 were identified. This information, combined with mRNA profiling, provided novel insights into the activities of regulatory pathways, and illustrates the potential of SPA-OATFA in detailed systems biology analysis of cell responses.
Collapse
Affiliation(s)
- Ji-Ying Qiao
- Medical Systems Biology Research Center, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Anckar J, Sistonen L. Heat Shock Factor 1 as a Coordinator of Stress and Developmental Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 594:78-88. [PMID: 17205677 DOI: 10.1007/978-0-387-39975-1_8] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The transition from normal growth conditions to stressful conditions is accompanied by a robust upregulation of heat shock proteins, which dampen the cytotoxicity caused by misfolded and denatured proteins. The most prominent part of this transition occurs on the transcriptional level. In mammals, protein-damaging stress leads to the activation of heat shock factor 1 (HSF1), which binds to upstream regulatory sequences in the promoters of heat shock genes. The activation of HSF1 proceeds through a multi-step pathway, involving a monomer-to-trimer transition, nuclear accumulation and extensive posttranslational modifications. In addition to its established role as the main regulator of heat shock genes, new data link HSF 1 to developmental pathways. In this chapter, we examine the established stress-related functions and prospect the intriguing role of HSF 1 as a developmental coordinator.
Collapse
Affiliation(s)
- Julius Anckar
- Department of Biology, Abo Akademi University, P.O. Box 123 FI-20521 Turku, Finland
| | | |
Collapse
|
32
|
Kowalczyk A, Guzik K, Slezak K, Dziedzic J, Rokita H. Heat shock protein and heat shock factor 1 expression and localization in vaccinia virus infected human monocyte derived macrophages. JOURNAL OF INFLAMMATION-LONDON 2005; 2:12. [PMID: 16246258 PMCID: PMC1283150 DOI: 10.1186/1476-9255-2-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Accepted: 10/24/2005] [Indexed: 12/25/2022]
Abstract
BACKGROUND Viruses remain one of the inducers of the stress response in the infected cells. Heat shock response induced by vaccinia virus (VV) infection was studied in vitro in human blood monocyte derived macrophages (MDMs) as blood cells usually constitute the primary site of the infection. METHODS Human blood monocytes were cultured for 12-14 days. The transcripts of heat shock factor 1 (HSF1), heat shock protein 70 (HSP70), heat shock protein 90 (HSP90) and two viral genes (E3L and F17R) were assayed by reverse transcriptase-polymerase chain reaction (RT-PCR), and the corresponding proteins measured by Western blot. Heat shock factor 1 DNA binding activities were estimated by electrophoretic mobility shift assay (EMSA) and its subcellular localization analyzed by immunocytofluorescence. RESULTS It appeared that infection with vaccinia virus leads to activation of the heat shock factor 1. Activation of HSF1 causes increased synthesis of an inducible form of the HSP70 both at the mRNA and the protein level. Although HSP90 mRNA was enhanced in vaccinia virus infected cells, the HSP90 protein content remained unchanged. At the time of maximum vaccinia virus gene expression, an inhibitory effect of the infection on the heat shock protein and the heat shock factor 1 was most pronounced. Moreover, at the early phase of the infection translocation of HSP70 and HSP90 from the cytoplasm to the nucleus of the infected cells was observed. CONCLUSION Preferential nuclear accumulation of HSP70, the major stress-inducible chaperone protein, suggests that VV employs this particular mechanism of cytoprotection to protect the infected cell rather than to help viral replication. The results taken together with our previous data on monocytes or MDMs infected with VV or S. aureus strongly argue that VV employs multiple cellular antiapoptotic/cytoprotective mechanisms to prolong viability and proinflammatory activity of the cells of monocytic-macrophage lineage.
Collapse
Affiliation(s)
- Aleksandra Kowalczyk
- Jagiellonian University, Faculty of Biotechnology; 7, Gronostajowa St., 30-387 Krakow, Poland
| | - Krzysztof Guzik
- Jagiellonian University, Faculty of Biotechnology; 7, Gronostajowa St., 30-387 Krakow, Poland
| | - Kinga Slezak
- Jagiellonian University, Faculty of Biotechnology; 7, Gronostajowa St., 30-387 Krakow, Poland
| | - Jakub Dziedzic
- Jagiellonian University, Faculty of Biotechnology; 7, Gronostajowa St., 30-387 Krakow, Poland
| | - Hanna Rokita
- Jagiellonian University, Faculty of Biotechnology; 7, Gronostajowa St., 30-387 Krakow, Poland
| |
Collapse
|
33
|
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
|
34
|
Hsiao CJJ, Stapleton SR. Characterization of Cd-induced molecular events prior to cellular damage in primary rat hepatocytes in culture: activation of the stress activated signal protein JNK and transcription factor AP-1. J Biochem Mol Toxicol 2005; 18:133-42. [PMID: 15252869 DOI: 10.1002/jbt.20018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The effect of Cadmium (Cd) on the expression of c-Jun N-terminal kinase (JNK), c-jun, and activator protein-1 (AP-1) has been investigated. We previously reported that Cd causes cell damage as indicated by increases in the cytotoxic parameters, lactate dehydrogenase and lipid peroxidation, and this damage was mediated by decreases in cellular concentration of glutathione. In the present study, we investigate the molecular events involved prior to the Cd-induced cellular toxicity and damage in primary rat hepatocytes. We propose that Cd, through the generation of reactive oxygen species (ROS) and prior to significant cellular damage, activates the stress activated signal protein JNK, regulates c-jun expression, and promotes the binding of a redox sensitive transcription factor AP-1. We show JNK activity and c-jun mRNA level significantly increased at 1 h and AP-1 DNA binding activity significantly enhanced at 3 h in the presence of 4 microM cadmium chloride. Blocking the Cd induction of JNK activity, c-jun mRNA level, and AP-1 binding activity using the antioxidants N-acetyl cysteine (10 mM) or carnosol (0.5 microg/mL) suggests a role for ROS. Blocking JNK activity and c-jun mRNA by SP600125 (20 microM), a JNK inhibitor, supports the role of JNK in transmission of signals induced by Cd.
Collapse
Affiliation(s)
- Chin-ju J Hsiao
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | | |
Collapse
|
35
|
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
|
36
|
Wang X, Grammatikakis N, Siganou A, Calderwood SK. Regulation of molecular chaperone gene transcription involves the serine phosphorylation, 14-3-3 epsilon binding, and cytoplasmic sequestration of heat shock factor 1. Mol Cell Biol 2003; 23:6013-26. [PMID: 12917326 PMCID: PMC180972 DOI: 10.1128/mcb.23.17.6013-6026.2003] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2002] [Revised: 02/04/2003] [Accepted: 05/13/2003] [Indexed: 11/20/2022] Open
Abstract
Heat shock factor 1 (HSF1) regulates the transcription of molecular chaperone hsp genes. However, the cellular control mechanisms that regulate HSF1 activity are not well understood. In this study, we have demonstrated for the first time that human HSF1 binds to the essential cell signaling protein 14-3-3 epsilon. Binding of HSF1 to 14-3-3 epsilon occurs in cells in which extracellular signal regulated kinase (ERK) is activated and blockade of the ERK pathway by treatment with the specific ERK pathway inhibitor PD98059 in vivo strongly suppresses the binding. We previously showed that ERK1 phosphorylates HSF1 on serine 307 and leads to secondary phosphorylation by glycogen synthase kinase 3 (GSK3) on serine 303 within the regulatory domain and that these phosphorylation events repress HSF1. We show here that HSF1 binding to 14-3-3 epsilon requires HSF1 phosphorylation on serines 303 and 307. Furthermore, the serine phosphorylation-dependent binding of HSF1 to 14-3-3 epsilon results in the transcriptional repression of HSF1 and its sequestration in the cytoplasm. Leptomycin B, a specific inhibitor of nuclear export receptor CRM1, was found to reverse the cytoplasmic sequestration of HSF1 mediated by 14-3-3 epsilon, suggesting that CRM1/14-3-3 epsilon directed nuclear export plays a major role in repression of HSF1 by the ERK/GSK3/14-3-3 epsilon pathway. Our experiments indicate a novel pathway for HSF1 regulation and suggest a mechanism for suppression of its activity during cellular proliferation.
Collapse
Affiliation(s)
- XiaoZhe Wang
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
37
|
Hung WJ, Roberson RS, Taft J, Wu DY. Human BAG-1 proteins bind to the cellular stress response protein GADD34 and interfere with GADD34 functions. Mol Cell Biol 2003; 23:3477-86. [PMID: 12724406 PMCID: PMC164759 DOI: 10.1128/mcb.23.10.3477-3486.2003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cellular stress response protein GADD34 mediates growth arrest and apoptosis in response to DNA damage, negative growth signals, and protein malfolding. GADD34 binds to protein phosphatase PP1 and can attenuate the translational elongation of key transcriptional factors through dephosphorylation of eukaryotic initiation factor 2alpha (eIF2alpha). Recently, we reported the involvement of human SNF5/INI1 (hSNF5/INI1) protein in the functions of GADD34 and showed that hSNF5/INI1 binds GADD34 and stimulates the bound PP1 phosphatase activity. To better understand the regulatory and functional mechanisms of GADD34, we undertook a yeast two-hybrid screen with full-length GADD34 as bait in order to identify additional protein partners of GADD34. We report here that human cochaperone protein BAG-1 interacts with GADD34 in vitro and in SW480 cells treated with the proteasome inhibitor z-LLL-B to induce apoptosis. Two other proteins, Hsp70/Hsc70 and PP1, associate reversibly with the GADD34-BAG-1 complex, and their dissociation is promoted by ATP. BAG-1 negatively modulates GADD34-bound PP1 activity, and the expression of BAG-1 isoforms can also mask GADD34-mediated inhibition of colony formation and suppression of transcription. Our findings suggest that BAG-1 may function to suppress the GADD34-mediated cellular stress response and support a role for BAG-1 in the survival of cells undergoing stress.
Collapse
Affiliation(s)
- Wesley J Hung
- Division of Medical Oncology, Department of Medicine, Veterans Administration Puget Sound Health Care System, Seattle Division, 1660 S. Columbian Way, Seattle, WA 98108, USA
| | | | | | | |
Collapse
|
38
|
Ahn SG, Thiele DJ. Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress. Genes Dev 2003; 17:516-28. [PMID: 12600944 PMCID: PMC195992 DOI: 10.1101/gad.1044503] [Citation(s) in RCA: 327] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The activation of eukaryotic heat shock protein (Hsp) gene expression occurs in response to a wide variety of cellular stresses including heat shock, hydrogen peroxide, uncoupled oxidative phosphorylation, infection, and inflammation. Biochemical and genetic studies have clearly demonstrated critical roles for mammalian heat shock factor 1 (HSF1) in stress-inducible Hsp gene expression, resistance to stress-induced programmed cell death, extra-embryonic development, and other biological functions. Activation of mammalian Hsp gene expression involves the stress-inducible conversion of HSF1 from the inactive monomer to the DNA-binding competent homotrimer. Although Hsp activation is a central conserved process in biology, the precise mechanisms for stress sensing and signaling to activate HSF1, and the mechanisms by which many distinct stresses activate HSF1, are poorly understood. In this report we demonstrate that recombinant mammalian HSF1 directly senses both heat and hydrogen peroxide to assemble into a homotrimer in a reversible and redox-regulated manner. The sensing of both stresses requires two cysteine residues within the HSF1 DNA-binding domain that are engaged in redox-sensitive disulfide bonds. HSF1 derivatives in which either or both cysteines were mutated are defective in stress-inducible trimerization and DNA binding, stress-inducible nuclear translocation and Hsp gene trans-activation, and in the protection of mouse cells from stress-induced apoptosis. This redox-dependent activation of HSF1 by heat and hydrogen peroxide establishes a common mechanism in the stress activation of Hsp gene expression by mammalian HSF1.
Collapse
Affiliation(s)
- Sang-Gun Ahn
- Department of Biological Chemistry, The University of Michigan Medical School, Ann Arbor, Michigan 48109-0606, USA
| | | |
Collapse
|
39
|
Fairfield DA, Kanicki AC, Lomax MI, Altschuler RA. Expression and localization of heat shock factor (Hsf) 1 in the rodent cochlea. Hear Res 2002; 173:109-18. [PMID: 12372640 DOI: 10.1016/s0378-5955(02)00607-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of heat shock factors (Hsfs) is one of the potential mechanisms for regulating the transcription of the heat shock proteins (Hsps) and certain other stress-responsive genes. Reverse transcription polymerase chain reaction (RT-PCR), Western blot and immunocytochemistry were used to examine the expression and localization of Hsf1, the stress-responsive member of the Hsf family, in the rat and mouse cochlea. Cerebellum was used as a positive control. Semi-quantitative RT-PCR of cochlear RNA revealed that Hsf1 was more highly expressed in a subfraction containing sensorineural epithelium and lateral wall than in a subfraction containing modiolus, with the alpha splice form predominant over the beta in both subfractions. Immunocytochemistry showed selective staining in the rodent cochlea. Hsf1 immunostaining was found in the nuclei of inner and outer hair cells in the organ of Corti, spiral ganglion cells in the modiolus, and cells in the marginal and intermediate layers of the stria vascularis. This is largely consistent with where Hsp70 induction is reported. Hsf1 activation following heat shock was examined by Western blot. Hyperthermia resulted in stress-induced Hsf1 hyperphosphorylation in cochlea as well as cerebellum. This hyperphosphorylation as well as the correlation of its localization with Hsp70 induction supports a role for Hsf1 in the cochlear stress response.
Collapse
Affiliation(s)
- Damon A Fairfield
- Kresge Hearing Research Institute, Department of Otolaryngology/Head Neck Surgery, The University of Michigan, 1301 East Ann Street, Ann Arbor 48109-0506, USA
| | | | | | | |
Collapse
|
40
|
Grably MR, Stanhill A, Tell O, Engelberg D. HSF and Msn2/4p can exclusively or cooperatively activate the yeast HSP104 gene. Mol Microbiol 2002; 44:21-35. [PMID: 11967066 DOI: 10.1046/j.1365-2958.2002.02860.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In an effort to understand how an accurate level of stress-specific expression is obtained, we studied the promoter of the yeast HSP104 gene. Through 5' deletions, we defined a 334 bp fragment upstream of the first coding AUG as sufficient and essential for maximal basal activity and a 260 bp fragment as sufficient and essential for heat shock responsiveness. These sequences contain heat shock elements (HSEs) and stress response elements (STREs) that cooperate to achieve maximal inducible expression. However, in the absence of one set of factors (e.g. in msn2Deltamsn4Delta cells) proper induction is obtained exclusively through HSEs. We also show that HSP104 is constitutively derepressed in ras2Delta cells. This derepression is achieved exclusively through activation of STREs, with no role for HSEs. Strikingly, in ras2Deltamsn2Deltamsn4Delta cells the HSP104 promoter is also derepressed, but in this strain derepression is mediated through HSEs, showing the flexibility and adaptation of the promoter. Thus, appropriate transcription of HSP104 is usually obtained through cooperation between the Msn2/4/STRE and the HSF/ HSE systems, but each factor could activate the promoter alone, backing up the other. Transcription control of HSP104 is adaptive and robust, ensuring proper expression under extreme conditions and in various mutants.
Collapse
Affiliation(s)
- Melanie R Grably
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | | | |
Collapse
|
41
|
Tomanek L, Somero GN. Interspecific- and acclimation-induced variation in levels of heat-shock proteins 70 (hsp70) and 90 (hsp90) and heat-shock transcription factor-1 (HSF1) in congeneric marine snails (genusTegula): implications for regulation ofhspgene expression. J Exp Biol 2002; 205:677-85. [PMID: 11907057 DOI: 10.1242/jeb.205.5.677] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
SUMMARYIn our previous studies of heat-shock protein (hsp) expression in congeneric marine gastropods of the genus Tegula, we observed interspecific and acclimation-induced variation in the temperatures at which heat-shock gene expression is induced (Ton). To investigate the factors responsible for these inter- and intraspecific differences in Ton, we tested the predictions of the ‘cellular thermometer’ model for the transcriptional regulation of hsp expression. According to this model, hsps not active in chaperoning unfolded proteins bind to a transcription factor, heat-shock factor-1 (HSF1), thereby reducing the levels of free HSF1 that are available to bind to the heat-shock element, a regulatory element upstream of hsp genes. Under stress, hsps bind to denatured proteins, releasing HSF1, which can now activate hsp gene transcription. Thus, elevated levels of heat-shock proteins of the 40, 70 and 90 kDa families (hsp 40, hsp70 and hsp90, respectively) would be predicted to elevate Ton. Conversely, elevated levels of HSF1 would be predicted to decrease Ton. Following laboratory acclimation to 13, 18 and 23°C, we used solid-phase immunochemistry (western analysis) to quantify endogenous levels of two hsp70 isoforms (hsp74 and hsp72), hsp90 and HSF1 in the low- to mid-intertidal species Tegula funebralis and in two subtidal to low-intertidal congeners, T. brunnea and T. montereyi. We found higher endogenous levels of hsp72 (a strongly heat-induced isoform) at 13 and 18°C in T. funebralis in comparison with T. brunnea and T. montereyi. However, T. funebralis also had higher levels of HSF1 than its congeners. The higher levels of HSF1 in T. funebralis cannot, within the framework of the cellular thermometer model, account for the higher Ton observed for this species, although they may explain why T. funebralis is able to induce the heat-shock response more rapidly than T. brunnea. However, the cellular thermometer model does appear to explain the cause of the increases in Ton that occurred during warm acclimation of the two subtidal species, in which warm acclimation was accompanied by increased levels of hsp72, hsp74 and hsp90, whereas levels of HSF1 remained stable. T. funebralis, which experiences greater heat stress than its subtidal congeners, consistently had higher ratios of hsp72 to hsp74 than its congeners, although the sum of levels of the two isoforms was similar for all three species except at the highest acclimation temperature (23°C). The ratio of hsp72 to hsp74 may provide a more accurate estimate of environmental heat stress than the total concentrations of both hsp70 isoforms.
Collapse
Affiliation(s)
- Lars Tomanek
- Hopkins Marine Station, Stanford University, Pacific Grove, CA 93950-3094, USA.
| | | |
Collapse
|
42
|
Manalo DJ, Liu AY. Resolution, detection, and characterization of redox conformers of human HSF1. J Biol Chem 2001; 276:23554-61. [PMID: 11320084 DOI: 10.1074/jbc.m011300200] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here an experimental protocol for the resolution, detection, and quantitation of the reduced and oxidized conformers of human heat shock factor 1 (hHSF1) and report on the effects in vitro and in vivo of redox-active agents on the redox status, structure, and function of hHSF1. We showed that diamide, a reagent that promotes disulfide bond formation, caused a loss of immunorecognition of the monomeric hHSF1 protein in a standard Western blot detection procedure. Modification of the Western blot procedure to include dithiothreitol in the equilibration and transfer buffers after gel electrophoresis allowed for the detection of a compact, intramolecularly disulfide cross-linked oxidized hHSF1 (ox-hHSF1) in the diamide-treated sample. The effect of diamide was blocked by pretreatment with N-ethylmaleimide and was reversed by dithiothreitol added to the sample prior to gel electrophoresis. Incubation with nitrosoglutathione at 42 degrees C also promoted the conversion of HSF1 to ox-HSF1; at 25 degrees C, however, nitrosoglutathione was by itself without effect but blocked the formation of ox-hHSF1 in the presence of diamide. The disulfide cross-linked ox-hHSF1 was monomeric and resistant to the in vitro heat-induced trimerization and activation. The possibility that ox-HSF1 may occur in oxidatively stressed cells was evaluated. Treatment of HeLa cells with 2 mm l-buthionine sulfoximine promoted the formation of ox-HSF1 and blocked the heat-induced activation of HSF DNA binding activity. Our result suggests that hHSF1 may have integrated redox chemistry of cysteine sulfhydryl into its functional responses.
Collapse
Affiliation(s)
- D J Manalo
- Graduate Program in Cell and Developmental Biology and Department of Cell Biology and Neuroscience, Rutgers State University, Piscataway, New Jersey 08854-8082, USA
| | | |
Collapse
|
43
|
Holmberg CI, Illman SA, Kallio M, Mikhailov A, Sistonen L. Formation of nuclear HSF1 granules varies depending on stress stimuli. Cell Stress Chaperones 2000; 5:219-28. [PMID: 11005380 PMCID: PMC312888 DOI: 10.1379/1466-1268(2000)005<0219:fonhgv>2.0.co;2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In concert with the stress-induced activation of human heat shock factor 1 (HSF1), the factor becomes inducibly phosphorylated and accumulates into nuclear granules. To date, these processes are not fully understood. Here, we show that although stress caused by the proteasome inhibitors MG132 and clasto-lactacystine beta-lactone induces the expression of Hsp70, the formation of HSF1 granules is affected differently in comparison to heat shock. Furthermore, proteasome inhibition increases serine phosphorylation on HSF1, but to a lesser extent than heat stress. Our results suggest that, depending on the type of stress stimulus, the multiple events associated with HSF1 activation might be affected differently.
Collapse
Affiliation(s)
- Carina I Holmberg
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, PO Box 123, FIN-20521 Turku, Finland and Department of Biochemistry and Pharmacy, Åbo Akademi University, PO Box 66, FIN-20521 Turku, Finland
| | - Sara A Illman
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, PO Box 123, FIN-20521 Turku, Finland and Department of Biochemistry and Pharmacy, Åbo Akademi University, PO Box 66, FIN-20521 Turku, Finland
| | - Marko Kallio
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, PO Box 123, FIN-20521 Turku, Finland
| | - Andrey Mikhailov
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, PO Box 123, FIN-20521 Turku, Finland
| | - Lea Sistonen
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, PO Box 123, FIN-20521 Turku, Finland
- Correspondence to: Lea Sistonen, Tel: +358 2 333 8028; Fax: +358 2 333 8000; .
| |
Collapse
|