1
|
Zhang H, Wang X. The Role of Protein Quantity Control in Polyglutamine Spinocerebellar Ataxias. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2575-2592. [PMID: 39052145 DOI: 10.1007/s12311-024-01722-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Polyglutamine spinocerebellar ataxias (polyQ SCAs) represent the most prevalent subtype of SCAs. The primary pathogenic mechanism is believed to be the gain-of-function neurotoxicity of polyQ proteins. Strategies such as enhancing the degradation or inhibiting the accumulation of these mutant proteins are pivotal for reducing their toxicity and slowing disease progression. The protein quality control (PQC) system, comprising primarily molecular chaperones and the ubiquitin‒proteasome system (UPS), is essential for maintaining protein homeostasis by regulating protein folding, trafficking, and degradation. Notably, polyQ proteins can disrupt the PQC system by sequestering its critical components and impairing its proteasomal functions. Therefore, restoring the PQC system through genetic or pharmacological interventions could potentially offer beneficial effects and alleviate the symptoms of the disease. Here, we will provide a review on the distribution, expression, and genetic or pharmacological intervention of protein quality control system in cellular or animal models of PolyQ SCAs.
Collapse
Affiliation(s)
- Hongfeng Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361005, Fujian, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China.
| | - Xin Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361005, Fujian, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
2
|
Fabri JHTM, Rocha MC, Fernandes CM, Campanella JEM, da Cunha AF, Del Poeta M, Malavazi I. The Heat Shock Transcription Factor HsfA Plays a Role in Membrane Lipids Biosynthesis Connecting Thermotolerance and Unsaturated Fatty Acid Metabolism in Aspergillus fumigatus. Microbiol Spectr 2023; 11:e0162723. [PMID: 37195179 PMCID: PMC10269545 DOI: 10.1128/spectrum.01627-23] [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: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
Thermotolerance is a remarkable virulence attribute of Aspergillus fumigatus, but the consequences of heat shock (HS) to the cell membrane of this fungus are unknown, although this structure is one of the first to detect changes in ambient temperature that imposes on the cell a prompt adaptative response. Under high-temperature stress, fungi trigger the HS response controlled by heat shock transcription factors, such as HsfA, which regulates the expression of heat shock proteins. In yeast, smaller amounts of phospholipids with unsaturated fatty acid (FA) chains are synthesized in response to HS, directly affecting plasma membrane composition. The addition of double bonds in saturated FA is catalyzed by Δ9-fatty acid desaturases, whose expression is temperature-modulated. However, the relationship between HS and saturated/unsaturated FA balance in membrane lipids of A. fumigatus in response to HS has not been investigated. Here, we found that HsfA responds to plasma membrane stress and has a role in sphingolipid and phospholipid unsaturated biosynthesis. In addition, we studied the A. fumigatus Δ9-fatty acid desaturase sdeA and discovered that this gene is essential and required for unsaturated FA biosynthesis, although it did not directly affect the total levels of phospholipids and sphingolipids. sdeA depletion significantly sensitizes mature A. fumigatus biofilms to caspofungin. Also, we demonstrate that hsfA controls sdeA expression, while SdeA and Hsp90 physically interact. Our results suggest that HsfA is required for the adaptation of the fungal plasma membrane to HS and point out a sharp relationship between thermotolerance and FA metabolism in A. fumigatus. IMPORTANCE Aspergillus fumigatus causes invasive pulmonary aspergillosis, a life-threatening infection accounting for high mortality rates in immunocompromised patients. The ability of this organism to grow at elevated temperatures is long recognized as an essential attribute for this mold to cause disease. A. fumigatus responds to heat stress by activating heat shock transcription factors and chaperones to orchestrate cellular responses that protect the fungus against damage caused by heat. Concomitantly, the cell membrane must adapt to heat and maintain physical and chemical properties such as the balance between saturated/unsaturated fatty acids. However, how A. fumigatus connects these two physiological responses is unclear. Here, we explain that HsfA affects the synthesis of complex membrane lipids such as phospholipids and sphingolipids and controls the enzyme SdeA, which produces monounsaturated fatty acids, raw material for membrane lipids. These findings suggest that forced dysregulation of saturated/unsaturated fatty acid balance might represent novel strategies for antifungal therapy.
Collapse
Affiliation(s)
- João Henrique Tadini Marilhano Fabri
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Marina Campos Rocha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Caroline Mota Fernandes
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Jonatas Erick Maimoni Campanella
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Anderson Ferreira da Cunha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, USA
- Veterans Administration Medical Center, Northport, New York, USA
| | - Iran Malavazi
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| |
Collapse
|
3
|
Kinetic principles underlying pioneer function of GAGA transcription factor in live cells. Nat Struct Mol Biol 2022; 29:665-676. [PMID: 35835866 PMCID: PMC10177624 DOI: 10.1038/s41594-022-00800-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/27/2022] [Indexed: 11/09/2022]
Abstract
How pioneer factors interface with chromatin to promote accessibility for transcription control is poorly understood in vivo. Here, we directly visualize chromatin association by the prototypical GAGA pioneer factor (GAF) in live Drosophila hemocytes. Single-particle tracking reveals that most GAF is chromatin bound, with a stable-binding fraction showing nucleosome-like confinement residing on chromatin for more than 2 min, far longer than the dynamic range of most transcription factors. These kinetic properties require the full complement of GAF's DNA-binding, multimerization and intrinsically disordered domains, and are autonomous from recruited chromatin remodelers NURF and PBAP, whose activities primarily benefit GAF's neighbors such as Heat Shock Factor. Evaluation of GAF kinetics together with its endogenous abundance indicates that, despite on-off dynamics, GAF constitutively and fully occupies major chromatin targets, thereby providing a temporal mechanism that sustains open chromatin for transcriptional responses to homeostatic, environmental and developmental signals.
Collapse
|
4
|
Heat Shock Factors in Protein Quality Control and Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:181-199. [PMID: 36472823 DOI: 10.1007/978-3-031-12966-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Proper regulation of cellular protein quality control is crucial for cellular health. It appears that the protein quality control machinery is subjected to distinct regulation in different cellular contexts such as in somatic cells and in germ cells. Heat shock factors (HSFs) play critical role in the control of quality of cellular proteins through controlling expression of many genes encoding different proteins including those for inducible protein chaperones. Mammalian cells exert distinct mechanism of cellular functions through maintenance of tissue-specific HSFs. Here, we have discussed different HSFs and their functions including those during spermatogenesis. We have also discussed the different heat shock proteins induced by the HSFs and their activities in those contexts. We have also identified several small molecule activators and inhibitors of HSFs from different sources reported so far.
Collapse
|
5
|
Lai WKM, Mariani L, Rothschild G, Smith ER, Venters BJ, Blanda TR, Kuntala PK, Bocklund K, Mairose J, Dweikat SN, Mistretta K, Rossi MJ, James D, Anderson JT, Phanor SK, Zhang W, Zhao Z, Shah AP, Novitzky K, McAnarney E, Keogh MC, Shilatifard A, Basu U, Bulyk ML, Pugh BF. A ChIP-exo screen of 887 Protein Capture Reagents Program transcription factor antibodies in human cells. Genome Res 2021; 31:1663-1679. [PMID: 34426512 PMCID: PMC8415381 DOI: 10.1101/gr.275472.121] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/07/2021] [Indexed: 12/22/2022]
Abstract
Antibodies offer a powerful means to interrogate specific proteins in a complex milieu. However, antibody availability and reliability can be problematic, whereas epitope tagging can be impractical in many cases. To address these limitations, the Protein Capture Reagents Program (PCRP) generated over a thousand renewable monoclonal antibodies (mAbs) against human presumptive chromatin proteins. However, these reagents have not been widely field-tested. We therefore performed a screen to test their ability to enrich genomic regions via chromatin immunoprecipitation (ChIP) and a variety of orthogonal assays. Eight hundred eighty-seven unique antibodies against 681 unique human transcription factors (TFs) were assayed by ultra-high-resolution ChIP-exo/seq, generating approximately 1200 ChIP-exo data sets, primarily in a single pass in one cell type (K562). Subsets of PCRP mAbs were further tested in ChIP-seq, CUT&RUN, STORM super-resolution microscopy, immunoblots, and protein binding microarray (PBM) experiments. About 5% of the tested antibodies displayed high-confidence target (i.e., cognate antigen) enrichment across at least one assay and are strong candidates for additional validation. An additional 34% produced ChIP-exo data that were distinct from background and thus warrant further testing. The remaining 61% were not substantially different from background, and likely require consideration of a much broader survey of cell types and/or assay optimizations. We show and discuss the metrics and challenges to antibody validation in chromatin-based assays.
Collapse
Affiliation(s)
- William K M Lai
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Luca Mariani
- Division of Genetics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Edwin R Smith
- Simpson Querrey Institute for Epigenetics and the Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | - Thomas R Blanda
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Prashant K Kuntala
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Kylie Bocklund
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Joshua Mairose
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Sarah N Dweikat
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Katelyn Mistretta
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Matthew J Rossi
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Daniela James
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - James T Anderson
- Division of Genetics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sabrina K Phanor
- Division of Genetics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wanwei Zhang
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Zibo Zhao
- Simpson Querrey Institute for Epigenetics and the Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Avani P Shah
- Simpson Querrey Institute for Epigenetics and the Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | - Ali Shilatifard
- Simpson Querrey Institute for Epigenetics and the Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Uttiya Basu
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - B Franklin Pugh
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
6
|
Lang BJ, Guerrero ME, Prince TL, Okusha Y, Bonorino C, Calderwood SK. The functions and regulation of heat shock proteins; key orchestrators of proteostasis and the heat shock response. Arch Toxicol 2021; 95:1943-1970. [PMID: 34003342 DOI: 10.1007/s00204-021-03070-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Cells respond to protein-damaging (proteotoxic) stress by activation of the Heat Shock Response (HSR). The HSR provides cells with an enhanced ability to endure proteotoxic insults and plays a crucial role in determining subsequent cell death or survival. The HSR is, therefore, a critical factor that influences the toxicity of protein stress. While named for its vital role in the cellular response to heat stress, various components of the HSR system and the molecular chaperone network execute essential physiological functions as well as responses to other diverse toxic insults. The effector molecules of the HSR, the Heat Shock Factors (HSFs) and Heat Shock Proteins (HSPs), are also important regulatory targets in the progression of neurodegenerative diseases and cancers. Modulation of the HSR and/or its extended network have, therefore, become attractive treatment strategies for these diseases. Development of effective therapies will, however, require a detailed understanding of the HSR, important features of which continue to be uncovered and are yet to be completely understood. We review recently described and hallmark mechanistic principles of the HSR, the regulation and functions of HSPs, and contexts in which the HSR is activated and influences cell fate in response to various toxic conditions.
Collapse
Affiliation(s)
- Benjamin J Lang
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Martin E Guerrero
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), 5500, Mendoza, Argentina
| | - Thomas L Prince
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Yuka Okusha
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Cristina Bonorino
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brasil.,Department of Surgery, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
7
|
Moreno R, Banerjee S, Jackson AW, Quinn J, Baillie G, Dixon JE, Dinkova-Kostova AT, Edwards J, de la Vega L. The stress-responsive kinase DYRK2 activates heat shock factor 1 promoting resistance to proteotoxic stress. Cell Death Differ 2021; 28:1563-1578. [PMID: 33268814 PMCID: PMC8166837 DOI: 10.1038/s41418-020-00686-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/23/2022] Open
Abstract
To survive proteotoxic stress, cancer cells activate the proteotoxic-stress response pathway, which is controlled by the transcription factor heat shock factor 1 (HSF1). This pathway supports cancer initiation, cancer progression and chemoresistance and thus is an attractive therapeutic target. As developing inhibitors against transcriptional regulators, such as HSF1 is challenging, the identification and targeting of upstream regulators of HSF1 present a tractable alternative strategy. Here we demonstrate that in triple-negative breast cancer (TNBC) cells, the dual specificity tyrosine-regulated kinase 2 (DYRK2) phosphorylates HSF1, promoting its nuclear stability and transcriptional activity. DYRK2 depletion reduces HSF1 activity and sensitises TNBC cells to proteotoxic stress. Importantly, in tumours from TNBC patients, DYRK2 levels positively correlate with active HSF1 and associates with poor prognosis, suggesting that DYRK2 could be promoting TNBC. These findings identify DYRK2 as a key modulator of the HSF1 transcriptional programme and a potential therapeutic target.
Collapse
Affiliation(s)
- Rita Moreno
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Sourav Banerjee
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093-0721, USA
| | - Angus W Jackson
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Jean Quinn
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gregg Baillie
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jack E Dixon
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093-0721, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Joanne Edwards
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Laureano de la Vega
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK.
| |
Collapse
|
8
|
Tandon V, de la Vega L, Banerjee S. Emerging roles of DYRK2 in cancer. J Biol Chem 2021; 296:100233. [PMID: 33376136 PMCID: PMC7948649 DOI: 10.1074/jbc.rev120.015217] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the CMGC kinase DYRK2 has been reported as a tumor suppressor across various cancers triggering major antitumor and proapoptotic signals in breast, colon, liver, ovary, brain, and lung cancers, with lower DYRK2 expression correlated with poorer prognosis in patients. Contrary to this, various medicinal chemistry studies reported robust antiproliferative properties of DYRK2 inhibitors, whereas unbiased 'omics' and genome-wide association study-based studies identified DYRK2 as a highly overexpressed kinase in various patient tumor samples. A major paradigm shift occurred in the last 4 years when DYRK2 was found to regulate proteostasis in cancer via a two-pronged mechanism. DYRK2 phosphorylated and activated the 26S proteasome to enhance degradation of misfolded/tumor-suppressor proteins while also promoting the nuclear stability and transcriptional activity of its substrate, heat-shock factor 1 triggering protein folding. Together, DYRK2 regulates proteostasis and promotes protumorigenic survival for specific cancers. Indeed, potent and selective small-molecule inhibitors of DYRK2 exhibit in vitro and in vivo anti-tumor activity in triple-negative breast cancer and myeloma models. However, with conflicting and contradictory reports across different cancers, the overarching role of DYRK2 remains enigmatic. Specific cancer (sub)types coupled to spatiotemporal interactions with substrates could decide the procancer or anticancer role of DYRK2. The current review aims to provide a balanced and critical appreciation of the literature to date, highlighting top substrates such as p53, c-Myc, c-Jun, heat-shock factor 1, proteasome, or NOTCH1, to discuss DYRK2 inhibitors available to the scientific community and to shed light on this duality of protumorigenic and antitumorigenic roles of DYRK2.
Collapse
Affiliation(s)
- Vasudha Tandon
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Laureano de la Vega
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Sourav Banerjee
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
9
|
Wang Y, Chen Z, Li Y, Ma L, Zou Y, Wang X, Yin C, Pan L, Shen Y, Jia J, Yuan J, Zhang G, Yang C, Ge J, Zou Y, Gong H. Low density lipoprotein receptor related protein 6 (LRP6) protects heart against oxidative stress by the crosstalk of HSF1 and GSK3β. Redox Biol 2020; 37:101699. [PMID: 32905882 PMCID: PMC7486456 DOI: 10.1016/j.redox.2020.101699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Low density lipoprotein receptor-related protein 6 (LRP6), a Wnt co-receptor, induces multiple functions in various organs. We recently reported cardiac specific LRP6 deficiency caused cardiac dysfunction in mice. Whether cardiomyocyte-expressed LRP6 protects hearts against ischemic stress is largely unknown. Here, we investigated the effects of cardiac LRP6 in response to ischemic reperfusion (I/R) injury. Tamoxifen inducible cardiac specific LRP6 overexpression mice were generated to build I/R model by occlusion of the left anterior descending (LAD) coronary artery for 40 min and subsequent release of specific time. Cardiac specific LRP6 overexpression significantly ameliorated myocardial I/R injury as characterized by the improved cardiac function, strain pattern and infarct area at 24 h after reperfusion. I/R induced-apoptosis and endoplasmic reticulum (ER) stress were greatly inhibited by LRP6 overexpression in cardiomyocytes. LRP6 overexpression enhanced the expression of heat shock transcription factor-1(HSF1) and heat shock proteins (HSPs), the level of p-glycogen synthase kinase 3β(GSK3β)(S9) and p-AMPK under I/R. HSF1 inhibitor deteriorated the apoptosis and decreased p-GSK3β(S9) level in LRP6 overexpressed -cardiomyocytes treated with H2O2. Si-HSF1 or overexpression of active GSK3β significantly attenuated the increased expression of HSF1 and p-AMPK, and the inhibition of apoptosis and ER stress induced by LRP6 overexpression in H2O2-treated cardiomyocytes. AMPK inhibitor suppressed the increase in p-GSK3β (S9) level but didn't alter HSF1 nucleus expression in LRP6 overexpressed-cardiomyocytes treated with H2O2. Active GSK3β, but not AMPK inhibitor, attenuated the inhibition of ubiquitination of HSF1 induced by LRP6-overexpressed-cardiomyocytes treated with H2O2. LRP6 overexpression increased interaction of HSF1 and GSK3β which may be involved in the reciprocal regulation under oxidative stress. In conclusion, cardiac LRP6 overexpression significantly inhibits cardiomyocyte apoptosis and ameliorates myocardial I/R injury by the crosstalk of HSF1 and GSK3β signaling.
Collapse
Affiliation(s)
- Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhidan Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Leilei Ma
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yan Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Le Pan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yi Shen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianguo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jie Yuan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guoping Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chunjie Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
van Marion DMS, Lanters EAH, Ramos KS, Li J, Wiersma M, Baks-te Bulte L, J. Q. M. Muskens A, Boersma E, de Groot NMS, Brundel BJJM. Evaluating Serum Heat Shock Protein Levels as Novel Biomarkers for Atrial Fibrillation. Cells 2020; 9:E2105. [PMID: 32947824 PMCID: PMC7564530 DOI: 10.3390/cells9092105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Staging of atrial fibrillation (AF) is essential to understanding disease progression and the accompanied increase in therapy failure. Blood-based heat shock protein (HSP) levels may enable staging of AF and the identification of patients with higher risk for AF recurrence after treatment. Objective: This study evaluates the relationship between serum HSP levels, presence of AF, AF stage and AF recurrence following electrocardioversion (ECV) or pulmonary vein isolation (PVI). Methods: To determine HSP27, HSP70, cardiovascular (cv)HSP and HSP60 levels, serum samples were collected from control patients without AF and patients with paroxysmal atrial fibrillation (PAF), persistent (PeAF) and longstanding persistent (LSPeAF) AF, presenting for ECV or PVI, prior to intervention and at 3-, 6- and 12-months post-PVI. Results: The study population (n = 297) consisted of 98 control and 199 AF patients admitted for ECV (n = 98) or PVI (n = 101). HSP27, HSP70, cvHSP and HSP60 serum levels did not differ between patients without or with PAF, PeAF or LSPeAF. Additionally, baseline HSP levels did not correlate with AF recurrence after ECV or PVI. However, in AF patients with AF recurrence, HSP27 levels were significantly elevated post-PVI relative to baseline, compared to patients without recurrence. Conclusions: No association was observed between baseline HSP levels and the presence of AF, AF stage or AF recurrence. However, HSP27 levels were increased in serum samples of patients with AF recurrence within one year after PVI, suggesting that HSP27 levels may predict recurrence of AF after ablative therapy.
Collapse
Affiliation(s)
- Denise M. S. van Marion
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Eva A. H. Lanters
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Kennedy S. Ramos
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Jin Li
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Marit Wiersma
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
- Netherlands Heart Institute, 3511EP Utrecht, The Netherlands
| | - Luciënne Baks-te Bulte
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Agnes J. Q. M. Muskens
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Natasja M. S. de Groot
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| |
Collapse
|
11
|
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: 74] [Impact Index Per Article: 18.5] [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
|
12
|
Abstract
Atrial fibrillation (AF), the most common progressive and age-related cardiac arrhythmia, affects millions of people worldwide. AF is associated with common risk factors, including hypertension, diabetes mellitus, and obesity, and serious complications such as stroke and heart failure. Notably, AF is progressive in nature, and because current treatment options are mainly symptomatic, they have only a moderate effect on prevention of arrhythmia progression. Hereto, there is an urgent unmet need to develop mechanistic treatments directed at root causes of AF. Recent research findings indicate a key role for inflammasomes and derailed proteostasis as root causes of AF. Here, we elaborate on the molecular mechanisms of these 2 emerging key pathways driving the pathogenesis of AF. First the role of NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome on AF pathogenesis and cardiomyocyte remodeling is discussed. Then we highlight pathways of proteostasis derailment, including exhaustion of cardioprotective heat shock proteins, disruption of cytoskeletal proteins via histone deacetylases, and the recently discovered DNA damage-induced nicotinamide adenine dinucleotide+ depletion to underlie AF. Moreover, potential interactions between the inflammasomes and proteostasis pathways are discussed and possible therapeutic targets within these pathways indicated.
Collapse
Affiliation(s)
- Na Li
- From the Department of Medicine (Cardiovascular Research) (N.L.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (N.L.), Baylor College of Medicine, Houston, TX.,Cardiovascular Research Institute (N.L.), Baylor College of Medicine, Houston, TX
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, the Netherlands (B.J.J.M.B.)
| |
Collapse
|
13
|
Katiyar A, Fujimoto M, Tan K, Kurashima A, Srivastava P, Okada M, Takii R, Nakai A. HSF1 is required for induction of mitochondrial chaperones during the mitochondrial unfolded protein response. FEBS Open Bio 2020; 10:1135-1148. [PMID: 32302062 PMCID: PMC7262932 DOI: 10.1002/2211-5463.12863] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 01/09/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt ) is characterized by the transcriptional induction of mitochondrial chaperone and protease genes in response to impaired mitochondrial proteostasis and is regulated by ATF5 and CHOP in mammalian cells. However, the detailed mechanisms underlying the UPRmt are currently unclear. Here, we show that HSF1 is required for activation of mitochondrial chaperone genes, including HSP60, HSP10, and mtHSP70, in mouse embryonic fibroblasts during inhibition of matrix chaperone TRAP1, protease Lon, or electron transfer complex 1 activity. HSF1 bound constitutively to mitochondrial chaperone gene promoters, and we observed that its occupancy was remarkably enhanced at different levels during the UPRmt . Furthermore, HSF1 supported the maintenance of mitochondrial function under the same conditions. These results demonstrate that HSF1 is required for induction of mitochondrial chaperones during the UPRmt , and thus, it may be one of the guardians of mitochondrial function under conditions of impaired mitochondrial proteostasis.
Collapse
Affiliation(s)
- Arpit Katiyar
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Ke Tan
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
- Present address:
Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei ProvinceCollege of Life SciencesHebei Normal UniversityShijiazhuangHebei050024China
| | - Ai Kurashima
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Pratibha Srivastava
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Mariko Okada
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Ryosuke Takii
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| | - Akira Nakai
- Department of Biochemistry and Molecular BiologyYamaguchi University School of MedicineUbeJapan
| |
Collapse
|
14
|
Tian X, Zhou N, Yuan J, Lu L, Zhang Q, Wei M, Zou Y, Yuan L. Heat shock transcription factor 1 regulates exercise-induced myocardial angiogenesis after pressure overload via HIF-1α/VEGF pathway. J Cell Mol Med 2020; 24:2178-2188. [PMID: 31930683 PMCID: PMC7011135 DOI: 10.1111/jcmm.14872] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Exercise training is believed to have a positive effect on cardiac hypertrophy after hypertension. However, its mechanism is still not fully understood. Herein, our findings suggest that heat shock transcription factor 1 (HSF1) improves exercise‐initiated myocardial angiogenesis after pressure overload. A sustained narrowing of the diagonal aorta (TAC) and moderately‐ intense exercise training protocol were imposed on HSF1 heterozygote (KO) and their littermate wild‐type (WT) male mice. After two months, the cardiac function was assessed using the adaptive responses to exercise training, or TAC, or both of them such as catheterization and echocardiography. The HE stains assessed the area of myocyte cross‐sectional. The Western blot and real‐time PCR measured the levels of expression for heat shock factor 1 (HSF1), vascular endothelial growth factor (VEGF) and hypoxia inducible factor‐1 alpha (HIF‐1α) in cardiac tissues. The anti‐CD31 antibody immunohistochemical staining was done to examine how exercise training influenced cardiac ontogeny. The outcome illustrated that exercise training significantly improved the cardiac ontogeny in TAC mice, which was convoyed by elevated levels of expression for VEGF and HIF‐1α and preserved the heart microvascular density. More importantly, HSF1 deficiency impaired these effects induced by exercise training in TAC mice. In conclusion, exercise training encourages cardiac ontogeny by means of HSF1 activation and successive HIF‐1α/VEGF up‐regulation in endothelial cells during continued pressure overload.
Collapse
Affiliation(s)
- Xu Tian
- Department of Kinesiology, Institute of Physical Education, Shanghai Normal University, Shanghai, China
| | - Ning Zhou
- Section of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Yuan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| | - Le Lu
- Department of Kinesiology, Institute of Physical Education, Shanghai Normal University, Shanghai, China
| | - Qi Zhang
- Department of Kinesiology, Institute of Physical Education, Shanghai Normal University, Shanghai, China
| | - Minmin Wei
- Department of Kinesiology, Institute of Physical Education, Shanghai Normal University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| | - Lingyan Yuan
- Department of Kinesiology, Institute of Physical Education, Shanghai Normal University, Shanghai, China
| |
Collapse
|
15
|
Gao Y, Kim S, Lee YI, Lee J. Cellular Stress-Modulating Drugs Can Potentially Be Identified by in Silico Screening with Connectivity Map (CMap). Int J Mol Sci 2019; 20:ijms20225601. [PMID: 31717493 PMCID: PMC6888006 DOI: 10.3390/ijms20225601] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/27/2022] Open
Abstract
Accompanied by increased life span, aging-associated diseases, such as metabolic diseases and cancers, have become serious health threats. Recent studies have documented that aging-associated diseases are caused by prolonged cellular stresses such as endoplasmic reticulum (ER) stress, mitochondrial stress, and oxidative stress. Thus, ameliorating cellular stresses could be an effective approach to treat aging-associated diseases and, more importantly, to prevent such diseases from happening. However, cellular stresses and their molecular responses within the cell are typically mediated by a variety of factors encompassing different signaling pathways. Therefore, a target-based drug discovery method currently being used widely (reverse pharmacology) may not be adequate to uncover novel drugs targeting cellular stresses and related diseases. The connectivity map (CMap) is an online pharmacogenomic database cataloging gene expression data from cultured cells treated individually with various chemicals, including a variety of phytochemicals. Moreover, by querying through CMap, researchers may screen registered chemicals in silico and obtain the likelihood of drugs showing a similar gene expression profile with desired and chemopreventive conditions. Thus, CMap is an effective genome-based tool to discover novel chemopreventive drugs.
Collapse
Affiliation(s)
- Yurong Gao
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (Y.G.); (S.K.)
| | - Sungwoo Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (Y.G.); (S.K.)
| | - Yun-Il Lee
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence: (Y.-I.L.); (J.L.)
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (Y.G.); (S.K.)
- Correspondence: (Y.-I.L.); (J.L.)
| |
Collapse
|
16
|
Takii R, Fujimoto M, Matsumoto M, Srivastava P, Katiyar A, Nakayama KI, Nakai A. The pericentromeric protein shugoshin 2 cooperates with HSF1 in heat shock response and RNA Pol II recruitment. EMBO J 2019; 38:e102566. [PMID: 31657478 DOI: 10.15252/embj.2019102566] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/17/2022] Open
Abstract
The recruitment of RNA polymerase II (Pol II) to core promoters is highly regulated during rapid induction of genes. In response to heat shock, heat shock transcription factor 1 (HSF1) is activated and occupies heat shock gene promoters. Promoter-bound HSF1 recruits general transcription factors and Mediator, which interact with Pol II, but stress-specific mechanisms of Pol II recruitment are unclear. Here, we show in comparative analyses of HSF1 paralogs and their mutants that HSF1 interacts with the pericentromeric adaptor protein shugoshin 2 (SGO2) during heat shock in mouse cells, in a manner dependent on inducible phosphorylation of HSF1 at serine 326, and recruits SGO2 to the HSP70 promoter. SGO2-mediated binding and recruitment of Pol II with a hypophosphorylated C-terminal domain promote expression of HSP70, implicating SGO2 as one of the coactivators that facilitate Pol II recruitment by HSF1. Furthermore, the HSF1-SGO2 complex supports cell survival and maintenance of proteostasis in heat shock conditions. These results exemplify a proteotoxic stress-specific mechanism of Pol II recruitment, which is triggered by phosphorylation of HSF1 during the heat shock response.
Collapse
Affiliation(s)
- Ryosuke Takii
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Masaki Matsumoto
- Division of Proteomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Pratibha Srivastava
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Arpit Katiyar
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Keiich I Nakayama
- Division of Proteomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| |
Collapse
|
17
|
Zhang L, Hu Z, Zhang Y, Huang J, Yang X, Wang J. Proteomics analysis of proteins interacting with heat shock factor 1 in squamous cell carcinoma of the cervix. Oncol Lett 2019; 18:2568-2575. [PMID: 31402952 DOI: 10.3892/ol.2019.10539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/07/2019] [Indexed: 01/08/2023] Open
Abstract
Protein interactions are crucial for maintaining homeostasis. Heat shock factor 1 (HSF1), a transcription factor that interacts with various proteins, is highly expressed in squamous cell carcinoma (SCC) of the cervix. The aim of the present study was to investigate the protein interaction profile of HSF1 in cervical SCC. Proteins interacting with HSF1 in SCC tissue and non-cancerous control (Ctrl) tissue were obtained by immunoprecipitation, separated by SDS-PAGE, identified by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry and analyzed using bioinformatics methods. A total of 220 and 241 proteins were identified by mass spectrometry in the tissues of Ctrl and SCC samples, respectively, among which 172 were detected exclusively in SCC (Pro-S), 151 exclusively in Ctrl (Pro-C) and 69 in both groups (Pro-B). The protein interaction profiles were different in each group; the STRING database identified three proteins that interacted with HSF1 directly, including insulin-like growth factor 1 receptor and small nuclear RNA-activating protein complex subunit 4 in Pro-C and small ubiquitin-related modifier 1 in Pro-S. Functional enrichment analysis of Gene Ontology revealed that the top terms were alternative splicing in Pro-S and polymorphism in Pro-C. In Pro-S, more categories were related to protein modification, such as phosphorylation, ubiquitination and acetylation. Therefore, HSF1 may influence the occurrence and development of cervical SCC by interacting with specific proteins.
Collapse
Affiliation(s)
- Lingli Zhang
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zhe Hu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Ying Zhang
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jinzhi Huang
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xuefen Yang
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jiafeng Wang
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
18
|
Patinen T, Adinolfi S, Cortés CC, Härkönen J, Jawahar Deen A, Levonen AL. Regulation of stress signaling pathways by protein lipoxidation. Redox Biol 2019; 23:101114. [PMID: 30709792 PMCID: PMC6859545 DOI: 10.1016/j.redox.2019.101114] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 12/30/2022] Open
Abstract
Enzymatic and non-enzymatic oxidation of unsaturated fatty acids gives rise to reactive species that covalently modify nucleophilic residues within redox sensitive protein sensors in a process called lipoxidation. This triggers adaptive signaling pathways that ultimately lead to increased resistance to stress. In this graphical review, we will provide an overview of pathways affected by protein lipoxidation and the key signaling proteins being altered, focusing on the KEAP1-NRF2 and heat shock response pathways. We review the mechanisms by which lipid peroxidation products can serve as second messengers and evoke cellular responses via covalent modification of key sensors of altered cellular environment, ultimately leading to adaptation to stress.
Collapse
Affiliation(s)
- Tommi Patinen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Simone Adinolfi
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Carlos Cruz Cortés
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland; Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City MX-07360, Mexico
| | - Jouni Härkönen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Ashik Jawahar Deen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Anna-Liisa Levonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland.
| |
Collapse
|
19
|
Liu P, Chen X, Zhu J, Li B, Chen Z, Wang G, Sun H, Xu Z, Zhao Z, Zhou C, Xie C, Lou L, Zhu W. Design, Synthesis and Pharmacological Evaluation of Novel Hsp90N-terminal Inhibitors Without Induction of Heat Shock Response. ChemistryOpen 2019; 8:344-353. [PMID: 30976475 PMCID: PMC6437812 DOI: 10.1002/open.201900055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/28/2019] [Indexed: 01/24/2023] Open
Abstract
Heat shock protein 90 (Hsp90) is a potential oncogenic target. However, Hsp90 inhibitors in clinical trial induce heat shock response, resulting in drug resistance and inefficiency. In this study, we designed and synthesized a series of novel triazine derivatives (A1‐26, B1‐13, C1‐23) as Hsp90 inhibitors. Compound A14 directly bound to Hsp90 in a different manner from traditional Hsp90 inhibitors, and degraded client proteins, but did not induce the concomitant activation of Hsp72. Importantly, A14 exhibited the most potent anti‐proliferation ability by inducing autophagy, with the IC50 values of 0.1 μM and 0.4 μM in A549 and SK‐BR‐3 cell lines, respectively. The in
vivo study demonstrated that A14 could induce autophagy and degrade Hsp90 client proteins in tumor tissues, and exhibit anti‐tumor activity in A549 lung cancer xenografts. Therefore, the compound A14 with potent antitumor activity and unique pharmacological characteristics is a novel Hsp90 inhibitor for developing anticancer agent without heat shock response.
Collapse
Affiliation(s)
- Peng Liu
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Xiangling Chen
- Division of Anti-Tumor Pharmacology Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Jianming Zhu
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Bo Li
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Zhaoqiang Chen
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Guimin Wang
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Haiguo Sun
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Zhijian Xu
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Zhixin Zhao
- Division of Anti-Tumor Pharmacology Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Chen Zhou
- Division of Anti-Tumor Pharmacology Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Chengying Xie
- Division of Anti-Tumor Pharmacology Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Liguang Lou
- Division of Anti-Tumor Pharmacology Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China
| | - Weiliang Zhu
- Key Laboratory of Receptor Research Drug Discovery and Design Center Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China.,University of Chinese Academy of Sciences No.19 A Yuquan Road Beijing 100049 China.,Open Studio for Druggability Research of Marine Natural Products Pilot National Laboratory for Marine Science and Technology (Qingdao) 1 Wenhai Road, Aoshanwei, Jimo Qingdao 266237 China
| |
Collapse
|
20
|
van Marion DM, Hu X, Zhang D, Hoogstra-Berends F, Seerden JPG, Loen L, Heeres A, Steen H, Henning RH, Brundel BJ. Screening of novel HSP-inducing compounds to conserve cardiomyocyte function in experimental atrial fibrillation. Drug Des Devel Ther 2019; 13:345-364. [PMID: 30705583 PMCID: PMC6342224 DOI: 10.2147/dddt.s176924] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The heat shock protein (HSP) inducer, geranylgeranylacetone (GGA), was previously found to protect against atrial fibrillation (AF) remodeling in experimental model systems. Clinical application of GGA in AF is limited, due to low systemic concentrations owing to the hydrophobic character of GGA. Objectives To identify novel HSP-inducing compounds, with improved physicochemical properties, that prevent contractile dysfunction in experimental model systems for AF. Methods Eighty-one GGA-derivatives were synthesized and explored for their HSP-inducing properties by assessment of HSP expression in HL-1 cardiomyocytes pretreated with or without a mild heat shock (HS), followed by incubation with 10 µM GGA or GGA-derivative. Subsequently, the most potent HSP-inducers were tested for preservation of calcium transient (CaT) amplitudes or heart wall contraction in pretreated tachypaced HL-1 cardiomyocytes (with or without HSPB1 siRNA) and Drosophilas, respectively. Finally, CaT recovery in tachypaced HL-1 cardiomyocytes posttreated with GGA or protective GGA-derivatives was determined. Results Thirty GGA-derivatives significantly induced HSPA1A expression after HS, and seven showed exceeding HSPA1A expression compared to GGA. GGA and nine GGA-derivatives protected significantly from tachypacing (TP)-induced CaT loss, which was abrogated by HSPB1 suppression. GGA and four potent GGA-derivatives protected against heart wall dysfunction after TP compared to non-paced control Drosophilas. Of these compounds, GGA and three GGA-derivatives induced a significant restoration from CaT loss after TP of HL-1 cardiomyocytes. Conclusion We identified novel GGA-derivatives with improved physicochemical properties compared to GGA. GGA-derivatives, particularly GGA*-59, boost HSP expression resulting in prevention and restoration from TP-induced remodeling, substantiating their role as novel therapeutics in clinical AF.
Collapse
Affiliation(s)
- Denise Ms van Marion
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Xu Hu
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Deli Zhang
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Femke Hoogstra-Berends
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Andre Heeres
- Syncom BV, Groningen, The Netherlands.,Hanze University of Applied Sciences, Groningen, The Netherlands
| | | | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Bianca Jjm Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| |
Collapse
|
21
|
Rossin F, Villella VR, D'Eletto M, Farrace MG, Esposito S, Ferrari E, Monzani R, Occhigrossi L, Pagliarini V, Sette C, Cozza G, Barlev NA, Falasca L, Fimia GM, Kroemer G, Raia V, Maiuri L, Piacentini M. TG2 regulates the heat-shock response by the post-translational modification of HSF1. EMBO Rep 2018; 19:embr.201745067. [PMID: 29752334 DOI: 10.15252/embr.201745067] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/24/2018] [Accepted: 04/13/2018] [Indexed: 01/24/2023] Open
Abstract
Heat-shock factor 1 (HSF1) is the master transcription factor that regulates the response to proteotoxic stress by controlling the transcription of many stress-responsive genes including the heat-shock proteins. Here, we show a novel molecular mechanism controlling the activation of HSF1. We demonstrate that transglutaminase type 2 (TG2), dependent on its protein disulphide isomerase activity, triggers the trimerization and activation of HSF1 regulating adaptation to stress and proteostasis impairment. In particular, we find that TG2 loss of function correlates with a defect in the nuclear translocation of HSF1 and in its DNA-binding ability to the HSP70 promoter. We show that the inhibition of TG2 restores the unbalance in HSF1-HSP70 pathway in cystic fibrosis (CF), a human disorder characterized by deregulation of proteostasis. The absence of TG2 leads to an increase of about 40% in CFTR function in a new experimental CF mouse model lacking TG2. Altogether, these results indicate that TG2 plays a key role in the regulation of cellular proteostasis under stressful cellular conditions through the modulation of the heat-shock response.
Collapse
Affiliation(s)
- Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Valeria Rachela Villella
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | | | - Speranza Esposito
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Ferrari
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Romina Monzani
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Occhigrossi
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Vittoria Pagliarini
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Claudio Sette
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Giorgio Cozza
- Department of Molecular Medicine, University of Padua, Padova, Italy
| | - Nikolai A Barlev
- Gene Expression Laboratory, Institute of Cytology, Saint-Petersburg, Russia
| | - Laura Falasca
- National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy
| | - Gian Maria Fimia
- National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy.,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Guido Kroemer
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Luigi Maiuri
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy.,SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy .,National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy
| |
Collapse
|
22
|
Pujols J, Santos J, Pallarès I, Ventura S. The Disordered C-Terminus of Yeast Hsf1 Contains a Cryptic Low-Complexity Amyloidogenic Region. Int J Mol Sci 2018; 19:ijms19051384. [PMID: 29734798 PMCID: PMC5983738 DOI: 10.3390/ijms19051384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 02/08/2023] Open
Abstract
Response mechanisms to external stress rely on networks of proteins able to activate specific signaling pathways to ensure the maintenance of cell proteostasis. Many of the proteins mediating this kind of response contain intrinsically disordered regions, which lack a defined structure, but still are able to interact with a wide range of clients that modulate the protein function. Some of these interactions are mediated by specific short sequences embedded in the longer disordered regions. Because the physicochemical properties that promote functional and abnormal interactions are similar, it has been shown that, in globular proteins, aggregation-prone and binding regions tend to overlap. It could be that the same principle applies for disordered protein regions. In this context, we show here that a predicted low-complexity interacting region in the disordered C-terminus of the stress response master regulator heat shock factor 1 (Hsf1) protein corresponds to a cryptic amyloid region able to self-assemble into fibrillary structures resembling those found in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jordi Pujols
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
| | - Jaime Santos
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
| | - Irantzu Pallarès
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Barcelona), Spain.
| |
Collapse
|
23
|
Stress pre-conditioning with temperature, UV and gamma radiation induces tolerance against phosphine toxicity. PLoS One 2018; 13:e0195349. [PMID: 29672544 PMCID: PMC5909616 DOI: 10.1371/journal.pone.0195349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/20/2018] [Indexed: 11/19/2022] Open
Abstract
Phosphine is the only general use fumigant for the protection of stored grain, though its long-term utility is threatened by the emergence of highly phosphine-resistant pests. Given this precarious situation, it is essential to identify factors, such as stress preconditioning, that interfere with the efficacy of phosphine fumigation. We used Caenorhabditis elegans as a model organism to test the effect of pre-exposure to heat and cold shock, UV and gamma irradiation on phosphine potency. Heat shock significantly increased tolerance to phosphine by 3-fold in wild-type nematodes, a process that was dependent on the master regulator of the heat shock response, HSF-1. Heat shock did not, however, increase the resistance of a strain carrying the phosphine resistance mutation, dld-1(wr4), and cold shock did not alter the response to phosphine of either strain. Pretreatment with the LD50 of UV (18 J cm-2) did not alter phosphine tolerance in wild-type nematodes, but the LD50 (33 J cm-2) of the phosphine resistant strain (dld-1(wr4)) doubled the level of resistance. In addition, exposure to a mild dose of gamma radiation (200 Gy) elevated the phosphine tolerance by ~2-fold in both strains.
Collapse
|
24
|
Krakowiak J, Zheng X, Patel N, Feder ZA, Anandhakumar J, Valerius K, Gross DS, Khalil AS, Pincus D. Hsf1 and Hsp70 constitute a two-component feedback loop that regulates the yeast heat shock response. eLife 2018; 7:31668. [PMID: 29393852 PMCID: PMC5809143 DOI: 10.7554/elife.31668] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/01/2018] [Indexed: 01/29/2023] Open
Abstract
Models for regulation of the eukaryotic heat shock response typically invoke a negative feedback loop consisting of the transcriptional activator Hsf1 and a molecular chaperone. Previously we identified Hsp70 as the chaperone responsible for Hsf1 repression and constructed a mathematical model that recapitulated the yeast heat shock response (Zheng et al., 2016). The model was based on two assumptions: dissociation of Hsp70 activates Hsf1, and transcriptional induction of Hsp70 deactivates Hsf1. Here we validate these assumptions. First, we severed the feedback loop by uncoupling Hsp70 expression from Hsf1 regulation. As predicted by the model, Hsf1 was unable to efficiently deactivate in the absence of Hsp70 transcriptional induction. Next, we mapped a discrete Hsp70 binding site on Hsf1 to a C-terminal segment known as conserved element 2 (CE2). In vitro, CE2 binds to Hsp70 with low affinity (9 µM), in agreement with model requirements. In cells, removal of CE2 resulted in increased basal Hsf1 activity and delayed deactivation during heat shock, while tandem repeats of CE2 sped up Hsf1 deactivation. Finally, we uncovered a role for the N-terminal domain of Hsf1 in negatively regulating DNA binding. These results reveal the quantitative control mechanisms underlying the heat shock response.
Collapse
Affiliation(s)
- Joanna Krakowiak
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Xu Zheng
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Nikit Patel
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States
| | - Zoë A Feder
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Jayamani Anandhakumar
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States
| | - Kendra Valerius
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - David S Gross
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, United States
| | - Ahmad S Khalil
- Whitehead Institute for Biomedical Research, Cambridge, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, United States
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, United States
| |
Collapse
|
25
|
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: 1.8] [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
|
26
|
Nieto A, Pérez Ishiwara DG, Orozco E, Sánchez Monroy V, Gómez García C. A Novel Heat Shock Element (HSE) in Entamoeba histolytica that Regulates the Transcriptional Activation of the EhPgp5 Gene in the Presence of Emetine Drug. Front Cell Infect Microbiol 2017; 7:492. [PMID: 29238701 PMCID: PMC5712549 DOI: 10.3389/fcimb.2017.00492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/15/2017] [Indexed: 01/01/2023] Open
Abstract
Transcriptional regulation of the multidrug resistance EhPgp5 gene in Entamoeba histolytica is induced by emetine stress. EhPgp5 overexpression alters the chloride-dependent currents that cause trophozoite swelling, diminishing induced programmed cell death (PCD) susceptibility. In contrast, antisense inhibition of P-glycoprotein (PGP) expression produces synchronous death of trophozoites and the enhancement of the biochemical and morphological characteristics of PCD induced by G418. Transcriptional gene regulation analysis identified a 59 bp region at position −170 to −111 bp promoter as putative emetine response elements (EREs). However, insights into transcription factors controlling EhPgp5 gene transcription are missing; to fill this knowledge gap, we used deletion studies and transient CAT activity assays. Our findings suggested an activating motif (−151 to −136 bp) that corresponds to a heat shock element (HSE). Gel-shift assays, UV-crosslinking, binding protein purification, and western blotting assays revealed proteins of 94, 66, 62, and 51 kDa binding to the EhPgp5 HSE that could be heat shock-like transcription factors that regulate the transcriptional activation of the EhPgp5 gene in the presence of emetine drug.
Collapse
Affiliation(s)
- Alma Nieto
- Laboratorio de Biomedicina Molecular I, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - David G Pérez Ishiwara
- Laboratorio de Biomedicina Molecular I, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Virginia Sánchez Monroy
- Laboratorio de Biomedicina Molecular I, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Consuelo Gómez García
- Laboratorio de Biomedicina Molecular I, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
27
|
Takii R, Fujimoto M, Matsuura Y, Wu F, Oshibe N, Takaki E, Katiyar A, Akashi H, Makino T, Kawata M, Nakai A. HSF1 and HSF3 cooperatively regulate the heat shock response in lizards. PLoS One 2017; 12:e0180776. [PMID: 28686674 PMCID: PMC5501597 DOI: 10.1371/journal.pone.0180776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/21/2017] [Indexed: 01/01/2023] Open
Abstract
Cells cope with temperature elevations, which cause protein misfolding, by expressing heat shock proteins (HSPs). This adaptive response is called the heat shock response (HSR), and it is regulated mainly by heat shock transcription factor (HSF). Among the four HSF family members in vertebrates, HSF1 is a master regulator of HSP expression during proteotoxic stress including heat shock in mammals, whereas HSF3 is required for the HSR in birds. To examine whether only one of the HSF family members possesses the potential to induce the HSR in vertebrate animals, we isolated cDNA clones encoding lizard and frog HSF genes. The reconstructed phylogenetic tree of vertebrate HSFs demonstrated that HSF3 in one species is unrelated with that in other species. We found that the DNA-binding activity of both HSF1 and HSF3 in lizard and frog cells was induced in response to heat shock. Unexpectedly, overexpression of lizard and frog HSF3 as well as HSF1 induced HSP70 expression in mouse cells during heat shock, indicating that the two factors have the potential to induce the HSR. Furthermore, knockdown of either HSF3 or HSF1 markedly reduced HSP70 induction in lizard cells and resistance to heat shock. These results demonstrated that HSF1 and HSF3 cooperatively regulate the HSR at least in lizards, and suggest complex mechanisms of the HSR in lizards as well as frogs.
Collapse
Affiliation(s)
- Ryosuke Takii
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Mitsuaki Fujimoto
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Yuki Matsuura
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Fangxu Wu
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Namiko Oshibe
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Eiichi Takaki
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Arpit Katiyar
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
| | - Hiroshi Akashi
- Department of Ecology and Evolutionary Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Takashi Makino
- Department of Ecology and Evolutionary Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Masakado Kawata
- Department of Ecology and Evolutionary Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Akira Nakai
- Departments of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Japan
- * E-mail:
| |
Collapse
|
28
|
Yin J, Jiang XY, Qi W, Ji CG, Xie XL, Zhang DX, Cui ZJ, Wang CK, Bai Y, Wang J, Jiang HQ. piR-823 contributes to colorectal tumorigenesis by enhancing the transcriptional activity of HSF1. Cancer Sci 2017; 108:1746-1756. [PMID: 28618124 PMCID: PMC5581525 DOI: 10.1111/cas.13300] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/02/2017] [Accepted: 06/10/2017] [Indexed: 12/13/2022] Open
Abstract
Piwi-interacting RNAs (piRNAs), a novel class of small non-coding RNAs, were first discovered in germline cells and are thought to silence transposons in spermatogenesis. Recently, piRNAs have also been identified in somatic tissues, and aberrant expression of piRNAs in tumor tissues may be implicated in carcinogenesis. However, the function of piR-823 in colorectal cancer (CRC) remains unclear. Here, we first found that piR-823 was significantly upregulated in CRC tissues compared with its expression in the adjacent tissues. Inhibition of piR-823 suppressed cell proliferation, arrested the cell cycle in the G1 phase and induced cell apoptosis in CRC cell lines HCT116 and DLD-1, whereas overexpression of piR-823 promoted cell proliferation in normal colonic epithelial cell line FHC. Interestingly, Inhibition of piR-823 repressed the expression of heat shock protein (HSP) 27, 60, 70. Furthermore, elevated HSPs expression partially abolished the effect of piR-823 on cell proliferation and apoptosis. In addition, we further demonstrated that piR-823 increased the transcriptional activity of HSF1, the common transcription factor of HSPs, by binding to HSF1 and promoting its phosphorylation at Ser326. Our study reveals that piR-823 plays a tumor-promoting role by upregulating phosphorylation and transcriptional activity of HSF1 and suggests piR-823 as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Jie Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Xiao-Yu Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Wei Qi
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Chen-Guang Ji
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Xiao-Li Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Dong-Xuan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Zi-Jin Cui
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Cun-Kai Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Yun Bai
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| | - Jia Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei.,Ronghe Biotechnology Co., Ltd, Shijiazhuang, Hebei, China
| | - Hui-Qing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, Hebei
| |
Collapse
|
29
|
Saia-Cereda VM, Santana AG, Schmitt A, Falkai P, Martins-de-Souza D. The Nuclear Proteome of White and Gray Matter from Schizophrenia Postmortem Brains. MOLECULAR NEUROPSYCHIATRY 2017; 3:37-52. [PMID: 28879200 PMCID: PMC5582429 DOI: 10.1159/000477299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SCZ) is a serious neuropsychiatric disorder that manifests through several symptoms from early adulthood. Numerous studies over the last decades have led to significant advances in increasing our understanding of the factors involved in SCZ. For example, mass spectrometry-based proteomic analysis has provided important insights by uncovering protein dysfunctions inherent to SCZ. Here, we present a comprehensive analysis of the nuclear proteome of postmortem brain tissues from corpus callosum (CC) and anterior temporal lobe (ATL). We show an overview of the role of deregulated nuclear proteins in these two main regions of the brain: the first, mostly composed of glial cells and axons of neurons, and the second, represented mainly by neuronal cell bodies. These samples were collected from SCZ patients in an attempt to characterize the role of the nucleus in the disease process. With the ATL nucleus enrichment, we found 224 proteins present at different levels, and 76 of these were nuclear proteins. In the CC analysis, we identified 119 present at different levels, and 24 of these were nuclear proteins. The differentially expressed nuclear proteins of ATL are mainly associated with the spliceosome, whereas those of the CC region are associated with calcium/calmodulin signaling.
Collapse
Affiliation(s)
- Verônica M. Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Aline G. Santana
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
- Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- UNICAMP's Neurobiology Center, Campinas, Brazil
| |
Collapse
|
30
|
Kim HJ, Lee JJ, Cho JH, Jeong J, Park AY, Kang W, Lee KJ. Heterogeneous nuclear ribonucleoprotein K inhibits heat shock-induced transcriptional activity of heat shock factor 1. J Biol Chem 2017; 292:12801-12812. [PMID: 28592492 DOI: 10.1074/jbc.m117.774992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 06/06/2017] [Indexed: 12/22/2022] Open
Abstract
When cells are exposed to heat shock and various other stresses, heat shock factor 1 (HSF1) is activated, and the heat shock response (HSR) is elicited. To better understand the molecular regulation of the HSR, we used 2D-PAGE-based proteome analysis to screen for heat shock-induced post-translationally modified cellular proteins. Our analysis revealed that two protein spots typically present on 2D-PAGE gels and containing heterogeneous nuclear ribonucleoprotein K (hnRNP K) with trioxidized Cys132 disappeared after the heat shock treatment and reappeared during recovery, but the total amount of hnRNP K protein remained unchanged. We next tested whether hnRNP K plays a role in HSR by regulating HSF1 and found that hnRNP K inhibits HSF1 activity, resulting in reduced expression of hsp70 and hsp27 mRNAs. hnRNP K also reduced binding affinity of HSF1 to the heat shock element by directly interacting with HSF1 but did not affect HSF1 phosphorylation-dependent activation or nuclear localization. hnRNP K lost its ability to induce these effects when its Cys132 was substituted with Ser, Asp, or Glu. These findings suggest that hnRNP K inhibits transcriptional activity of HSF1 by inhibiting its binding to heat shock element and that the oxidation status of Cys132 in hnRNP K is critical for this inhibition.
Collapse
Affiliation(s)
- Hee-Jung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Jae-Jin Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Jin-Hwan Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Jaeho Jeong
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - A Young Park
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Wonmo Kang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Kong-Joo Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea.
| |
Collapse
|
31
|
Sala AJ, Bott LC, Morimoto RI. Shaping proteostasis at the cellular, tissue, and organismal level. J Cell Biol 2017; 216:1231-1241. [PMID: 28400444 PMCID: PMC5412572 DOI: 10.1083/jcb.201612111] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 01/22/2023] Open
Abstract
The proteostasis network (PN) regulates protein synthesis, folding, transport, and degradation to maintain proteome integrity and limit the accumulation of protein aggregates, a hallmark of aging and degenerative diseases. In multicellular organisms, the PN is regulated at the cellular, tissue, and systemic level to ensure organismal health and longevity. Here we review these three layers of PN regulation and examine how they collectively maintain cellular homeostasis, achieve cell type-specific proteomes, and coordinate proteostasis across tissues. A precise understanding of these layers of control has important implications for organismal health and could offer new therapeutic approaches for neurodegenerative diseases and other chronic disorders related to PN dysfunction.
Collapse
Affiliation(s)
- Ambre J Sala
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208
| | - Laura C Bott
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208
| |
Collapse
|
32
|
Li J, Chauve L, Phelps G, Brielmann RM, Morimoto RI. E2F coregulates an essential HSF developmental program that is distinct from the heat-shock response. Genes Dev 2016; 30:2062-2075. [PMID: 27688402 PMCID: PMC5066613 DOI: 10.1101/gad.283317.116] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/06/2016] [Indexed: 01/08/2023]
Abstract
Heat-shock factor (HSF) is the master transcriptional regulator of the heat-shock response (HSR) and is essential for stress resilience. HSF is also required for metazoan development; however, its function and regulation in this process are poorly understood. Here, we characterize the genomic distribution and transcriptional activity of Caenorhabditis elegans HSF-1 during larval development and show that the developmental HSF-1 transcriptional program is distinct from the HSR. HSF-1 developmental activation requires binding of E2F/DP to a GC-rich motif that facilitates HSF-1 binding to a heat-shock element (HSE) that is degenerate from the consensus HSE sequence and adjacent to the E2F-binding site at promoters. In contrast, induction of the HSR is independent of these promoter elements or E2F/DP and instead requires a distinct set of tandem canonical HSEs. Together, E2F and HSF-1 directly regulate a gene network, including a specific subset of chaperones, to promote protein biogenesis and anabolic metabolism, which are essential in development.
Collapse
Affiliation(s)
- Jian Li
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Laetitia Chauve
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Grace Phelps
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Renée M Brielmann
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|
33
|
Knippertz I, Deinzer A, Dörrie J, Schaft N, Nettelbeck DM, Steinkasserer A. Transcriptional Targeting of Mature Dendritic Cells with Adenoviral Vectors via a Modular Promoter System for Antigen Expression and Functional Manipulation. J Immunol Res 2016; 2016:6078473. [PMID: 27446966 PMCID: PMC4942663 DOI: 10.1155/2016/6078473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/29/2016] [Indexed: 02/06/2023] Open
Abstract
To specifically target dendritic cells (DCs) to simultaneously express different therapeutic transgenes for inducing immune responses against tumors, we used a combined promoter system of adenoviral vectors. We selected a 216 bp short Hsp70B' core promoter induced by a mutated, constitutively active heat shock factor (mHSF) 1 to drive strong gene expression of therapeutic transgenes MelanA, BclxL, and IL-12p70 in HeLa cells, as well as in mature DCs (mDCs). As this involves overexpressing mHSF1, we first evaluated the resulting effects on DCs regarding upregulation of heat shock proteins and maturation markers, toxicity, cytokine profile, and capacity to induce antigen-specific CD8(+) T cells. Second, we generated the two-vector-based "modular promoter" system, where one vector contains the mHSF1 under the control of the human CD83 promoter, which is specifically active only in DCs and after maturation. mHSF1, in turn, activates the Hsp70B' core promotor-driven expression of transgenes MelanA and IL-12p70 in the DC-like cell line XS52 and in human mature and hence immunogenic DCs, but not in tolerogenic immature DCs. These in vitro experiments provide the basis for an in vivo targeting of mature DCs for the expression of multiple transgenes. Therefore, this modular promoter system represents a promising tool for future DC-based immunotherapies in vivo.
Collapse
Affiliation(s)
- Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Andrea Deinzer
- Department of Immune Modulation at the Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Dirk M. Nettelbeck
- German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| |
Collapse
|
34
|
Piri N, Kwong JMK, Gu L, Caprioli J. Heat shock proteins in the retina: Focus on HSP70 and alpha crystallins in ganglion cell survival. Prog Retin Eye Res 2016; 52:22-46. [PMID: 27017896 PMCID: PMC4842330 DOI: 10.1016/j.preteyeres.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/14/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Heat shock proteins (HSPs) belong to a superfamily of stress proteins that are critical constituents of a complex defense mechanism that enhances cell survival under adverse environmental conditions. Cell protective roles of HSPs are related to their chaperone functions, antiapoptotic and antinecrotic effects. HSPs' anti-apoptotic and cytoprotective characteristics, their ability to protect cells from a variety of stressful stimuli, and the possibility of their pharmacological induction in cells under pathological stress make these proteins an attractive therapeutic target for various neurodegenerative diseases; these include Alzheimer's, Parkinson's, Huntington's, prion disease, and others. This review discusses the possible roles of HSPs, particularly HSP70 and small HSPs (alpha A and alpha B crystallins) in enhancing the survival of retinal ganglion cells (RGCs) in optic neuropathies such as glaucoma, which is characterized by progressive loss of vision caused by degeneration of RGCs and their axons in the optic nerve. Studies in animal models of RGC degeneration induced by ocular hypertension, optic nerve crush and axotomy show that upregulation of HSP70 expression by hyperthermia, zinc, geranyl-geranyl acetone, 17-AAG (a HSP90 inhibitor), or through transfection of retinal cells with AAV2-HSP70 effectively supports the survival of injured RGCs. RGCs survival was also stimulated by overexpression of alpha A and alpha B crystallins. These findings provide support for translating the HSP70- and alpha crystallin-based cell survival strategy into therapy to protect and rescue injured RGCs from degeneration associated with glaucomatous and other optic neuropathies.
Collapse
Affiliation(s)
- Natik Piri
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, CA 90095, USA.
| | - Jacky M K Kwong
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Lei Gu
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Joseph Caprioli
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
35
|
Hentze N, Le Breton L, Wiesner J, Kempf G, Mayer MP. Molecular mechanism of thermosensory function of human heat shock transcription factor Hsf1. eLife 2016; 5. [PMID: 26785146 PMCID: PMC4775227 DOI: 10.7554/elife.11576] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/18/2016] [Indexed: 01/06/2023] Open
Abstract
The heat shock response is a universal homeostatic cell autonomous reaction of organisms to cope with adverse environmental conditions. In mammalian cells, this response is mediated by the heat shock transcription factor Hsf1, which is monomeric in unstressed cells and upon activation trimerizes, and binds to promoters of heat shock genes. To understand the basic principle of Hsf1 activation we analyzed temperature-induced alterations in the conformational dynamics of Hsf1 by hydrogen exchange mass spectrometry. We found a temperature-dependent unfolding of Hsf1 in the regulatory region happening concomitant to tighter packing in the trimerization region. The transition to the active DNA binding-competent state occurred highly cooperative and was concentration dependent. Surprisingly, Hsp90, known to inhibit Hsf1 activation, lowered the midpoint temperature of trimerization and reduced cooperativity of the process thus widening the response window. Based on our data we propose a kinetic model of Hsf1 trimerization.
Collapse
Affiliation(s)
- Nikolai Hentze
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Laura Le Breton
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Jan Wiesner
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Georg Kempf
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Matthias P Mayer
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
36
|
Neudegger T, Verghese J, Hayer-Hartl M, Hartl FU, Bracher A. Structure of human heat-shock transcription factor 1 in complex with DNA. Nat Struct Mol Biol 2016; 23:140-6. [PMID: 26727489 DOI: 10.1038/nsmb.3149] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023]
Abstract
Heat-shock transcription factor 1 (HSF1) has a central role in mediating the protective response to protein conformational stresses in eukaryotes. HSF1 consists of an N-terminal DNA-binding domain (DBD), a coiled-coil oligomerization domain, a regulatory domain and a transactivation domain. Upon stress, HSF1 trimerizes via its coiled-coil domain and binds to the promoters of heat shock protein-encoding genes. Here, we present cocrystal structures of the human HSF1 DBD in complex with cognate DNA. A comparative analysis of the HSF1 paralog Skn7 from Chaetomium thermophilum showed that single amino acid changes in the DBD can switch DNA binding specificity, thus revealing the structural basis for the interaction of HSF1 with cognate DNA. We used a crystal structure of the coiled-coil domain of C. thermophilum Skn7 to develop a model of the active human HSF1 trimer in which HSF1 embraces the heat-shock-element DNA.
Collapse
Affiliation(s)
- Tobias Neudegger
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jacob Verghese
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Manajit Hayer-Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
37
|
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: 75] [Impact Index Per Article: 8.3] [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
|
38
|
Pasqua T, Filice E, Mazza R, Quintieri AM, Carmela Cerra M, Iannacone R, Melfi D, Indiveri C, Gattuso A, Angelone T. Cardiac and hepatic role of r-AtHSP70: basal effects and protection against ischemic and sepsis conditions. J Cell Mol Med 2015; 19:1492-503. [PMID: 25904190 PMCID: PMC4511348 DOI: 10.1111/jcmm.12491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022] Open
Abstract
Heat shock proteins (HSPs), highly conserved in all organisms, act as molecular chaperones activated by several stresses. The HSP70 class of stress-induced proteins is the most studied subtype in cardiovascular and inflammatory disease. Because of the high similarity between plant and mammalian HSP70, the aim of this work was to evaluate whether recombinant HSP70 of plant origin (r-AtHSP70) was able to protect rat cardiac and hepatic function under ischemic and sepsis conditions. We demonstrated for the first time that, in ex vivo isolated and perfused rat heart, exogenous r-AtHSP70 exerted direct negative inotropic and lusitropic effects via Akt/endothelial nitric oxide synthase pathway, induced post-conditioning cardioprotection via Reperfusion Injury Salvage Kinase and Survivor Activating Factor Enhancement pathways, and did not cause hepatic damage. In vivo administration of r-AtHSP70 protected both heart and liver against lipopolysaccharide-dependent sepsis, as revealed by the reduced plasma levels of interleukin-1β, tumour necrosis factor alpha, aspartate aminotransferase and alanine aminotransferase. These results suggest exogenous r-AtHSP70 as a molecular modulator able to protect myocardial function and to prevent cardiac and liver dysfunctions during inflammatory conditions.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Elisabetta Filice
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Rosa Mazza
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Anna Maria Quintieri
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Maria Carmela Cerra
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy.,National Institute of Cardiovascular Research, Bologna, Italy
| | - Rina Iannacone
- ALSIA-Research Center Metapontum Agrobios, Metaponto (MT), Italy
| | - Donato Melfi
- ALSIA-Research Center Metapontum Agrobios, Metaponto (MT), Italy
| | - Cesare Indiveri
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Alfonsina Gattuso
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Tommaso Angelone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende (CS), Italy.,National Institute of Cardiovascular Research, Bologna, Italy
| |
Collapse
|
39
|
Abstract
Loss of protein homeostasis (proteostasis) is a common feature of aging and disease that is characterized by the appearance of nonnative protein aggregates in various tissues. Protein aggregation is routinely suppressed by the proteostasis network (PN), a collection of macromolecular machines that operate in diverse ways to maintain proteome integrity across subcellular compartments and between tissues to ensure a healthy life span. Here, we review the composition, function, and organizational properties of the PN in the context of individual cells and entire organisms and discuss the mechanisms by which disruption of the PN, and related stress response pathways, contributes to the initiation and progression of disease. We explore emerging evidence that disease susceptibility arises from early changes in the composition and activity of the PN and propose that a more complete understanding of the temporal and spatial properties of the PN will enhance our ability to develop effective treatments for protein conformational diseases.
Collapse
Affiliation(s)
- Johnathan Labbadia
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208;
| | | |
Collapse
|
40
|
Abstract
Heat shock proteins are molecular chaperones with a central role in protein folding and cellular protein homeostasis. They also play major roles in the development of cancer and in recent years have emerged as promising therapeutic targets. In this review, we discuss the known molecular mechanisms of various heat shock protein families and their involvement in cancer and in particular, multiple myeloma. In addition, we address the current progress and challenges in pharmacologically targeting these proteins as anti-cancer therapeutic strategies.
Collapse
|
41
|
Vera M, Pani B, Griffiths LA, Muchardt C, Abbott CM, Singer RH, Nudler E. The translation elongation factor eEF1A1 couples transcription to translation during heat shock response. eLife 2014; 3:e03164. [PMID: 25233275 PMCID: PMC4164936 DOI: 10.7554/elife.03164] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/14/2014] [Indexed: 01/26/2023] Open
Abstract
Translation elongation factor eEF1A has a well-defined role in protein synthesis. In this study, we demonstrate a new role for eEF1A: it participates in the entire process of the heat shock response (HSR) in mammalian cells from transcription through translation. Upon stress, isoform 1 of eEF1A rapidly activates transcription of HSP70 by recruiting the master regulator HSF1 to its promoter. eEF1A1 then associates with elongating RNA polymerase II and the 3'UTR of HSP70 mRNA, stabilizing it and facilitating its transport from the nucleus to active ribosomes. eEF1A1-depleted cells exhibit severely impaired HSR and compromised thermotolerance. In contrast, tissue-specific isoform 2 of eEF1A does not support HSR. By adjusting transcriptional yield to translational needs, eEF1A1 renders HSR rapid, robust, and highly selective; thus, representing an attractive therapeutic target for numerous conditions associated with disrupted protein homeostasis, ranging from neurodegeneration to cancer.
Collapse
Affiliation(s)
- Maria Vera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, United States
- Département de Biologie du Développement et Cellules Souches, Institut Pasteur, CNRS URA2578, Paris, France
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, United States
| | - Bibhusita Pani
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Lowri A Griffiths
- Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Christian Muchardt
- Département de Biologie du Développement et Cellules Souches, Institut Pasteur, CNRS URA2578, Paris, France
| | - Catherine M Abbott
- Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, United States
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, United States
- Howard Hughes Medical Institute, New York University School of Medicine, New York, United States
| |
Collapse
|
42
|
van Oosten-Hawle P, Morimoto RI. Transcellular chaperone signaling: an organismal strategy for integrated cell stress responses. ACTA ACUST UNITED AC 2014; 217:129-36. [PMID: 24353212 DOI: 10.1242/jeb.091249] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of each cell within a metazoan to adapt to and survive environmental and physiological stress requires cellular stress-response mechanisms, such as the heat shock response (HSR). Recent advances reveal that cellular proteostasis and stress responses in metazoans are regulated by multiple layers of intercellular communication. This ensures that an imbalance of proteostasis that occurs within any single tissue 'at risk' is protected by a compensatory activation of a stress response in adjacent tissues that confers a community protective response. While each cell expresses the machinery for heat shock (HS) gene expression, the HSR is regulated cell non-autonomously in multicellular organisms, by neuronal signaling to the somatic tissues, and by transcellular chaperone signaling between somatic tissues and from somatic tissues to neurons. These cell non-autonomous processes ensure that the organismal HSR is orchestrated across multiple tissues and that transmission of stress signals between tissues can also override the neuronal control to reset cell- and tissue-specific proteostasis. Here, we discuss emerging concepts and insights into the complex cell non-autonomous mechanisms that control stress responses in metazoans and highlight the importance of intercellular communication for proteostasis maintenance in multicellular organisms.
Collapse
Affiliation(s)
- Patricija van Oosten-Hawle
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | | |
Collapse
|
43
|
Rhoads RP, Baumgard LH, Suagee JK, Sanders SR. Nutritional interventions to alleviate the negative consequences of heat stress. Adv Nutr 2013; 4:267-76. [PMID: 23674792 PMCID: PMC3650495 DOI: 10.3945/an.112.003376] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Energy metabolism is a highly coordinated process, and preferred fuel(s) differ among tissues. The hierarchy of substrate use can be affected by physiological status and environmental factors including high ambient temperature. Unabated heat eventually overwhelms homeothermic mechanisms resulting in heat stress, which compromises animal health, farm animal production, and human performance. Various aspects of heat stress physiology have been extensively studied, yet a clear understanding of the metabolic changes occurring at the cellular, tissue, and whole-body levels in response to an environmental heat load remains ill-defined. For reasons not yet clarified, circulating nonesterified fatty acid levels are reduced during heat stress, even in the presence of elevated stress hormones (epinephrine, glucagon, and cortisol), and heat-stressed animals often have a blunted lipolytic response to catabolic signals. Either directly because of or in coordination with this, animals experiencing environmental hyperthermia exhibit a shift toward carbohydrate use. These metabolic alterations occur coincident with increased circulating basal and stimulated plasma insulin concentrations. Limited data indicate that proper insulin action is necessary to effectively mount a response to heat stress and minimize heat-induced damage. Consistent with this idea, nutritional interventions targeting increased insulin action may improve tolerance and productivity during heat stress. Further research is warranted to uncover the effects of heat on parameters associated with energy metabolism so that more appropriate and effective treatment methodologies can be designed.
Collapse
Affiliation(s)
- Robert P Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| | | | | | | |
Collapse
|
44
|
Desai S, Liu Z, Yao J, Patel N, Chen J, Wu Y, Ahn EEY, Fodstad O, Tan M. Heat shock factor 1 (HSF1) controls chemoresistance and autophagy through transcriptional regulation of autophagy-related protein 7 (ATG7). J Biol Chem 2013; 288:9165-76. [PMID: 23386620 PMCID: PMC3610989 DOI: 10.1074/jbc.m112.422071] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/04/2013] [Indexed: 01/07/2023] Open
Abstract
Heat shock factor 1 (HSF1), a master regulator of heat shock responses, plays an important role in tumorigenesis. In this study we demonstrated that HSF1 is required for chemotherapeutic agent-induced cytoprotective autophagy through transcriptional up-regulation of autophagy-related gene ATG7. Interestingly, this is independent of the HSF1 heat shock response function. Treatment of cancer cells with the FDA-approved chemotherapeutic agent carboplatin induced autophagy and growth inhibition, which were significantly increased upon knockdown of HSF1. Mechanistic studies revealed that HSF1 regulates autophagy by directly binding to ATG7 promoter and transcriptionally up-regulating its expression. Significantly, breast cancer patient sample study revealed that a higher ATG7 expression level is associated with poor patient survival. This novel finding was further confirmed by analysis of two independent patient databases, demonstrating a prognostic value of ATG7. Furthermore, a strong positive correlation was observed between levels of HSF1 and ATG7 in triple-negative breast cancer patient samples, thus validating our in vitro findings. This is the first study identifying a critical role for HSF1 in controlling cytoprotective autophagy through regulation of ATG7, which is distinct from the HSF1 function in the heat shock response. This is also the first study demonstrating a prognostic value of ATG7 in breast cancer patients. These findings strongly argue that combining chemotherapeutic agents with autophagy inhibition by repressing HSF1/ATG7 axis represents a promising strategy for future cancer treatment.
Collapse
Affiliation(s)
- Shruti Desai
- From the Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604
| | - Zixing Liu
- From the Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604
| | - Jun Yao
- the Department of Neuro-oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Nishant Patel
- the Department of Mathematics, Northwest Florida State College, Niceville, Florida 32578
| | - Jieqing Chen
- the Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Yun Wu
- the Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Erin Eun-Young Ahn
- From the Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604
| | - Oystein Fodstad
- the Department of Tumor Biology, Norwegian Radium Hospital, University of Oslo, 0310 Oslo, Norway, and
| | - Ming Tan
- From the Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604
- the Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama 36604
| |
Collapse
|
45
|
Non-specific protein modifications by a phytochemical induce heat shock response for self-defense. PLoS One 2013; 8:e58641. [PMID: 23536805 PMCID: PMC3594166 DOI: 10.1371/journal.pone.0058641] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 02/05/2013] [Indexed: 01/02/2023] Open
Abstract
Accumulated evidence shows that some phytochemicals provide beneficial effects for human health. Recently, a number of mechanistic studies have revealed that direct interactions between phytochemicals and functional proteins play significant roles in exhibiting their bioactivities. However, their binding selectivities to biological molecules are considered to be lower due to their small and simple structures. In this study, we found that zerumbone, a bioactive sesquiterpene, binds to numerous proteins with little selectivity. Similar to heat-denatured proteins, zerumbone-modified proteins were recognized by heat shock protein 90, a constitutive molecular chaperone, leading to heat shock factor 1-dependent heat shock protein induction in hepa1c1c7 mouse hepatoma cells. Furthermore, oral administration of this phytochemical up-regulated heat shock protein expressions in the livers of Sprague-Dawley rats. Interestingly, pretreatment with zerumbone conferred a thermoresistant phenotype to hepa1c1c7 cells as well as to the nematode Caenorhabditis elegans. It is also important to note that several phytochemicals with higher hydrophobicity or electrophilicity, including phenethyl isothiocyanate and curcumin, markedly induced heat shock proteins, whereas most of the tested nutrients did not. These results suggest that non-specific protein modifications by xenobiotic phytochemicals cause mild proteostress, thereby inducing heat shock response and leading to potentiation of protein quality control systems. We considered these bioactivities to be xenohormesis, an adaptation mechanism against xenobiotic chemical stresses. Heat shock response by phytochemicals may be a fundamental mechanism underlying their various bioactivities.
Collapse
|
46
|
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: 6.8] [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
|
47
|
Dai B, Gong A, Jing Z, Aldape KD, Kang SH, Sawaya R, Huang S. Forkhead box M1 is regulated by heat shock factor 1 and promotes glioma cells survival under heat shock stress. J Biol Chem 2012. [PMID: 23192351 DOI: 10.1074/jbc.m112.379362] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The forkhead box M1 (FoxM1) is a key transcription factor regulating multiple aspects of cell biology. Prior studies have shown that FoxM1 is overexpressed in a variety of human tumors, including brain tumor, and plays a critical role in cancer development and progression. In this study we found that FoxM1 was up-regulated by heat shock factor 1 (HSF1) under heat shock stress condition in multiple cell lines. Knockdown of HSF1 with HSF1 siRNA or inhibition of HSF1 with a HSF1 inhibitor abrogated heat shock-induced expression of FoxM1. Genetic deletion of HSF1 in mouse embryo fibroblast cells also abolished heat shock stress-induced FoxM1 expression. Moreover, we showed that HSF1 directly bound to FoxM1 promoter and increased FoxM1 promoter activity. Furthermore, we demonstrated that FoxM1 was required for the G(2)-M phase progression through regulating Cdc2, Cdc20, and Cdc25B under a mild heat shock stress but enhanced cell survival under lethal heat shock stress condition. Finally, in human glioblastoma specimens, FoxM1 overexpression correlated with elevated HSF1 expression. Our results indicate that FoxM1 is regulated by HSF1 and is critical for HSF1-mediated heat shock response. We demonstrated a novel mechanism of stress resistance controlled by HSF1 and a new HSF-FoxM1 connection that mediates cellular thermotolerance.
Collapse
Affiliation(s)
- Bingbing Dai
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Kloster-Landsberg M, Herbomel G, Wang I, Derouard J, Vourc'h C, Usson Y, Souchier C, Delon A. Cellular response to heat shock studied by multiconfocal fluorescence correlation spectroscopy. Biophys J 2012; 103:1110-9. [PMID: 22995483 PMCID: PMC3446677 DOI: 10.1016/j.bpj.2012.07.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 07/21/2012] [Accepted: 07/27/2012] [Indexed: 10/27/2022] Open
Abstract
Heat shock triggers a transient and ubiquitous response, the function of which is to protect cells against stress-induced damage. The heat-shock response is controlled by a key transcription factor known as heat shock factor 1 (HSF1). We have developed a multiconfocal fluorescence correlation spectroscopy setup to measure the dynamics of HSF1 during the course of the heat-shock response. The system combines a spatial light modulator, to address several points of interest, and an electron-multiplying charge-coupled camera for fast multiconfocal recording of the photon streams. Autocorrelation curves with a temporal resolution of 14 μs were analyzed before and after heat shock on eGFP and HSF1-eGFP-expressing cells. Evaluation of the dynamic parameters of a diffusion-and-binding model showed a slower HSF1 diffusion after heat shock. It is also observed that the dissociation rate decreases after heat shock, whereas the association rate is not affected. In addition, thanks to the multiconfocal fluorescence correlation spectroscopy system, up to five spots could be simultaneously located in each cell nucleus. This made it possible to quantify the intracellular variability of the diffusion constant of HSF1, which is higher than that of inert eGFP molecules and increases after heat shock. This finding is consistent with the fact that heat-shock response is associated with an increase of HSF1 interactions with DNA and cannot be explained even partially by heat-induced modifications of nuclear organization.
Collapse
Affiliation(s)
- Meike Kloster-Landsberg
- University of Grenoble I/Centre National de la Recherche Scientifique, Laboratoire Interdisciplinaire de Physique, Grenoble, France
| | - Gaëtan Herbomel
- University of Grenoble I/Institut National de la Santé et de la Recherche Médicale, Institut Albert Bonniot, U823 team 10, Grenoble, France
| | - Irène Wang
- University of Grenoble I/Centre National de la Recherche Scientifique, Laboratoire Interdisciplinaire de Physique, Grenoble, France
| | - Jacques Derouard
- University of Grenoble I/Centre National de la Recherche Scientifique, Laboratoire Interdisciplinaire de Physique, Grenoble, France
| | - Claire Vourc'h
- University of Grenoble I/Institut National de la Santé et de la Recherche Médicale, Institut Albert Bonniot, U823 team 10, Grenoble, France
| | - Yves Usson
- University of Grenoble I/Centre National de la Recherche Scientifique, Laboratoire TIMC-IMAG, Grenoble, France
| | - Catherine Souchier
- University of Grenoble I/Institut National de la Santé et de la Recherche Médicale, Institut Albert Bonniot, U823 team 10, Grenoble, France
| | - Antoine Delon
- University of Grenoble I/Centre National de la Recherche Scientifique, Laboratoire Interdisciplinaire de Physique, Grenoble, France
| |
Collapse
|
49
|
Sriram K, Rodriguez-Fernandez M, Doyle FJ. A detailed modular analysis of heat-shock protein dynamics under acute and chronic stress and its implication in anxiety disorders. PLoS One 2012; 7:e42958. [PMID: 22937003 PMCID: PMC3425570 DOI: 10.1371/journal.pone.0042958] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 07/16/2012] [Indexed: 01/01/2023] Open
Abstract
Physiological and psychological stresses cause anxiety disorders such as depression and post-traumatic stress disorder (PTSD) and induce drastic changes at a molecular level in the brain. To counteract this stress, the heat-shock protein (HSP) network plays a vital role in restoring the homeostasis of the system. To study the stress-induced dynamics of heat-shock network, we analyzed three modules of the HSP90 network—namely trimerization reactions, phosphorylation–dephosphorylation reactions, and the conversion of HSP90 from an open to a closed conformation—and constructed a corresponding nonlinear differential equation model based on mass action kinetics laws. The kinetic parameters of the model were obtained through global optimization, and sensitivity analyses revealed that the most sensitive parameters are the kinase and phosphatase that drive the phosphorylation–dephosphorylation reactions. Bifurcation analysis carried out with the estimated kinetic parameters of the model with stress as bifurcation parameter revealed the occurrence of “mushroom”, a type of complex dynamics in which S-shaped and Z-shaped hysteretic bistable forms are present together. We mapped the molecular events responsible for generating the mushroom dynamics under stress and interpreted the occurrence of the S-shaped hysteresis to a normal level of stress, and the Z-shaped hysteresis to the HSP90 variations under acute and chronic stress in the fear conditioned system, and further, we hypothesized that this can be extended to stress-related disorders such as depression and PTSD in humans. Finally, we studied the effect of parameter variations on the mushroom dynamics to get insight about the role of phosphorylation–dephosphorylation parameters in HSP90 network in bringing about complex dynamics such as isolas, where the stable steady states in a bistable system are isolated and separated from each other and not connected by an unstable steady state.
Collapse
Affiliation(s)
- K. Sriram
- Institute of Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California, United States of America
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
- Indraprastha Institute of Information Technology (IIIT), Delhi, India
| | - Maria Rodriguez-Fernandez
- Institute of Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California, United States of America
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Francis J. Doyle
- Institute of Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California, United States of America
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
- * E-mail:
| |
Collapse
|
50
|
Ullery JC, Marnett LJ. Protein modification by oxidized phospholipids and hydrolytically released lipid electrophiles: Investigating cellular responses. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2424-35. [PMID: 22562025 DOI: 10.1016/j.bbamem.2012.04.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/05/2012] [Accepted: 04/17/2012] [Indexed: 12/17/2022]
Abstract
Oxygen is essential for the growth and function of mammalian cells. However, imbalances in oxygen or abnormalities in the ability of a cell to respond to oxygen levels can result in oxidative stress. Oxidative stress plays an important role in a number of diseases including atherosclerosis, rheumatoid arthritis, cancer, neurodegenerative diseases and asthma. When membrane lipids are exposed to high levels of oxygen or derived oxidants, they undergo lipid peroxidation to generate oxidized phospholipids (oxPL). Continual exposure to oxidants and decomposition of oxPL results in the formation of reactive electrophiles, such as 4-hydroxy-2-nonenal (HNE). Reactive lipid electrophiles have been shown to covalently modify DNA and proteins. Furthermore, exposure of cells to lipid electrophiles results in the activation of cytoprotective signaling pathways in order to promote cell survival and recovery from oxidant stress. However, if not properly managed by cellular detoxification mechanisms, the continual exposure of cells to electrophiles results in cytotoxicity. The following perspective will discuss the biological importance of lipid electrophile protein adducts including current strategies employed to identify and isolate protein adducts of lipid electrophiles as well as approaches to define cellular signaling mechanisms altered upon exposure to electrophiles. This article is part of a Special Issue entitled: Oxidized phospholipids-their properties and interactions with proteins.
Collapse
Affiliation(s)
- Jody C Ullery
- Department of Biochemistry, Vanderbilt Institute of Chemical Biology, Nashville, TN, USA
| | | |
Collapse
|