1
|
Zhong Q, Wang X, Wei R, Liu F, Alamin M, Sun J, Gui L. Equisetin inhibits adiposity through AMPK-dependent regulation of brown adipocyte differentiation. Heliyon 2024; 10:e25458. [PMID: 38327434 PMCID: PMC10847917 DOI: 10.1016/j.heliyon.2024.e25458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
Obesity has a significant impact on endocrine function, which leads to metabolic diseases including diabetes, insulin resistance, and other complications associated with obesity. Development of effective and safe anti-obesity drugs is imperative and necessary. Equisetin (EQST), a tetramate-containing marine fungal product, was reported to inhibit bacterial fatty acid synthesis and affect mitochondrial metabolism. It is tempting to speculate that EQST might have anti-obesity effects. This study was designed to explore anti-obesity effects and underlying mechanism of EQST on 3T3-L1 adipocytes differentiated from 3T3-L1 cells. Oil Red O staining showed that EQST reduced lipid accumulation in 3T3-L1 adipocytes. Quantitative real-time polymerase chain reaction and Western blot analysis revealed that EQST significantly inhibited expression of adipogenesis/lipogenesis-related genes C/ebp-α, Ppar-γ, Srebp1c, Fas, and reduced protein levels. There was also increased expression of key genes and protein levels involved in lipolysis (Perilipin, Atgl, Hsl), brown adipocyte differentiation (Prdm16, Ucp1), mitochondrial biogenesis (Pgc1α, Tfam) and β-oxidation Acsl1, Cpt1. Moreover, mitochondrial content, their membrane potential ΔΨM, and respiratory chain genes Mt-Co1, Cox7a1, Cox8b, and Cox4 (and protein) exhibited marked increase in expression upon EQST treatment, along with increased protein levels. Importantly, EQST induced expression and activation of AMPK, which was compromised by the AMPK inhibitor dorsomorphin, leading to rescue of EQST-downregulated Fas expression and a reduction of the EQST-increased expression of Pgc1α, Ucp1, and Cox4. Together, EQST robustly promotes fat clearance through the AMPK pathway, these results supporting EQST as a strong candidate for the development into an anti-obesity therapeutic agent.
Collapse
Affiliation(s)
- Qin Zhong
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Clinical Medical Research Center, Affiliated Hospital of Guizhou Medical University No.28 Beijing Road, Guiyang City, Guizhou Province 550001, China
| | - Xian Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
| | - Ruiran Wei
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Department of Basic Medical Sciences, Clinical College of Anhui Medical University, No.69 Meishan Road Hefei City, Anhui Province 230031, China
| | - Fang Liu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
| | - Md Alamin
- Department of Biology, College of Life Sciences, Southern Medical University of Science and Technology, No.1088 Xueyuan Road, Shenzhen City, Guangdong Province 518055, China
| | - Jiajia Sun
- Institute of Obstetrics and Gynecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, No.1120 Lianhua Road, Futian District, Shenzhen City, Guangdong Province 518000, China
| | - Liming Gui
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Institute of Obstetrics and Gynecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, No.1120 Lianhua Road, Futian District, Shenzhen City, Guangdong Province 518000, China
| |
Collapse
|
2
|
Kapuganti RS, Alone DP. Current understanding of genetics and epigenetics in pseudoexfoliation syndrome and glaucoma. Mol Aspects Med 2023; 94:101214. [PMID: 37729850 DOI: 10.1016/j.mam.2023.101214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Pseudoexfoliation is a complex, progressive, and systemic age-related disorder. The early stage of deposition of extracellular fibrillar material on ocular and extraocular tissues is termed as pseudoexfoliation syndrome (PEXS). The severe advanced stage is known as pseudoexfoliation glaucoma (PEXG), which involves increased intraocular pressure and optic nerve damage. Through genome-wide association and candidate gene studies, PEX has been associated with numerous genetic risk variants in various gene loci. However, the genetic basis of the disease fails to explain certain features of PEX pathology, such as the progressive nature of the disease, asymmetric ocular manifestation, age-related onset, and only a subset of PEXS individuals developing PEXG. Increasing evidence shows an interplay of genetic and epigenetic factors in the pathology of complex, multifactorial diseases. In this review, we have discussed the genetic basis of the disease and the emerging contribution of epigenetic regulations in PEX pathogenesis, focusing on DNA methylation and non-coding RNAs. Aberrant methylation patterns, histone modifications, and post-transcriptional regulation by microRNAs lead to aberrant gene expression changes. We have reviewed these aberrant epigenetic changes in PEX pathology and their effect on molecular pathways associated with PEX. We have further discussed some possible genetic/epigenetic-based diagnoses and therapeutics for PEX. Although studies to understand the role of epigenetic regulations in PEX are just emerging, epigenetic modifications contribute significantly to PEX pathogenesis and may pave the way for better and targeted therapeutics.
Collapse
Affiliation(s)
- Ramani Shyam Kapuganti
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Debasmita Pankaj Alone
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
3
|
Liu Y, Wang J, Liu B, Xu ZY. Dynamic regulation of DNA methylation and histone modifications in response to abiotic stresses in plants. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2022; 64:2252-2274. [PMID: 36149776 DOI: 10.1111/jipb.13368] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
DNA methylation and histone modification are evolutionarily conserved epigenetic modifications that are crucial for the expression regulation of abiotic stress-responsive genes in plants. Dynamic changes in gene expression levels can result from changes in DNA methylation and histone modifications. In the last two decades, how epigenetic machinery regulates abiotic stress responses in plants has been extensively studied. Here, based on recent publications, we review how DNA methylation and histone modifications impact gene expression regulation in response to abiotic stresses such as drought, abscisic acid, high salt, extreme temperature, nutrient deficiency or toxicity, and ultraviolet B exposure. We also review the roles of epigenetic mechanisms in the formation of transgenerational stress memory. We posit that a better understanding of the epigenetic underpinnings of abiotic stress responses in plants may facilitate the design of more stress-resistant or -resilient crops, which is essential for coping with global warming and extreme environments.
Collapse
Affiliation(s)
- Yutong Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Jie Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Bao Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Zheng-Yi Xu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
4
|
Combined Assay of rDNA and SatIII Copy Numbers as an Individual Profile of Stress Resistance, Longevity, Fertility and Disease Predisposition. J Pers Med 2022; 12:jpm12101752. [PMID: 36294891 PMCID: PMC9604575 DOI: 10.3390/jpm12101752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
The ribosomal DNA and pericentromeric satellite repeats are two important types of moderately repeated sequences existing in the human genome. They are functionally involved in the universal stress response. There is accumulating evidence that the copy number variation (CNV) of the repeat units is a novel factor modulating the stress response and, thus, has phenotypic manifestations. The ribosomal repeat copy number plays a role in stress resistance, lifespan, in vitro fertilization chances, disease progression and aging, while the dynamics of the satellite copy number are a sort of indicator of the current stress state. Here, we review some facts showing that a combined assay of rDNA and SatII/III abundance can provide valuable individual data ("stress profile") indicating not only the inherited adaptive reserve but also the stress duration and acute or chronic character of the stress. Thus, the repeat count could have applications in personalized medicine in the future.
Collapse
|
5
|
Vourc’h C, Dufour S, Timcheva K, Seigneurin-Berny D, Verdel A. HSF1-Activated Non-Coding Stress Response: Satellite lncRNAs and Beyond, an Emerging Story with a Complex Scenario. Genes (Basel) 2022; 13:genes13040597. [PMID: 35456403 PMCID: PMC9032817 DOI: 10.3390/genes13040597] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/21/2022] Open
Abstract
In eukaryotes, the heat shock response is orchestrated by a transcription factor named Heat Shock Factor 1 (HSF1). HSF1 is mostly characterized for its role in activating the expression of a repertoire of protein-coding genes, including the heat shock protein (HSP) genes. Remarkably, a growing set of reports indicate that, upon heat shock, HSF1 also targets various non-coding regions of the genome. Focusing primarily on mammals, this review aims at reporting the identity of the non-coding genomic sites directly bound by HSF1, and at describing the molecular function of the long non-coding RNAs (lncRNAs) produced in response to HSF1 binding. The described non-coding genomic targets of HSF1 are pericentric Satellite DNA repeats, (sub)telomeric DNA repeats, Short Interspersed Nuclear Element (SINE) repeats, transcriptionally active enhancers and the NEAT1 gene. This diverse set of non-coding genomic sites, which already appears to be an integral part of the cellular response to stress, may only represent the first of many. Thus, the study of the evolutionary conserved heat stress response has the potential to emerge as a powerful cellular context to study lncRNAs, produced from repeated or unique DNA regions, with a regulatory function that is often well-documented but a mode of action that remains largely unknown.
Collapse
Affiliation(s)
- Claire Vourc’h
- Université de Grenoble Alpes (UGA), 38700 La Tronche, France
- Correspondence: (C.V.); (A.V.)
| | - Solenne Dufour
- Institute for Advanced Biosciences (IAB), Centre de Recherche UGA/Inserm U 1209/CNRS UMR 5309, Site Santé-Allée des Alpes, 38700 La Tronche, France; (S.D.); (K.T.); (D.S.-B.)
| | - Kalina Timcheva
- Institute for Advanced Biosciences (IAB), Centre de Recherche UGA/Inserm U 1209/CNRS UMR 5309, Site Santé-Allée des Alpes, 38700 La Tronche, France; (S.D.); (K.T.); (D.S.-B.)
| | - Daphné Seigneurin-Berny
- Institute for Advanced Biosciences (IAB), Centre de Recherche UGA/Inserm U 1209/CNRS UMR 5309, Site Santé-Allée des Alpes, 38700 La Tronche, France; (S.D.); (K.T.); (D.S.-B.)
| | - André Verdel
- Institute for Advanced Biosciences (IAB), Centre de Recherche UGA/Inserm U 1209/CNRS UMR 5309, Site Santé-Allée des Alpes, 38700 La Tronche, France; (S.D.); (K.T.); (D.S.-B.)
- Correspondence: (C.V.); (A.V.)
| |
Collapse
|
6
|
The Role of Human Satellite III (1q12) Copy Number Variation in the Adaptive Response during Aging, Stress, and Pathology: A Pendulum Model. Genes (Basel) 2021; 12:genes12101524. [PMID: 34680920 PMCID: PMC8535310 DOI: 10.3390/genes12101524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/18/2022] Open
Abstract
The pericentric satellite III (SatIII or Sat3) and II tandem repeats recently appeared to be transcribed under stress conditions, and the transcripts were shown to play an essential role in the universal stress response. In this paper, we review the role of human-specific SatIII copy number variation (CNV) in normal stress response, aging and pathology, with a focus on 1q12 loci. We postulate a close link between transcription of SatII/III repeats and their CNV. The accrued body of data suggests a hypothetical universal mechanism, which provides for SatIII copy gain during the stress response, alongside with another, more hypothetical reverse mechanism that might reduce the mean SatIII copy number, likely via the selection of cells with excessively large 1q12 loci. Both mechanisms, working alternatively like swings of the pendulum, may ensure the balance of SatIII copy numbers and optimum stress resistance. This model is verified on the most recent data on SatIII CNV in pathology and therapy, aging, senescence and response to genotoxic stress in vitro.
Collapse
|
7
|
Livernois AM, Mallard BA, Cartwright SL, Cánovas A. Heat stress and immune response phenotype affect DNA methylation in blood mononuclear cells from Holstein dairy cows. Sci Rep 2021; 11:11371. [PMID: 34059695 PMCID: PMC8166884 DOI: 10.1038/s41598-021-89951-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/29/2021] [Indexed: 11/08/2022] Open
Abstract
Heat stress negatively affects health and production in cows. Examining the cellular response to heat stress could reveal underlying protective molecular mechanisms associated with superior resilience and ultimately enable selection for more resilient cattle. This type of investigation is increasingly important as future predictions for the patterns of heat waves point to increases in frequency, severity, and duration. Cows identified as high immune responders based on High Immune Response technology (HIR) have lower disease occurrence compared to their average and low immune responder herd-mates. In this study, our goal was to identify epigenetic differences between high and low immune responder cows in response to heat stress. We examined genome-wide DNA methylation of blood mononuclear cells (BMCs) isolated from high and low cows, before and after in vitro heat stress. We identified differential methylation of promoter regions associated with a variety of biological processes including immune function, stress response, apoptosis, and cell signalling. The specific differentially methylated promoter regions differed between samples from high and low cows, and results revealed pathways associated with cellular protection during heat stress.
Collapse
Affiliation(s)
- A M Livernois
- Deptartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada.
| | - B A Mallard
- Deptartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - S L Cartwright
- Deptartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - A Cánovas
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
8
|
Prince TL, Lang BJ, Guerrero-Gimenez ME, Fernandez-Muñoz JM, Ackerman A, Calderwood SK. HSF1: Primary Factor in Molecular Chaperone Expression and a Major Contributor to Cancer Morbidity. Cells 2020; 9:E1046. [PMID: 32331382 PMCID: PMC7226471 DOI: 10.3390/cells9041046] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023] Open
Abstract
Heat shock factor 1 (HSF1) is the primary component for initiation of the powerful heat shock response (HSR) in eukaryotes. The HSR is an evolutionarily conserved mechanism for responding to proteotoxic stress and involves the rapid expression of heat shock protein (HSP) molecular chaperones that promote cell viability by facilitating proteostasis. HSF1 activity is amplified in many tumor contexts in a manner that resembles a chronic state of stress, characterized by high levels of HSP gene expression as well as HSF1-mediated non-HSP gene regulation. HSF1 and its gene targets are essential for tumorigenesis across several experimental tumor models, and facilitate metastatic and resistant properties within cancer cells. Recent studies have suggested the significant potential of HSF1 as a therapeutic target and have motivated research efforts to understand the mechanisms of HSF1 regulation and develop methods for pharmacological intervention. We review what is currently known regarding the contribution of HSF1 activity to cancer pathology, its regulation and expression across human cancers, and strategies to target HSF1 for cancer therapy.
Collapse
Affiliation(s)
- Thomas L. Prince
- Department of Molecular Functional Genomics, Geisinger Clinic, Danville, PA 17821, USA
| | - Benjamin J. Lang
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Martin E. Guerrero-Gimenez
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Buenos Aires B1657, Argentina
| | - Juan Manuel Fernandez-Muñoz
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Buenos Aires B1657, Argentina
| | - Andrew Ackerman
- Department of Molecular Functional Genomics, Geisinger Clinic, Danville, PA 17821, USA
| | - Stuart K. Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Nuño-Cabanes C, Ugidos M, Tarazona S, Martín-Expósito M, Ferrer A, Rodríguez-Navarro S, Conesa A. A multi-omics dataset of heat-shock response in the yeast RNA binding protein Mip6. Sci Data 2020; 7:69. [PMID: 32109230 PMCID: PMC7046740 DOI: 10.1038/s41597-020-0412-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/10/2020] [Indexed: 11/25/2022] Open
Abstract
Gene expression is a biological process regulated at different molecular levels, including chromatin accessibility, transcription, and RNA maturation and transport. In addition, these regulatory mechanisms have strong links with cellular metabolism. Here we present a multi-omics dataset that captures different aspects of this multi-layered process in yeast. We obtained RNA-seq, metabolomics, and H4K12ac ChIP-seq data for wild-type and mip6Δ strains during a heat-shock time course. Mip6 is an RNA-binding protein that contributes to RNA export during environmental stress and is informative of the contribution of post-transcriptional regulation to control cellular adaptations to environmental changes. The experiment was performed in quadruplicate, and the different omics measurements were obtained from the same biological samples, which facilitates the integration and analysis of data using covariance-based methods. We validate our dataset by showing that ChIP-seq, RNA-seq and metabolomics signals recapitulate existing knowledge about the response of ribosomal genes and the contribution of trehalose metabolism to heat stress. Raw data, processed data and preprocessing scripts are made available.
Collapse
Grants
- PROMETEO/2016/093 Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
- PROMETEO/2016/093 Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
- PROMETEO/2016/093 Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
- PROMETEO/2016/093 Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
- PROMETEO/2016/093 Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
- Regional Government of Valencia | Conselleria d'Educació, Investigació, Cultura i Esport (Conselleria d'Educació, Investigació, Cultura i Esport de la Generalitat Valenciana)
Collapse
Affiliation(s)
- Carme Nuño-Cabanes
- Gene Expression and RNA Metabolism Laboratory, Instituto de Biomedicina de Valencia (CSIC). Jaume Roig, 11, E-46010, Valencia, Spain
- Gene Expression and RNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yúfera, E-46012, Valencia, Spain
| | - Manuel Ugidos
- Gene Expression and RNA Metabolism Laboratory, Instituto de Biomedicina de Valencia (CSIC). Jaume Roig, 11, E-46010, Valencia, Spain
- Gene Expression and RNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yúfera, E-46012, Valencia, Spain
| | - Sonia Tarazona
- Department of Applied Statistics, Operations Research and Quality, Universitat Politècnica de València (UPV), Valencia, Spain
| | - Manuel Martín-Expósito
- Gene Expression and RNA Metabolism Laboratory, Instituto de Biomedicina de Valencia (CSIC). Jaume Roig, 11, E-46010, Valencia, Spain
| | - Alberto Ferrer
- Department of Applied Statistics, Operations Research and Quality, Universitat Politècnica de València (UPV), Valencia, Spain
| | - Susana Rodríguez-Navarro
- Gene Expression and RNA Metabolism Laboratory, Instituto de Biomedicina de Valencia (CSIC). Jaume Roig, 11, E-46010, Valencia, Spain.
- Gene Expression and RNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yúfera, E-46012, Valencia, Spain.
| | - Ana Conesa
- Microbiology and Cell Science Department, University of Florida, Gainesville, Florida, USA.
- Institute for Food and Agricultural Reserach, University of Florida, Gainesville, Florida, USA.
- Genetics Institute, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
10
|
Kovács D, Sigmond T, Hotzi B, Bohár B, Fazekas D, Deák V, Vellai T, Barna J. HSF1Base: A Comprehensive Database of HSF1 (Heat Shock Factor 1) Target Genes. Int J Mol Sci 2019; 20:ijms20225815. [PMID: 31752429 PMCID: PMC6888953 DOI: 10.3390/ijms20225815] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022] Open
Abstract
HSF1 (heat shock factor 1) is an evolutionarily conserved master transcriptional regulator of the heat shock response (HSR) in eukaryotic cells. In response to high temperatures, HSF1 upregulates genes encoding molecular chaperones, also called heat shock proteins, which assist the refolding or degradation of damaged intracellular proteins. Accumulating evidence reveals however that HSF1 participates in several other physiological and pathological processes such as differentiation, immune response, and multidrug resistance, as well as in ageing, neurodegenerative demise, and cancer. To address how HSF1 controls these processes one should systematically analyze its target genes. Here we present a novel database called HSF1Base (hsf1base.org) that contains a nearly comprehensive list of HSF1 target genes identified so far. The list was obtained by manually curating publications on individual HSF1 targets and analyzing relevant high throughput transcriptomic and chromatin immunoprecipitation data derived from the literature and the Yeastract database. To support the biological relevance of HSF1 targets identified by high throughput methods, we performed an enrichment analysis of (potential) HSF1 targets across different tissues/cell types and organisms. We found that general HSF1 functions (targets are expressed in all tissues/cell types) are mostly related to cellular proteostasis. Furthermore, HSF1 targets that are conserved across various animal taxa operate mostly in cellular stress pathways (e.g., autophagy), chromatin remodeling, ribosome biogenesis, and ageing. Together, these data highlight diverse roles for HSF1, expanding far beyond the HSR.
Collapse
Affiliation(s)
- Dániel Kovács
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
| | - Tímea Sigmond
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
| | - Bernadette Hotzi
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
| | - Balázs Bohár
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
- Earlham Institute, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich NR4 7UA, UK
| | - Dávid Fazekas
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
- Earlham Institute, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich NR4 7UA, UK
| | - Veronika Deák
- Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, University of Technology, H-1111 Budapest, Hungary;
| | - Tibor Vellai
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, H-1117 Budapest, Hungary
- Correspondence: (T.V.); (J.B.); Tel.: +36-1-372-2500 (ext. 8684) (T.V.); +36-1-372-2500 (ext. 8349) (J.B.); Fax: +36-1-372-2641 (T.V.)
| | - János Barna
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary; (D.K.); (T.S.); (B.H.); (B.B.); (D.F.)
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, H-1117 Budapest, Hungary
- Correspondence: (T.V.); (J.B.); Tel.: +36-1-372-2500 (ext. 8684) (T.V.); +36-1-372-2500 (ext. 8349) (J.B.); Fax: +36-1-372-2641 (T.V.)
| |
Collapse
|
11
|
Mahady L, Nadeem M, Malek-Ahmadi M, Chen K, Perez SE, Mufson EJ. Frontal Cortex Epigenetic Dysregulation During the Progression of Alzheimer's Disease. J Alzheimers Dis 2019; 62:115-131. [PMID: 29439356 DOI: 10.3233/jad-171032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although the frontal cortex plays an important role in cognitive function and undergoes neuronal dysfunction in Alzheimer's disease (AD), the factors driving these cellular alterations remain unknown. Recent studies suggest that alterations in epigenetic regulation play a pivotal role in this process in AD. We evaluated frontal cortex histone deacetylase (HDAC) and sirtuin (SIRT) levels in tissue obtained from subjects with a premortem diagnosis of no-cognitive impairment (NCI), mild cognitive impairment (MCI), mild to moderate AD (mAD), and severe AD (sAD) using quantitative western blotting. Immunoblots revealed significant increases in HDAC1 and HDAC3 in MCI and mAD, followed by a decrease in sAD compared to NCI. HDAC2 levels remained stable across clinical groups. HDAC4 was significantly increased in MCI and mAD, but not in sAD compared to NCI. HDAC6 significantly increased during disease progression, while SIRT1 decreased in MCI, mAD, and sAD compared to NCI. HDAC1 levels negatively correlated with perceptual speed, while SIRT1 positively correlated with perceptual speed, episodic memory, global cognitive score, and Mini-Mental State Examination. HDAC1 positively, while SIRT1 negatively correlated with cortical neurofibrillary tangle counts. These findings suggest that dysregulation of epigenetic proteins contribute to neuronal dysfunction and cognitive decline in the early stage of AD.
Collapse
Affiliation(s)
- Laura Mahady
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA.,Arizona State University Interdisciplinary Graduate Program in Neuroscience, Tempe, AZ, USA
| | - Muhammad Nadeem
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
12
|
Fog CK, Zago P, Malini E, Solanko LM, Peruzzo P, Bornaes C, Magnoni R, Mehmedbasic A, Petersen NHT, Bembi B, Aerts JFMG, Dardis A, Kirkegaard T. The heat shock protein amplifier arimoclomol improves refolding, maturation and lysosomal activity of glucocerebrosidase. EBioMedicine 2018; 38:142-153. [PMID: 30497978 PMCID: PMC6306395 DOI: 10.1016/j.ebiom.2018.11.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Gaucher Disease is caused by mutations of the GBA gene which encodes the lysosomal enzyme acid beta-glucosidase (GCase). GBA mutations commonly affect GCase function by perturbing its protein homeostasis rather than its catalytic activity. Heat shock proteins are well known cytoprotective molecules with functions in protein homeostasis and lysosomal function and their manipulation has been suggested as a potential therapeutic strategy for GD. The investigational drug arimoclomol, which is in phase II/III clinical trials, is a well-characterized HSP amplifier and has been extensively clinically tested. Importantly, arimoclomol efficiently crosses the blood-brain-barrier presenting an opportunity to target the neurological manifestations of GD, which remains without a disease-modifying therapy. METHODS We used a range of biological and biochemical in vitro assays to assess the effect of arimoclomol on GCase activity in ex vivo systems of primary fibroblasts and neuronal-like cells from GD patients. FINDINGS We found that arimoclomol induced relevant HSPs such as ER-resident HSP70 (BiP) and enhanced the folding, maturation, activity, and correct cellular localization of mutated GCase across several genotypes including the common L444P and N370S mutations in primary cells from GD patients. These effects where recapitulated in a human neuronal model of GD obtained by differentiation of multipotent adult stem cells. INTERPRETATION These data demonstrate the potential of HSP-targeting therapies in GCase-deficiencies and strongly support the clinical development of arimoclomol as a potential therapeutic option for the neuronopathic forms of GD. FUNDING The research was funded by Orphazyme A/S, Copenhagen, Denmark.
Collapse
Affiliation(s)
- Cathrine K Fog
- Orphazyme A/S, Ole Maaloes vej 3, DK-2200 Copenhagen, Denmark
| | - Paola Zago
- Regional Coordinator Centre for Rare Diseases, Academic Hospital "Santa Maria della Misericordia", Udine, Italy
| | - Erika Malini
- Regional Coordinator Centre for Rare Diseases, Academic Hospital "Santa Maria della Misericordia", Udine, Italy
| | | | - Paolo Peruzzo
- Regional Coordinator Centre for Rare Diseases, Academic Hospital "Santa Maria della Misericordia", Udine, Italy
| | - Claus Bornaes
- Orphazyme A/S, Ole Maaloes vej 3, DK-2200 Copenhagen, Denmark
| | | | | | | | - Bruno Bembi
- Regional Coordinator Centre for Rare Diseases, Academic Hospital "Santa Maria della Misericordia", Udine, Italy
| | | | - Andrea Dardis
- Regional Coordinator Centre for Rare Diseases, Academic Hospital "Santa Maria della Misericordia", Udine, Italy
| | | |
Collapse
|
13
|
de Barros FRO, Paula-Lopes FF. Cellular and epigenetic changes induced by heat stress in bovine preimplantation embryos. Mol Reprod Dev 2018; 85:810-820. [DOI: 10.1002/mrd.23040] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 06/26/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Flavia R. O. de Barros
- Department of Biological Sciences; Federal University of São Paulo; São Paulo Brazil
- School of Bioprocess Engineering and Biotechnology, Federal University of Technology; Parana Brazil
| | | |
Collapse
|
14
|
Neckers L, Blagg B, Haystead T, Trepel JB, Whitesell L, Picard D. Methods to validate Hsp90 inhibitor specificity, to identify off-target effects, and to rethink approaches for further clinical development. Cell Stress Chaperones 2018; 23:467-482. [PMID: 29392504 PMCID: PMC6045531 DOI: 10.1007/s12192-018-0877-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
The molecular chaperone Hsp90 is one component of a highly complex and interactive cellular proteostasis network (PN) that participates in protein folding, directs misfolded and damaged proteins for destruction, and participates in regulating cellular transcriptional responses to environmental stress, thus promoting cell and organismal survival. Over the last 20 years, it has become clear that various disease states, including cancer, neurodegeneration, metabolic disorders, and infection by diverse microbes, impact the PN. Among PN components, Hsp90 was among the first to be pharmacologically targeted with small molecules. While the number of Hsp90 inhibitors described in the literature has dramatically increased since the first such small molecule was described in 1994, it has become increasingly apparent that not all of these agents have been sufficiently validated for specificity, mechanism of action, and lack of off-target effects. Given the less than expected activity of Hsp90 inhibitors in cancer-related human clinical trials, a re-evaluation of potentially confounding off-target effects, as well as confidence in target specificity and mechanism of action, is warranted. In this commentary, we provide feasible approaches to achieve these goals and we discuss additional considerations to improve the clinical efficacy of Hsp90 inhibitors in treating cancer and other diseases.
Collapse
Affiliation(s)
- Len Neckers
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, 20892, USA.
| | - Brian Blagg
- Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Luke Whitesell
- Whitehead Institute, Cambridge, MA, 02142, USA
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève, 1211, Geneva 4, Switzerland.
| |
Collapse
|
15
|
Yu J, Li Y, Wang T, Zhong X. Modification of N6-methyladenosine RNA methylation on heat shock protein expression. PLoS One 2018; 13:e0198604. [PMID: 29902206 PMCID: PMC6002042 DOI: 10.1371/journal.pone.0198604] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/22/2018] [Indexed: 11/18/2022] Open
Abstract
This study was conducted to investigate effect of N6-methyladenosine (m6A) RNA methylation on Heat shock proteins (HSPs) and dissect the profile of HSP RNA methylation. The results showed that m6A methyltransferases METTL3 mRNA was decreased in responses to heat shock stress in HepG2 cells, but m6A-specific binding protein YTHDF2 mRNA was upregulated in a manner similar to HSP70 induction. Immunofluorescence staining showed that the majority of YTHDF2 was present in the cytosol, however, nearly all YTHDF2 translocated from the cytosol into the nucleus after heat shock. METTL3 knockdown significantly changed HSP70, HSP60, and HSP27 mRNA expression in HepG2 cells using siRNA, however, mRNA lifetime was not impacted. Silence of YTHDF2 using siRNA did not change expression of HSP70, but significantly increased HSP90, HSP60, and HSPB1 mRNA expression. In addition, m6A-seq revealed that HSP m6A methylation peaks are mainly enriched on exons and around stop codons, and shows a unique distribution profile in the 5'UTR and 3'UTR. Knockdown of METTL3 changed the methylation patterns of HSPs transcript. In conclusion, m6A RNA methylation regulates HSP gene expression. Differential expression of HSPs modulated by m6A may depend on the m6A site and abundance of the target gene. This finding provides insights into new regulatory mechanisms of HSPs in normal and stress situations.
Collapse
Affiliation(s)
- Jiayao Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People’s Republic of China
| | - Yi Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People’s Republic of China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People’s Republic of China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
16
|
Col E, Hoghoughi N, Dufour S, Penin J, Koskas S, Faure V, Ouzounova M, Hernandez-Vargash H, Reynoird N, Daujat S, Folco E, Vigneron M, Schneider R, Verdel A, Khochbin S, Herceg Z, Caron C, Vourc'h C. Bromodomain factors of BET family are new essential actors of pericentric heterochromatin transcriptional activation in response to heat shock. Sci Rep 2017; 7:5418. [PMID: 28710461 PMCID: PMC5511177 DOI: 10.1038/s41598-017-05343-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 05/30/2017] [Indexed: 11/10/2022] Open
Abstract
The heat shock response is characterized by the transcriptional activation of both hsp genes and noncoding and repeated satellite III DNA sequences located at pericentric heterochromatin. Both events are under the control of Heat Shock Factor I (HSF1). Here we show that under heat shock, HSF1 recruits major cellular acetyltransferases, GCN5, TIP60 and p300 to pericentric heterochromatin leading to a targeted hyperacetylation of pericentric chromatin. Redistribution of histone acetylation toward pericentric region in turn directs the recruitment of Bromodomain and Extra-Terminal (BET) proteins BRD2, BRD3, BRD4, which are required for satellite III transcription by RNAP II. Altogether we uncover here a critical role for HSF1 in stressed cells relying on the restricted use of histone acetylation signaling over pericentric heterochromatin (HC).
Collapse
Affiliation(s)
- Edwige Col
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Neda Hoghoughi
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Solenne Dufour
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Jessica Penin
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Sivan Koskas
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Virginie Faure
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Maria Ouzounova
- International Agency for Research on Cancer (IARC), 69008, Lyon, France
| | | | - Nicolas Reynoird
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Sylvain Daujat
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Strasbourg, France
| | - Eric Folco
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Marc Vigneron
- UMR 7242, Ecole Supérieure de Biotechnologie de Strasbourg (ESBS), 300 boulevard Sebastien Brant, CS 10413, 67412, Illkirch, France
| | - Robert Schneider
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Strasbourg, France
- Institute of Functional Epigenetics, Helmholtz Zentrum Muenchen, Ingolstaedter Landstr 1, 85754, Neuherberg, Germany
| | - André Verdel
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Saadi Khochbin
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC), 69008, Lyon, France
| | - Cécile Caron
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Claire Vourc'h
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France.
| |
Collapse
|
17
|
Kisliouk T, Cramer T, Meiri N. Methyl CpG level at distal part of heat-shock protein promoter HSP70 exhibits epigenetic memory for heat stress by modulating recruitment of POU2F1-associated nucleosome-remodeling deacetylase (NuRD) complex. J Neurochem 2017; 141:358-372. [PMID: 28278364 DOI: 10.1111/jnc.14014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/09/2017] [Accepted: 02/24/2017] [Indexed: 02/06/2023]
Abstract
Depending on its stringency, exposure to heat in early life leads to either resilience or vulnerability to heat stress later in life. We hypothesized that epigenetic alterations in genes belonging to the cell proteostasis pathways are attributed to long-term responses to heat stress. Epigenetic regulation of the mRNA expression of the molecular chaperone heat-shock protein (HSP) 70 (HSPA2) was evaluated in the chick hypothalamus during the critical period of thermal-control establishment on day 3 post-hatch and during heat challenge on day 10. Both the level and duration of HSP70 expression during heat challenge a week after heat conditioning were more pronounced in chicks conditioned under harsh versus mild temperature. Analyzing different segments of the promoter in vitro indicated that methylation of a distal part altered its transcriptional activity. In parallel, DNA-methylation level of this segment in vivo was higher in harsh- compared to mild-heat-conditioned chicks. Hypermethylation of the HSP70 promoter in high-temperature-conditioned chicks was accompanied by a reduction in both POU Class 2 Homeobox 1 (POU2F1) binding and recruitment of the nucleosome remodeling deacetylase (NuRD) chromatin-remodeling complex. As a result, histone H3 acetylation levels at the HSP70 promoter were higher in harsh-temperature-conditioned chicks than in their mild-heat-conditioned counterparts. These results suggest that methylation level of a distal part of the HSP70 promoter and POU2F1 recruitment may reflect heat-stress-related epigenetic memory and may be useful in differentiating between individuals that are resilient or vulnerable to stress.
Collapse
Affiliation(s)
- Tatiana Kisliouk
- Institute of Animal Science, ARO, The Volcani Center, Rishon LeZion, Israel
| | - Tomer Cramer
- Institute of Animal Science, ARO, The Volcani Center, Rishon LeZion, Israel.,Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Rehovot, Israel
| | - Noam Meiri
- Institute of Animal Science, ARO, The Volcani Center, Rishon LeZion, Israel
| |
Collapse
|
18
|
Moruno-Manchon JF, Uzor NE, Blasco-Conesa MP, Mannuru S, Putluri N, Furr-Stimming EE, Tsvetkov AS. Inhibiting sphingosine kinase 2 mitigates mutant Huntingtin-induced neurodegeneration in neuron models of Huntington disease. Hum Mol Genet 2017; 26:1305-1317. [PMID: 28175299 PMCID: PMC6251541 DOI: 10.1093/hmg/ddx046] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/20/2017] [Accepted: 02/02/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington disease (HD) is the most common inherited neurodegenerative disorder. It has no cure. The protein huntingtin causes HD, and mutations to it confer toxic functions to the protein that lead to neurodegeneration. Thus, identifying modifiers of mutant huntingtin-mediated neurotoxicity might be a therapeutic strategy for HD. Sphingosine kinases 1 (SK1) and 2 (SK2) synthesize sphingosine-1-phosphate (S1P), a bioactive lipid messenger critically involved in many vital cellular processes, such as cell survival. In the nucleus, SK2 binds to and inhibits histone deacetylases 1 and 2 (HDAC1/2). Inhibiting both HDACs has been suggested as a potential therapy in HD. Here, we found that SK2 is nuclear in primary neurons and, unexpectedly, overexpressed SK2 is neurotoxic in a dose-dependent manner. SK2 promotes DNA double-strand breaks in cultured primary neurons. We also found that SK2 is hyperphosphorylated in the brain samples from a model of HD, the BACHD mice. These data suggest that the SK2 pathway may be a part of a pathogenic pathway in HD. ABC294640, an inhibitor of SK2, reduces DNA damage in neurons and increases survival in two neuron models of HD. Our results identify a novel regulator of mutant huntingtin-mediated neurotoxicity and provide a new target for developing therapies for HD.
Collapse
Affiliation(s)
- Jose F. Moruno-Manchon
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Ndidi-Ese Uzor
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Maria P. Blasco-Conesa
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Sishira Mannuru
- The University of Texas Medical Training Program, Houston, TX 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Erin E. Furr-Stimming
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Andrey S. Tsvetkov
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
19
|
Hussong M, Kaehler C, Kerick M, Grimm C, Franz A, Timmermann B, Welzel F, Isensee J, Hucho T, Krobitsch S, Schweiger MR. The bromodomain protein BRD4 regulates splicing during heat shock. Nucleic Acids Res 2016; 45:382-394. [PMID: 27536004 PMCID: PMC5224492 DOI: 10.1093/nar/gkw729] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 12/11/2022] Open
Abstract
The cellular response to heat stress is an ancient and evolutionarily highly conserved defence mechanism characterised by the transcriptional up-regulation of cyto-protective genes and a partial inhibition of splicing. These features closely resemble the proteotoxic stress response during tumor development. The bromodomain protein BRD4 has been identified as an integral member of the oxidative stress as well as of the inflammatory response, mainly due to its role in the transcriptional regulation process. In addition, there are also several lines of evidence implicating BRD4 in the splicing process. Using RNA-sequencing we found a significant increase in splicing inhibition, in particular intron retentions (IR), following heat treatment in BRD4-depleted cells. This leads to a decrease of mRNA abundancy of the affected transcripts, most likely due to premature termination codons. Subsequent experiments revealed that BRD4 interacts with the heat shock factor 1 (HSF1) such that under heat stress BRD4 is recruited to nuclear stress bodies and non-coding SatIII RNA transcripts are up-regulated. These findings implicate BRD4 as an important regulator of splicing during heat stress. Our data which links BRD4 to the stress induced splicing process may provide novel mechanisms of BRD4 inhibitors in regard to anti-cancer therapies.
Collapse
Affiliation(s)
- Michelle Hussong
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr.63-73, 14195 Berlin, Germany.,Department of Biology, Chemistry and Pharmacy, Free University Berlin, 14195 Berlin, Germany.,Functional Epigenomics, CCG, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Christian Kaehler
- Department of Biology, Chemistry and Pharmacy, Free University Berlin, 14195 Berlin, Germany.,Otto-Warburg Laboratory 'Neurodegenerative disorders', Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Martin Kerick
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr.63-73, 14195 Berlin, Germany.,Functional Epigenomics, CCG, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Christina Grimm
- Functional Epigenomics, CCG, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Alexandra Franz
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr.63-73, 14195 Berlin, Germany
| | - Bernd Timmermann
- Sequencing Core Facility, Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Franziska Welzel
- Otto-Warburg Laboratory 'Neurodegenerative disorders', Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University of Cologne, 50931 Cologne, Germany
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University of Cologne, 50931 Cologne, Germany
| | - Sylvia Krobitsch
- Otto-Warburg Laboratory 'Neurodegenerative disorders', Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Michal R Schweiger
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr.63-73, 14195 Berlin, Germany .,Functional Epigenomics, CCG, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| |
Collapse
|
20
|
Lenart P, Bienertová-Vašků J. Double strand breaks may be a missing link between entropy and aging. Mech Ageing Dev 2016; 157:1-6. [DOI: 10.1016/j.mad.2016.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/11/2016] [Accepted: 06/02/2016] [Indexed: 01/09/2023]
|
21
|
Calapre L, Gray ES, Kurdykowski S, David A, Hart P, Descargues P, Ziman M. Heat-mediated reduction of apoptosis in UVB-damaged keratinocytes in vitro and in human skin ex vivo. BMC DERMATOLOGY 2016; 16:6. [PMID: 27230291 PMCID: PMC4882820 DOI: 10.1186/s12895-016-0043-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 05/18/2016] [Indexed: 01/18/2023]
Abstract
Background UV radiation induces significant DNA damage in keratinocytes and is a known risk factor for skin carcinogenesis. However, it has been reported previously that repeated and simultaneous exposure to UV and heat stress increases the rate of cutaneous tumour formation in mice. Since constant exposure to high temperatures and UV are often experienced in the environment, the effects of exposure to UV and heat needs to be clearly addressed in human epidermal cells. Methods In this study, we determined the effects of repeated UVB exposure 1 kJ/m2 followed by heat (39 °C) to human keratinocytes. Normal human ex vivo skin models and primary keratinocytes (NHEK) were exposed once a day to UVB and/or heat stress for four consecutive days. Cells were then assessed for changes in proliferation, apoptosis and gene expression at 2 days post-exposure, to determine the cumulative and persistent effects of UV and/or heat in skin keratinocytes. Results Using ex vivo skin models and primary keratinocytes in vitro, we showed that UVB plus heat treated keratinocytes exhibit persistent DNA damage, as observed with UVB alone. However, we found that apoptosis was significantly reduced in UVB plus heat treated samples. Immunohistochemical and whole genome transcription analysis showed that multiple UVB plus heat exposures induced inactivation of the p53-mediated stress response. Furthermore, we demonstrated that repeated exposure to UV plus heat induced SIRT1 expression and a decrease in acetylated p53 in keratinocytes, which is consistent with the significant downregulation of p53-regulated pro-apoptotic and DNA damage repair genes in these cells. Conclusion Our results suggest that UVB-induced p53-mediated cell cycle arrest and apoptosis are reduced in the presence of heat stress, leading to increased survival of DNA damaged cells. Thus, exposure to UVB and heat stress may act synergistically to allow survival of damaged cells, which could have implications for initiation skin carcinogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12895-016-0043-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leslie Calapre
- School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, Perth, WA, 6027, Australia
| | - Elin S Gray
- School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, Perth, WA, 6027, Australia
| | | | - Anthony David
- GENOSKIN Centre Pierre Potier, Oncopole, Toulouse, France
| | - Prue Hart
- Telethon Kids Institute, University of Western Australia, 100 Roberts Road, Subiaco, Perth, 6008, Australia
| | | | - Mel Ziman
- School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, Perth, WA, 6027, Australia. .,Department of Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
22
|
Wang J, Jia ST, Jia S. New Insights into the Regulation of Heterochromatin. Trends Genet 2016; 32:284-294. [PMID: 27005444 DOI: 10.1016/j.tig.2016.02.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 10/22/2022]
Abstract
All living organisms are constantly exposed to stresses from internal biological processes and surrounding environments, which induce many adaptive changes in cellular physiology and gene expression programs. Unexpectedly, constitutive heterochromatin, which is generally associated with the stable maintenance of gene silencing, is also dynamically regulated in response to stimuli. In this review we discuss the mechanism of constitutive heterochromatin assembly, its dynamic nature, and its responses to environmental changes.
Collapse
Affiliation(s)
- Jiyong Wang
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Sharon T Jia
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Songtao Jia
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
23
|
A DNA element in the slo gene modulates ethanol tolerance. Alcohol 2016; 51:37-42. [PMID: 26992698 DOI: 10.1016/j.alcohol.2015.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 12/23/2015] [Accepted: 12/23/2015] [Indexed: 12/20/2022]
Abstract
In Drosophila, the slo gene encodes BK-type Ca(2+)-activated K(+) channels and is involved in producing rapid functional tolerance to sedation with ethanol. Drosophila are ideal for the study of functional ethanol tolerance because the adult does not acquire metabolic ethanol tolerance (Scholz, Ramond, Singh, & Heberlein, 2000). It has been shown that mutations in slo block the capacity to acquire tolerance, that sedation with ethanol vapor induces slo gene expression in the nervous system, and that transgenic induction of slo can phenocopy tolerance (Cowmeadow, Krishnan, & Atkinson, 2005; Cowmeadow et al., 2006). Here we use ethanol-induced histone acetylation to map a DNA regulatory element in the slo transcriptional control region and functionally test the element for a role in producing ethanol tolerance. Histone acetylation is commonly associated with activating transcription factors. We used the chromatin immunoprecipitation assay to map histone acetylation changes following ethanol sedation to identify an ethanol-responsive DNA element. Ethanol sedation induced an increase in histone acetylation over a 60 n DNA element called 6b, which is situated between the two ethanol-responsive neural promoters of the slo gene. Removal of the 6b element from the endogenous slo gene affected the production of functional ethanol tolerance as assayed in an ethanol-vapor recovery from sedation assay. Removal of element 6b extended the period of functional ethanol tolerance from ∼10 days to more than 21 days after a single ethanol-vapor sedation. This study demonstrates that mapping the position of ethanol-induced histone acetylation is an effective way to identify DNA regulatory elements that help to mediate the response of a gene to ethanol. Using this approach, we identified a DNA element, which is conserved among Drosophila species, and which is important for producing a behaviorally relevant ethanol response.
Collapse
|
24
|
Norouzitallab P, Baruah K, Biswas P, Vanrompay D, Bossier P. Probing the phenomenon of trained immunity in invertebrates during a transgenerational study, using brine shrimp Artemia as a model system. Sci Rep 2016; 6:21166. [PMID: 26876951 PMCID: PMC4753410 DOI: 10.1038/srep21166] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/15/2016] [Indexed: 01/05/2023] Open
Abstract
The invertebrate’s innate immune system was reported to show some form of adaptive features, termed trained immunity. However, the memory characteristics of innate immune system and the mechanisms behind such phenomena remain unclear. Using the invertebrate model Artemia, we verified the possibility or impossibility of trained immunity, examining the presence or absence of enduring memory against homologous and heterologous antigens (Vibrio spp.) during a transgenerational study. We also determined the mechanisms behind such phenomenon. Our results showed the occurrence of memory and partial discrimination in Artemia’s immune system, as manifested by increased resistance, for three successive generations, of the progenies of Vibrio-exposed ancestors towards a homologous bacterial strain, rather than to a heterologous strain. This increased resistance phenotype was associated with elevated levels of hsp70 and hmgb1 signaling molecules and alteration in the expression of key innate immunity-related genes. Our results also showed stochastic pattern in the acetylation and methylation levels of H4 and H3K4me3 histones, respectively, in the progenies whose ancestors were challenged. Overall results suggest that innate immune responses in invertebrates have the capacity to be trained, and epigenetic reprogramming of (selected) innate immune effectors is likely to have central place in the mechanisms leading to trained immunity.
Collapse
Affiliation(s)
- Parisa Norouzitallab
- Laboratory of Aquaculture &Artemia Reference Center, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Rozier 44, Ghent 9000, Belgium.,Lab of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure 653, Ghent 9000, Belgium
| | - Kartik Baruah
- Laboratory of Aquaculture &Artemia Reference Center, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Rozier 44, Ghent 9000, Belgium
| | - Priyanka Biswas
- Laboratory of Aquaculture &Artemia Reference Center, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Rozier 44, Ghent 9000, Belgium
| | - Daisy Vanrompay
- Lab of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure 653, Ghent 9000, Belgium
| | - Peter Bossier
- Laboratory of Aquaculture &Artemia Reference Center, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Rozier 44, Ghent 9000, Belgium
| |
Collapse
|
25
|
HERBOMEL G, GRICHINE A, FERTIN A, DELON A, VOURC'H C, SOUCHIER C, USSON Y. Wavelet transform analysis of chromatin texture changes during heat shock. J Microsc 2015; 262:295-305. [DOI: 10.1111/jmi.12363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/17/2015] [Indexed: 11/29/2022]
Affiliation(s)
- G. HERBOMEL
- INSERM, IAB, University Grenoble Alpes; Grenoble France
| | - A. GRICHINE
- INSERM, IAB, University Grenoble Alpes; Grenoble France
| | - A FERTIN
- CNRS, TIMC-IMAG, University Grenoble Alpes; Grenoble France
| | - A. DELON
- CNRS, LIPHY, University Grenoble Alpes; Grenoble France
| | - C. VOURC'H
- INSERM, IAB, University Grenoble Alpes; Grenoble France
| | - C. SOUCHIER
- INSERM, IAB, University Grenoble Alpes; Grenoble France
| | - Y. USSON
- CNRS, TIMC-IMAG, University Grenoble Alpes; Grenoble France
| |
Collapse
|
26
|
Miozzo F, Sabéran-Djoneidi D, Mezger V. HSFs, Stress Sensors and Sculptors of Transcription Compartments and Epigenetic Landscapes. J Mol Biol 2015; 427:3793-816. [DOI: 10.1016/j.jmb.2015.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/02/2015] [Accepted: 10/09/2015] [Indexed: 01/06/2023]
|
27
|
Qureshi IA, Mehler MF. Epigenetics and therapeutic targets mediating neuroprotection. Brain Res 2015; 1628:265-272. [PMID: 26236020 DOI: 10.1016/j.brainres.2015.07.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/14/2015] [Accepted: 07/22/2015] [Indexed: 11/29/2022]
Abstract
The rapidly evolving science of epigenetics is transforming our understanding of the nervous system in health and disease and holds great promise for the development of novel diagnostic and therapeutic approaches targeting neurological diseases. Increasing evidence suggests that epigenetic factors and mechanisms serve as important mediators of the pathogenic processes that lead to irrevocable neural injury and of countervailing homeostatic and regenerative responses. Epigenetics is, therefore, of considerable translational significance to the field of neuroprotection. In this brief review, we provide an overview of epigenetic mechanisms and highlight the emerging roles played by epigenetic processes in neural cell dysfunction and death and in resultant neuroprotective responses. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Irfan A Qureshi
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Mark F Mehler
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Ruth L. and David S. Gottesman Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Center for Epigenomics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
28
|
El Fatimy R, Miozzo F, Le Mouël A, Abane R, Schwendimann L, Sabéran-Djoneidi D, de Thonel A, Massaoudi I, Paslaru L, Hashimoto-Torii K, Christians E, Rakic P, Gressens P, Mezger V. Heat shock factor 2 is a stress-responsive mediator of neuronal migration defects in models of fetal alcohol syndrome. EMBO Mol Med 2015; 6:1043-61. [PMID: 25027850 PMCID: PMC4154132 DOI: 10.15252/emmm.201303311] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is a frequent cause of mental retardation. However, the molecular mechanisms underlying brain development defects induced by maternal alcohol consumption during pregnancy are unclear. We used normal and Hsf2-deficient mice and cell systems to uncover a pivotal role for heat shock factor 2 (HSF2) in radial neuronal migration defects in the cortex, a hallmark of fetal alcohol exposure. Upon fetal alcohol exposure, HSF2 is essential for the triggering of HSF1 activation, which is accompanied by distinctive post-translational modifications, and HSF2 steers the formation of atypical alcohol-specific HSF1-HSF2 heterocomplexes. This perturbs the in vivo binding of HSF2 to heat shock elements (HSEs) in genes that control neuronal migration in normal conditions, such as p35 or the MAPs (microtubule-associated proteins, such as Dclk1 and Dcx), and alters their expression. In the absence of HSF2, migration defects as well as alterations in gene expression are reduced. Thus, HSF2, as a sensor for alcohol stress in the fetal brain, acts as a mediator of the neuronal migration defects associated with FASD.
Collapse
Affiliation(s)
- Rachid El Fatimy
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Federico Miozzo
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Anne Le Mouël
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Ryma Abane
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Leslie Schwendimann
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Aurélie de Thonel
- INSERM UMR 866, Dijon, France Faculty of Medicine and Pharmacy, Univ Burgundy, Dijon, France
| | - Illiasse Massaoudi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Liliana Paslaru
- Carol Davila University of Medicine and Pharmacy Fundeni Hospital, Bucharest, Romania
| | - Kazue Hashimoto-Torii
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Elisabeth Christians
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, CNRS, Villefranche-sur-mer, France Sorbonne Universités UPMC Univ Paris 06, Villefranche-sur-mer, France
| | - Pasko Rakic
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Pierre Gressens
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Valérie Mezger
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| |
Collapse
|
29
|
Morano KA, Sistonen L, Mezger V. Heat shock in the springtime. Cell Stress Chaperones 2014; 19:753-61. [PMID: 25199949 PMCID: PMC4389858 DOI: 10.1007/s12192-014-0539-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 08/12/2014] [Accepted: 08/12/2014] [Indexed: 11/26/2022] Open
Abstract
A collaborative workshop dedicated to the discussion of heat shock factors in stress response, development, and disease was held on April 22-24, 2014 at the Université Paris Diderot in Paris, France. Recent years have witnessed an explosion of interest in these highly conserved transcription factors, with biological roles ranging from environmental sensing to human development and cancer.
Collapse
Affiliation(s)
- Kevin A. Morano
- />Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, Houston, TX 77030 USA
| | - Lea Sistonen
- />Department of Biosciences, Åbo Akademi University, BioCity, 20520 Turku, Finland
| | - Valérie Mezger
- />UMR7216 Epigenetics and Cell Fate, CNRS, F-75205 Paris Cedex 13, France
- />University Paris Diderot, Sorbonne Paris Cité, F-75205 Paris Cedex 13, France
| |
Collapse
|
30
|
Neueder A, Achilli F, Moussaoui S, Bates GP. Novel isoforms of heat shock transcription factor 1, HSF1γα and HSF1γβ, regulate chaperone protein gene transcription. J Biol Chem 2014; 289:19894-906. [PMID: 24855652 PMCID: PMC4106310 DOI: 10.1074/jbc.m114.570739] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The heat shock response, resulting in the production of heat shock proteins or molecular chaperones, is triggered by elevated temperature and a variety of other stressors. Its master regulator is heat shock transcription factor 1 (HSF1). Heat shock factors generally exist in multiple isoforms. The two known isoforms of HSF1 differ in the inclusion (HSF1α) or exclusion (HSF1β) of exon 11. Although there are some data concerning the differential expression patterns and transcriptional activities of HSF2 isoforms during development, little is known about the distinct properties of the HSF1 isoforms. Here we present evidence for two novel HSF1 isoforms termed HSF1γα and HSF1γβ, and we show that the HSF1 isoform ratio differentially regulates heat shock protein gene transcription. Hsf1γ isoforms are expressed in various mouse tissues and are translated into protein. Furthermore, after heat shock, HSF1γ isoforms are exported from the nucleus more rapidly or degraded more quickly than HSF1α or HSF1β. We also show that each individual HSF1 isoform is sufficient to induce the heat shock response and that expression of combinations of HSF1 isoforms, in particular HSF1α and HSF1β, results in a synergistic enhancement of the transcriptional response. In addition, HSF1γ isoforms potentially suppress the synergistic effect of HSF1α and HSF1β co-expression. Collectively, our observations suggest that the expression of HSF1 isoforms in a specific ratio provides an additional layer in the regulation of heat shock protein gene transcription.
Collapse
Affiliation(s)
- Andreas Neueder
- From the Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom and
| | - Francesca Achilli
- From the Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom and
| | - Saliha Moussaoui
- Neuroscience Discovery, Novartis Institute for Biomedical Research, CH-4002 Basel, Switzerland
| | - Gillian P Bates
- From the Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom and
| |
Collapse
|
31
|
Ingemann L, Kirkegaard T. Lysosomal storage diseases and the heat shock response: convergences and therapeutic opportunities. J Lipid Res 2014; 55:2198-210. [PMID: 24837749 DOI: 10.1194/jlr.r048090] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lysosomes play a vital role in the maintenance of cellular homeostasis through the recycling of cell constituents, a key metabolic function which is highly dependent on the correct function of the lysosomal hydrolases and membrane proteins, as well as correct membrane lipid stoichiometry and composition. The critical role of lysosomal functionality is evident from the severity of the diseases in which the primary lesion is a genetically defined loss-of-function of lysosomal hydrolases or membrane proteins. This group of diseases, known as lysosomal storage diseases (LSDs), number more than 50 and are associated with severe neurodegeneration, systemic disease, and early death, with only a handful of the diseases having a therapeutic option. Another key homeostatic system is the metabolic stress response or heat shock response (HSR), which is induced in response to a number of physiological and pathological stresses, such as protein misfolding and aggregation, endoplasmic reticulum stress, oxidative stress, nutrient deprivation, elevated temperature, viral infections, and various acute traumas. Importantly, the HSR and its cardinal members of the heat shock protein 70 family has been shown to protect against a number of degenerative diseases, including severe diseases of the nervous system. The cytoprotective actions of the HSR also include processes involving the lysosomal system, such as cell death, autophagy, and protection against lysosomal membrane permeabilization, and have shown promise in a number of LSDs. This review seeks to describe the emerging understanding of the interplay between these two essential metabolic systems, the lysosomes and the HSR, with a particular focus on their potential as a therapeutic target for LSDs.
Collapse
|
32
|
Abstract
This review summarizes the current understanding of the role of nuclear bodies in regulating gene expression. The compartmentalization of cellular processes, such as ribosome biogenesis, RNA processing, cellular response to stress, transcription, modification and assembly of spliceosomal snRNPs, histone gene synthesis and nuclear RNA retention, has significant implications for gene regulation. These functional nuclear domains include the nucleolus, nuclear speckle, nuclear stress body, transcription factory, Cajal body, Gemini of Cajal body, histone locus body and paraspeckle. We herein review the roles of nuclear bodies in regulating gene expression and their relation to human health and disease.
Collapse
Affiliation(s)
| | - Cornelius F. Boerkoel
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-604-875-2157; Fax: +1-604-875-2376
| |
Collapse
|
33
|
Norouzitallab P, Baruah K, Vandegehuchte M, Van Stappen G, Catania F, Bussche JV, Vanhaecke L, Sorgeloos P, Bossier P. Environmental heat stress induces epigenetic transgenerational inheritance of robustness in parthenogenetic
Artemia
model. FASEB J 2014; 28:3552-63. [DOI: 10.1096/fj.14-252049] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Parisa Norouzitallab
- Laboratory of AquacultureGhent UniversityGhentBelgium
- Artemia Reference CenterGhent UniversityGhentBelgium
| | - Kartik Baruah
- Laboratory of AquacultureGhent UniversityGhentBelgium
- Artemia Reference CenterGhent UniversityGhentBelgium
| | - Michiel Vandegehuchte
- Laboratory of Environmental Toxicology and Aquatic EcologyGhent UniversityGhentBelgium
| | - Gilbert Van Stappen
- Laboratory of AquacultureGhent UniversityGhentBelgium
- Artemia Reference CenterGhent UniversityGhentBelgium
| | - Francesco Catania
- Institute for Evolution and Biodiversity, University of MünsterMünsterGermany
| | | | - Lynn Vanhaecke
- Laboratory of Chemical AnalysisGhent UniversityMerelbekeBelgium
| | - Patrick Sorgeloos
- Laboratory of AquacultureGhent UniversityGhentBelgium
- Artemia Reference CenterGhent UniversityGhentBelgium
| | - Peter Bossier
- Laboratory of AquacultureGhent UniversityGhentBelgium
- Artemia Reference CenterGhent UniversityGhentBelgium
| |
Collapse
|
34
|
Li X, Ghezzi A, Pohl JB, Bohm AY, Atkinson NS. A DNA element regulates drug tolerance and withdrawal in Drosophila. PLoS One 2013; 8:e75549. [PMID: 24086565 PMCID: PMC3781064 DOI: 10.1371/journal.pone.0075549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/19/2013] [Indexed: 01/05/2023] Open
Abstract
Drug tolerance and withdrawal are insidious responses to drugs of abuse; the first increases drug consumption while the second punishes abstention. Drosophila generate functional tolerance to benzyl alcohol sedation by increasing neural expression of the slo BK-type Ca(2+) activated K(+) channel gene. After drug clearance this change produces a withdrawal phenotype-increased seizure susceptibility. The drug-induced histone modification profile identified the 6b element (60 nt) as a drug responsive element. Genomic deletion of 6b produces the allele, slo (Δ6b), that reacts more strongly to the drug with increased induction, a massive increase in the duration of tolerance, and an increase in the withdrawal phenotype yet does not alter other slo-dependent behaviors. The 6b element is a homeostatic regulator of BK channel gene expression and is the first cis-acting DNA element shown to specifically affect the duration of a drug action.
Collapse
Affiliation(s)
- Xiaolei Li
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, the University of Texas at Austin, Austin, Texas, United States of America
| | - Alfredo Ghezzi
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, the University of Texas at Austin, Austin, Texas, United States of America
| | - Jascha B. Pohl
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, the University of Texas at Austin, Austin, Texas, United States of America
| | - Arun Y. Bohm
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, the University of Texas at Austin, Austin, Texas, United States of America
| | - Nigel S. Atkinson
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, the University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
35
|
Kus-Liśkiewicz M, Polańska J, Korfanty J, Olbryt M, Vydra N, Toma A, Widłak W. Impact of heat shock transcription factor 1 on global gene expression profiles in cells which induce either cytoprotective or pro-apoptotic response following hyperthermia. BMC Genomics 2013; 14:456. [PMID: 23834426 PMCID: PMC3711851 DOI: 10.1186/1471-2164-14-456] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 07/01/2013] [Indexed: 11/23/2022] Open
Abstract
Background Elevated temperatures induce activation of the heat shock transcription factor 1 (HSF1) which in somatic cells leads to heat shock proteins synthesis and cytoprotection. However, in the male germ cells (spermatocytes) caspase-3 dependent apoptosis is induced upon HSF1 activation and spermatogenic cells are actively eliminated. Results To elucidate a mechanism of such diverse HSF1 activity we carried out genome-wide transcriptional analysis in control and heat-shocked cells, either spermatocytes or hepatocytes. Additionally, to identify direct molecular targets of active HSF1 we used chromatin immunoprecipitation assay (ChIP) combined with promoter microarrays (ChIP on chip). Genes that are differently regulated after HSF1 binding during hyperthermia in both types of cells have been identified. Despite HSF1 binding to promoter sequences in both types of cells, strong up-regulation of Hsps and other genes typically activated by the heat shock was observed only in hepatocytes. In spermatocytes HSF1 binding correlates with transcriptional repression on a large scale. HSF1-bound and negatively regulated genes encode mainly for proteins required for cell division, involved in RNA processing and piRNA biogenesis. Conclusions Observed suppression of the transcription could lead to genomic instability caused by meiotic recombination disturbances, which in turn might induce apoptosis of spermatogenic cells. We propose that HSF1-dependent induction of cell death is caused by the simultaneous repression of many genes required for spermatogenesis, which guarantees the elimination of cells damaged during heat shock. Such activity of HSF1 prevents transmission of damaged genetic material to the next generation.
Collapse
Affiliation(s)
- Małgorzata Kus-Liśkiewicz
- Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | | | | | | | | | | | | |
Collapse
|
36
|
Oshlag JZ, Devasthanam AS, Tomasi TB. Mild hyperthermia enhances the expression and induces oscillations in the Dicer protein. Int J Hyperthermia 2013; 29:51-61. [PMID: 23311378 DOI: 10.3109/02656736.2012.753471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To investigate whether mild heat stress at 39.5°C altered Dicer protein and miRNA expression patterns in several cell types. METHODS Multiple human and mouse cell types were cultured during the course of 9 h at temperatures from 37°C to 39.5°C. Dicer mRNA levels and microRNAs were quantified by TaqMan RT-qPCR assays and Dicer protein by western blotting. RESULTS Dicer protein was substantially elevated on western analysis in response to heat stress at 39.5°C in the absence of significant changes in Dicer mRNA by RT-qPCR. CONCLUSIONS Heat-induced regulation of Dicer expression occurs primarily post- transcriptionally, and the expression levels of Dicer protein are increased and often oscillate in response to fever-range hyperthermia in multiple mouse and human cells. Our studies suggest a potential role for Dicer and microRNAs in the response to mild thermal stress. Additional studies on the mechanisms involved in the stress-induced oscillations of Dicer protein and microRNAs will be of interest.
Collapse
Affiliation(s)
- Julian Z Oshlag
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | |
Collapse
|
37
|
Brandman O, Stewart-Ornstein J, Wong D, Larson A, Williams CC, Li GW, Zhou S, King D, Shen PS, Weibezahn J, Dunn JG, Rouskin S, Inada T, Frost A, Weissman JS. A ribosome-bound quality control complex triggers degradation of nascent peptides and signals translation stress. Cell 2013; 151:1042-54. [PMID: 23178123 PMCID: PMC3534965 DOI: 10.1016/j.cell.2012.10.044] [Citation(s) in RCA: 456] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/03/2012] [Accepted: 10/30/2012] [Indexed: 10/27/2022]
Abstract
The conserved transcriptional regulator heat shock factor 1 (Hsf1) is a key sensor of proteotoxic and other stress in the eukaryotic cytosol. We surveyed Hsf1 activity in a genome-wide loss-of-function library in Saccaromyces cerevisiae as well as ~78,000 double mutants and found Hsf1 activity to be modulated by highly diverse stresses. These included disruption of a ribosome-bound complex we named the Ribosome Quality Control Complex (RQC) comprising the Ltn1 E3 ubiquitin ligase, two highly conserved but poorly characterized proteins (Tae2 and Rqc1), and Cdc48 and its cofactors. Electron microscopy and biochemical analyses revealed that the RQC forms a stable complex with 60S ribosomal subunits containing stalled polypeptides and triggers their degradation. A negative feedback loop regulates the RQC, and Hsf1 senses an RQC-mediated translation-stress signal distinctly from other stresses. Our work reveals the range of stresses Hsf1 monitors and elucidates a conserved cotranslational protein quality control mechanism.
Collapse
Affiliation(s)
- Onn Brandman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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: 1.3] [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
|
39
|
de Thonel A, Le Mouël A, Mezger V. Transcriptional regulation of small HSP-HSF1 and beyond. Int J Biochem Cell Biol 2012; 44:1593-612. [PMID: 22750029 DOI: 10.1016/j.biocel.2012.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 12/16/2022]
Abstract
The members of the small heat shock protein (sHSP) family are molecular chaperones that play major roles in development, stress responses, and diseases, and have been envisioned as targets for therapy, particularly in cancer. The molecular mechanisms that regulate their transcription, in normal, stress, or pathological conditions, are characterized by extreme complexity and subtlety. Although historically linked to the heat shock transcription factors (HSFs), the stress-induced or developmental expression of the diverse members, including HSPB1/Hsp27/Hsp25, αA-crystallin/HSPB4, and αB-crystallin/HSPB5, relies on the combinatory effects of many transcription factors. Coupled with remarkably different cis-element architectures in the sHsp regulatory regions, they confer to each member its developmental expression or stress-inducibility. For example, multiple regulatory pathways coordinate the spatio-temporal expression of mouse αA-, αB-crystallin, and Hsp25 genes during lens development, through the action of master genes, like the large Maf family proteins and Pax6, but also HSF4. The inducibility of Hsp27 and αB-crystallin transcription by various stresses is exerted by HSF-dependent mechanisms, by which concomitant induction of Hsp27 and αB-crystallin expression is observed. In contrast, HSF-independent pathways can lead to αB-crystallin expression, but not to Hsp27 induction. Not surprisingly, deregulation of the expression of sHSP is associated with various pathologies, including cancer, neurodegenerative, or cardiac diseases. However, many questions remain to be addressed, and further elucidation of the developmental mechanisms of sHsp gene transcription might help to unravel the tissue- and stage-specific functions of this fascinating class of proteins, which might prove to be crucial for future therapeutic strategies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
|
40
|
do Rego TG, Roider HG, de Carvalho FAT, Costa IG. Inferring epigenetic and transcriptional regulation during blood cell development with a mixture of sparse linear models. Bioinformatics 2012; 28:2297-303. [PMID: 22730432 DOI: 10.1093/bioinformatics/bts362] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MOTIVATION Blood cell development is thought to be controlled by a circuit of transcription factors (TFs) and chromatin modifications that determine the cell fate through activating cell type-specific expression programs. To shed light on the interplay between histone marks and TFs during blood cell development, we model gene expression from regulatory signals by means of combinations of sparse linear regression models. RESULTS The mixture of sparse linear regression models was able to improve the gene expression prediction in relation to the use of a single linear model. Moreover, it performed an efficient selection of regulatory signals even when analyzing all TFs with known motifs (>600). The method identified interesting roles for histone modifications and a selection of TFs related to blood development and chromatin remodelling. AVAILABILITY The method and datasets are available from http://www.cin.ufpe.br/~igcf/SparseMix. CONTACT igcf@cin.ufpe.br SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Thais G do Rego
- Center of Informatics, Federal University of Pernambuco, Recife 50740-560, Brazil
| | | | | | | |
Collapse
|
41
|
Ontogenetic survey of histone modifications in an annelid. GENETICS RESEARCH INTERNATIONAL 2012; 2012:392903. [PMID: 22567386 PMCID: PMC3335605 DOI: 10.1155/2012/392903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/21/2011] [Indexed: 01/06/2023]
Abstract
Histone modifications are widely recognized for their fundamental importance in regulating gene expression in embryonic development in a wide range of eukaryotes, but they have received relatively little attention in the development of marine invertebrates. We surveyed histone modifications throughout the development of a marine annelid, Polydora cornuta, to determine if modifications could be detected immunohistochemically and if there were characteristic changes in modifications throughout ontogeny (surveyed at representative stages from oocyte to adult). We found a common time of onset for three histone modifications in early cleavage (H3K14ac, H3K9me, and H3K4me2), some differences in the distribution of modifications among germ layers, differences in epifluorescence intensity in specific cell lineages suggesting that hyperacetylation (H3K14ac) and hypermethylation (H3K9me) occur during differentiation, and an overall decrease in the distribution of modifications from larvae to adults. Although preliminary, these results suggest that histone modifications are involved in activating early development and differentiation in a marine invertebrate.
Collapse
|
42
|
Yao ZG, Zhang L, Huang L, Zhu H, Liu Y, Ma CM, Sheng SL, Qin C. Regional and cell-type specific distribution of HDAC2 in the adult mouse brain. Brain Struct Funct 2012; 218:563-73. [DOI: 10.1007/s00429-012-0416-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 04/06/2012] [Indexed: 12/27/2022]
|
43
|
Teves SS, Henikoff S. Heat shock reduces stalled RNA polymerase II and nucleosome turnover genome-wide. Genes Dev 2011; 25:2387-97. [PMID: 22085965 DOI: 10.1101/gad.177675.111] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Heat shock rapidly induces expression of a subset of genes while globally repressing transcription, making it an attractive system to study alterations in the chromatin landscape that accompany changes in gene regulation. We characterized these changes in Drosophila cells by profiling classical low-salt-soluble chromatin, RNA polymerase II (Pol II), and nucleosome turnover dynamics at single-base-pair resolution. With heat shock, low-salt-soluble chromatin and stalled Pol II levels were found to decrease within gene bodies, but no overall changes were detected at transcriptional start sites. Strikingly, nucleosome turnover decreased genome-wide within gene bodies upon heat shock in a pattern similar to that observed with inhibition of Pol II elongation, especially at genes involved in the heat-shock response. Relatively high levels of nucleosome turnover were also observed throughout the bodies of genes with paused Pol II. These observations suggest that down-regulation of transcription during heat shock involves reduced nucleosome mobility and that this process has evolved to promote heat-shock gene regulation. Our ability to precisely map both nucleosomal and subnucleosomal particles directly from low-salt-soluble chromatin extracts to assay changes in the chromatin landscape provides a simple general strategy for epigenome characterization.
Collapse
Affiliation(s)
- Sheila S Teves
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | |
Collapse
|
44
|
Zorzi E, Bonvini P. Inducible hsp70 in the regulation of cancer cell survival: analysis of chaperone induction, expression and activity. Cancers (Basel) 2011; 3:3921-56. [PMID: 24213118 PMCID: PMC3763403 DOI: 10.3390/cancers3043921] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/26/2011] [Accepted: 10/10/2011] [Indexed: 12/31/2022] Open
Abstract
Understanding the mechanisms that control stress is central to realize how cells respond to environmental and physiological insults. All the more important is to reveal how tumour cells withstand their harsher growth conditions and cope with drug-induced apoptosis, since resistance to chemotherapy is the foremost complication when curing cancer. Intensive research on tumour biology over the past number of years has provided significant insights into the molecular events that occur during oncogenesis, and resistance to anti-cancer drugs has been shown to often rely on stress response and expression of inducible heat shock proteins (HSPs). However, with respect to the mechanisms guarding cancer cells against proteotoxic stresses and the modulatory effects that allow their survival, much remains to be defined. Heat shock proteins are molecules responsible for folding newly synthesized polypeptides under physiological conditions and misfolded proteins under stress, but their role in maintaining the transformed phenotype often goes beyond their conventional chaperone activity. Expression of inducible HSPs is known to correlate with limited sensitivity to apoptosis induced by diverse cytotoxic agents and dismal prognosis of several tumour types, however whether cancer cells survive because of the constitutive expression of heat shock proteins or the ability to induce them when adapting to the hostile microenvironment remains to be elucidated. Clear is that tumours appear nowadays more "addicted" to heat shock proteins than previously envisaged, and targeting HSPs represents a powerful approach and a future challenge for sensitizing tumours to therapy. This review will focus on the anti-apoptotic role of heat shock 70kDa protein (Hsp70), and how regulatory factors that control inducible Hsp70 synthesis, expression and activity may be relevant for response to stress and survival of cancer cells.
Collapse
Affiliation(s)
- Elisa Zorzi
- OncoHematology Clinic of Pediatrics, University-Hospital of Padova, 35100 Padova, Italy; E-Mail:
| | - Paolo Bonvini
- OncoHematology Clinic of Pediatrics, University-Hospital of Padova, 35100 Padova, Italy; E-Mail:
- Fondazione Città della Speranza, 36030 Monte di Malo, Vicenza, Italy
| |
Collapse
|
45
|
Scharf KD, Berberich T, Ebersberger I, Nover L. The plant heat stress transcription factor (Hsf) family: structure, function and evolution. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1819:104-19. [PMID: 22033015 DOI: 10.1016/j.bbagrm.2011.10.002] [Citation(s) in RCA: 571] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/06/2011] [Accepted: 10/07/2011] [Indexed: 12/13/2022]
Abstract
Ten years after the first overview of a complete plant Hsf family was presented for Arabidopsis thaliana by Nover et al. [1], we compiled data for 252 Hsfs from nine plant species (five eudicots and four monocots) with complete or almost complete genome sequences. The new data set provides interesting insights into phylogenetic relationships within the Hsf family in plants and allows the refinement of their classification into distinct groups. Numerous publications over the last decade document the diversification and functional interaction of Hsfs as well as their integration into the complex stress signaling and response networks of plants. This article is part of a Special Issue entitled: Plant gene regulation in response to abiotic stress.
Collapse
Affiliation(s)
- Klaus-Dieter Scharf
- Molecular Cellbiology of Plants, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/M., Germany.
| | | | | | | |
Collapse
|
46
|
Labbadia J, Cunliffe H, Weiss A, Katsyuba E, Sathasivam K, Seredenina T, Woodman B, Moussaoui S, Frentzel S, Luthi-Carter R, Paganetti P, Bates GP. Altered chromatin architecture underlies progressive impairment of the heat shock response in mouse models of Huntington disease. J Clin Invest 2011; 121:3306-19. [PMID: 21785217 PMCID: PMC3148745 DOI: 10.1172/jci57413] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 06/03/2011] [Indexed: 01/05/2023] Open
Abstract
Huntington disease (HD) is a devastating neurodegenerative disorder for which there are no disease-modifying treatments. Previous studies have proposed that activation of the heat shock response (HSR) via the transcription factor heat shock factor 1 (HSF1) may be of therapeutic benefit. However, the effect of disease progression on the HSR and the therapeutic potential of this pathway are currently unknown. Here, we used a brain-penetrating HSP90 inhibitor and physiological, molecular, and behavioral readouts to demonstrate that pharmacological activation of HSF1 improves huntingtin aggregate load, motor performance, and other HD-related phenotypes in the R6/2 mouse model of HD. However, the beneficial effects of this treatment were transient and diminished with disease progression. Molecular analyses to understand the transient nature of these effects revealed altered chromatin architecture, reduced HSF1 binding, and impaired HSR accompanied disease progression in both the R6/2 transgenic and HdhQ150 knockin mouse models of HD. Taken together, our findings reveal that the HSR, a major inducible regulator of protein homeostasis and longevity, is disrupted in HD. Consequently, pharmacological induction of HSF1 as a therapeutic approach to HD is more complex than was previously anticipated.
Collapse
Affiliation(s)
- John Labbadia
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Helen Cunliffe
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Andreas Weiss
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Elena Katsyuba
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Kirupa Sathasivam
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Tamara Seredenina
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Ben Woodman
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Saliha Moussaoui
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Stefan Frentzel
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Ruth Luthi-Carter
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Paolo Paganetti
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Gillian P. Bates
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom.
Novartis Institutes for BioMedical Research, Neuroscience Discovery, Basel, Switzerland.
Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
47
|
Strenkert D, Schmollinger S, Sommer F, Schulz-Raffelt M, Schroda M. Transcription factor-dependent chromatin remodeling at heat shock and copper-responsive promoters in Chlamydomonas reinhardtii. THE PLANT CELL 2011; 23:2285-301. [PMID: 21705643 PMCID: PMC3160021 DOI: 10.1105/tpc.111.085266] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/09/2011] [Accepted: 06/07/2011] [Indexed: 05/19/2023]
Abstract
How transcription factors affect chromatin structure to regulate gene expression in response to changes in environmental conditions is poorly understood in the green lineage. To shed light on this issue, we used chromatin immunoprecipitation and formaldehyde-assisted isolation of regulatory elements to investigate the chromatin structure at target genes of HSF1 and CRR1, key transcriptional regulators of the heat shock and copper starvation responses, respectively, in the unicellular green alga Chlamydomonas reinhardtii. Generally, we detected lower nucleosome occupancy, higher levels of histone H3/4 acetylation, and lower levels of histone H3 Lys 4 (H3K4) monomethylation at promoter regions of active genes compared with inactive promoters and transcribed and intergenic regions. Specifically, we find that activated HSF1 and CRR1 transcription factors mediate the acetylation of histones H3/4, nucleosome eviction, remodeling of the H3K4 mono- and dimethylation marks, and transcription initiation/elongation. By this, HSF1 and CRR1 quite individually remodel and activate target promoters that may be inactive and embedded into closed chromatin (HSP22F/CYC6) or weakly active and embedded into partially opened (CPX1) or completely opened chromatin (HSP70A/CRD1). We also observed HSF1-independent histone H3/4 deacetylation at the RBCS2 promoter after heat shock, suggesting interplay of specific and presumably more generally acting factors to adapt gene expression to the new requirements of a changing environment.
Collapse
|
48
|
Mandrekar P. Epigenetic regulation in alcoholic liver disease. World J Gastroenterol 2011; 17:2456-64. [PMID: 21633650 PMCID: PMC3103803 DOI: 10.3748/wjg.v17.i20.2456] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/02/2011] [Accepted: 03/09/2011] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is characterized by steatosis or fat deposition in the liver and inflammation, which leads to cirrhosis and hepatocellular carcinoma. Induction of target genes without involving changes in DNA sequence seems to contribute greatly to liver injury. Chromatin modifications including alterations in histones and DNA, as well as post-transcriptional changes collectively referred to as epigenetic effects are altered by alcohol. Recent studies have pointed to a significant role for epigenetic mechanisms at the nucleosomal level influencing gene expression and disease outcome in ALD. Specifically, epigenetic alterations by alcohol include histone modifications such as changes in acetylation and phosphorylation, hypomethylation of DNA, and alterations in miRNAs. These modifications can be induced by alcohol-induced oxidative stress that results in altered recruitment of transcriptional machinery and abnormal gene expression. Delineating these mechanisms in initiation and progression of ALD is becoming a major area of interest. This review summarizes key epigenetic mechanisms that are dysregulated by alcohol in the liver. Alterations by alcohol in histone and DNA modifications, enzymes related to histone acetylation such as histone acetyltransferases, histone deacetylases and sirtuins, and methylation enzymes such as DNA methyltransferases are discussed. Chromatin modifications and miRNA alterations that result in immune cell dysfunction contributing to inflammatory cytokine production in ALD is reviewed. Finally, the role of alcohol-mediated oxidative stress in epigenetic regulation in ALD is described. A better understanding of these mechanisms is crucial for designing novel epigenetic based therapies to ameliorate ALD.
Collapse
|
49
|
Vabulas RM, Raychaudhuri S, Hayer-Hartl M, Hartl FU. Protein folding in the cytoplasm and the heat shock response. Cold Spring Harb Perspect Biol 2011; 2:a004390. [PMID: 21123396 DOI: 10.1101/cshperspect.a004390] [Citation(s) in RCA: 295] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Proteins generally must fold into precise three-dimensional conformations to fulfill their biological functions. In the cell, this fundamental process is aided by molecular chaperones, which act in preventing protein misfolding and aggregation. How this machinery assists newly synthesized polypeptide chains in navigating the complex folding energy landscape is now being understood in considerable detail. The mechanisms that ensure the maintenance of a functional proteome under normal and stress conditions are also of great medical relevance, as the aggregation of proteins that escape the cellular quality control underlies a range of debilitating diseases, including many age-of-onset neurodegenerative disorders.
Collapse
Affiliation(s)
- R Martin Vabulas
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | | | | | | |
Collapse
|
50
|
Sakurai H, Enoki Y. Novel aspects of heat shock factors: DNA recognition, chromatin modulation and gene expression. FEBS J 2010; 277:4140-9. [PMID: 20945530 DOI: 10.1111/j.1742-4658.2010.07829.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heat shock factor (HSF) is an evolutionarily conserved stress-response regulator that activates the transcription of heat shock protein genes, whose products maintain protein homeostasis under normal physiological conditions, as well as under conditions of stress. The promoter regions of the target genes contain a heat shock element consisting of multiple inverted repeats of the pentanucleotide sequence nGAAn. A single HSF of yeast can bind to heat shock elements that differ in the configuration of the nGAAn units and can regulate the transcription of various genes that function not only in stress resistance, but also in a broad range of biological processes. Mammalian cells have four HSF family members involved in different, but in some cases similar, biological functions, including stress resistance, cell differentiation and development. Mammalian HSF family members exhibit differential specificity for different types of heat shock elements, which, together with cell type-specific expression of HSFs is important in determining the target genes of each HSF. This minireview focuses on the molecular mechanisms of DNA recognition, chromatin modulation and gene expression by yeast and mammalian HSFs.
Collapse
Affiliation(s)
- Hiroshi Sakurai
- Department of Clinical Laboratory Science, Kanazawa University Graduate School of Medical Science, Ishikawa, Japan.
| | | |
Collapse
|