1
|
Nagai K, Nagai K, Iwaki M, Kobayashi T, Nogami A, Oka M, Saito S, Yoneda M. Frontiers of Collaboration between Primary Care and Specialists in the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease: A Review. Life (Basel) 2023; 13:2144. [PMID: 38004284 PMCID: PMC10672694 DOI: 10.3390/life13112144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease (NAFLD), is the most common liver disease. It has a rapidly growing patient population owing to the increasing prevalence of obesity and type 2 diabetes. Patients with MASLD are primarily treated by family physicians when fibrosis is absent or mild and by gastroenterologists/hepatologists when fibrosis is more advanced. It is imperative that a system for the appropriate treatment and surveillance of hepatocellular carcinoma be established in order to ensure that highly fibrotic cases are not overlooked among the large number of MASLD patients. Family physicians should check for viral hepatitis, autoimmune hepatitis, alcoholic liver disease, and drug-induced liver disease, and should evaluate fibrosis using NIT; gastroenterologists/hepatologists should perform liver biopsy, ultrasound elastography (260 units in Japan as of October 2023), and MR elastography (35 units in Japan as of October 2023). This review presents the latest findings in MASLD and the role, accuracy, and clinical use of NIT. It also describes the collaboration between Japanese primary care and gastroenterologists/hepatologists in Japan in the treatment of liver diseases, including MASLD.
Collapse
Affiliation(s)
- Koki Nagai
- Gastroenterology Division, National Hospital Organization Yokohama Medical Center, 3-60-2 Harajyuku, Totsuka-ku, Yokohama 245-8575, Japan;
| | - Kazuki Nagai
- Nagai Clinic, 1-7-25 Yokodai, Isogo-ku, Yokohama 235-0045, Japan;
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Hospital, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan; (M.I.); (T.K.); (A.N.)
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Hospital, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan; (M.I.); (T.K.); (A.N.)
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Hospital, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan; (M.I.); (T.K.); (A.N.)
| | - Masanao Oka
- OkaMedical, 1-19-18-3F Kamiookanishi, Kounan-ku, Yokohama 233-0002, Japan;
| | - Satoru Saito
- Sanno Hospital, 8-10-16 Akasaka, Minato-ku, Tokyo 107-0052, Japan;
| | - Masato Yoneda
- Gastroenterology Division, National Hospital Organization Yokohama Medical Center, 3-60-2 Harajyuku, Totsuka-ku, Yokohama 245-8575, Japan;
| |
Collapse
|
2
|
Du J, Li SK, Guan LY, Guo Z, Yin JF, Gao L, Kawanishi T, Shimada A, Zhang QP, Zheng LS, Liu YY, Feng XQ, Zhao L, Chen DY, Takeda H, Fan YB. Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos. iScience 2023; 26:107864. [PMID: 37766982 PMCID: PMC10520531 DOI: 10.1016/j.isci.2023.107864] [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: 05/09/2021] [Revised: 05/08/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The left-right symmetry breaking of vertebrate embryos requires nodal flow. However, the molecular mechanisms that mediate the asymmetric gene expression regulation under nodal flow remain elusive. Here, we report that heat shock factor 1 (HSF1) is asymmetrically activated in the Kupffer's vesicle of zebrafish embryos in the presence of nodal flow. Deficiency in HSF1 expression caused a significant situs inversus and disrupted gene expression asymmetry of nodal signaling proteins in zebrafish embryos. Further studies demonstrated that HSF1 is a mechanosensitive protein. The mechanical sensation ability of HSF1 is conserved in a variety of mechanical stimuli in different cell types. Moreover, cilia and Ca2+-Akt signaling axis are essential for the activation of HSF1 under mechanical stress in vitro and in vivo. Considering the conserved expression of HSF1 in organisms, these findings unveil a fundamental mechanism of gene expression regulation by mechanical clues during embryonic development and other physiological and pathological transformations.
Collapse
Affiliation(s)
- Jing Du
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Institute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, China
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Shu-Kai Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Liu-Yuan Guan
- Institute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, China
| | - Zheng Guo
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jiang-Fan Yin
- College of life science, Hebei Normal University, Shijiazhuang 050024, China
| | - Li Gao
- College of life science, Hebei Normal University, Shijiazhuang 050024, China
| | - Toru Kawanishi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Atsuko Shimada
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Qiu-Ping Zhang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Li-Sha Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yi-Yao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, China
| | - Lin Zhao
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Dong-Yan Chen
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Yu-Bo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| |
Collapse
|
3
|
Kovács D, Kovács M, Ahmed S, Barna J. Functional diversification of heat shock factors. Biol Futur 2022; 73:427-439. [PMID: 36402935 DOI: 10.1007/s42977-022-00138-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
Abstract
Heat shock transcription factors (HSFs) are widely known as master regulators of the heat shock response. In invertebrates, a single heat shock factor, HSF1, is responsible for the maintenance of protein homeostasis. In vertebrates, seven members of the HSF family have been identified, namely HSF1, HSF2, HSF3, HSF4, HSF5, HSFX, and HSFY, of which HSF1 and HSF2 are clearly associated with heat shock response, while HSF4 is involved in development. Other members of the family have not yet been studied as extensively. Besides their role in cellular proteostasis, HSFs influence a plethora of biological processes such as aging, development, cell proliferation, and cell differentiation, and they are implicated in several pathologies such as neurodegeneration and cancer. This is achieved by regulating the expression of a great variety of genes including chaperones. Here, we review our current knowledge on the function of HSF family members and important aspects that made possible the functional diversification of HSFs.
Collapse
Affiliation(s)
- Dániel Kovács
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - Márton Kovács
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - Saqib Ahmed
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - János Barna
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary. .,ELKH-ELTE Genetics Research Group, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary.
| |
Collapse
|
4
|
Lakhotia SC. Delayed discovery of Hsp60 and subsequent characterization of moonlighting functions of multiple Hsp60 genes in Drosophila: a personal historical perspective. J Genet 2022. [DOI: 10.1007/s12041-022-01389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
5
|
Morphis AC, Edwards SL, Erdenebat P, Kumar L, Li J. Auxin-Inducible Degron System Reveals Temporal-Spatial Roles of HSF-1 and Its Transcriptional Program in Lifespan Assurance. FRONTIERS IN AGING 2022; 3:899744. [PMID: 35899092 PMCID: PMC9309338 DOI: 10.3389/fragi.2022.899744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022]
Abstract
HSF-1 is a key regulator of cellular proteotoxic stress response and is required for animal lifespan. In C. elegans, HSF-1 mediated heat shock response (HSR) declines sharply on the first day of adulthood, and HSF-1 was proposed to function primarily during larval stages for lifespan assurance based on studies using RNAi. The tissue requirement for HSF-1 in lifespan, however, is not well understood. Using the auxin-inducible degron (AID) system, we manage to uncouple the roles of HSF-1 in development and longevity. In wild-type animals, we find HSF-1 is required during the whole self-reproductive period for lifespan. This period is extended in long-lived animals that have arrested germline stem cells (GSC) or reduced insulin/IGF-1 signaling (IIS). While depletion of HSF-1 from any major somatic tissues during development results in severe defects, HSF-1 primarily functions in the intestine and likely neural system of adults to support lifespan. Finally, by combining AID and genome-wide transcriptional analyses, we find HSF-1 directly activates the transcription of constitutively-expressed chaperone and co-chaperone genes among others in early adulthood, which underlies its roles in longevity assurance.
Collapse
Affiliation(s)
| | | | | | | | - Jian Li
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, United States
| |
Collapse
|
6
|
Singh G, Chakraborty S, Lakhotia SC. Elevation of major constitutive heat shock proteins is heat shock factor independent and essential for establishment and growth of Lgl loss and Yorkie gain-mediated tumors in Drosophila. Cell Stress Chaperones 2022; 27:431-448. [PMID: 35704239 PMCID: PMC9346025 DOI: 10.1007/s12192-022-01283-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 01/03/2023] Open
Abstract
Cancer cells generally overexpress heat shock proteins (Hsps), the major components of cellular stress response, to overcome and survive the diverse stresses. However, the specific roles of Hsps in initiation and establishment of cancers remain unclear. Using loss of Lgl-mediated epithelial tumorigenesis in Drosophila, we induced tumorigenic somatic clones of different genetic backgrounds to examine the temporal and spatial expression and roles of major heat shock proteins in tumor growth. The constitutively expressed Hsp83, Hsc70 (heat shock cognate), Hsp60 and Hsp27 show elevated levels in all cells of the tumorigenic clone since early stages, which persists till their transformation. However, the stress-inducible Hsp70 is expressd only in a few cells at later stage of established tumorous clones that show high F-actin aggregation. Intriguingly, levels of heat shock factor (HSF), the master regulator of Hsps, remain unaltered in these tumorous cells and its down-regulation does not affect tumorigenic growth of lgl- clones overexpressing Yorkie, although down-regulation of Hsp83 prevents their survival and growth. Interestingly, overexpression of HSF or Hsp83 in lgl- cells makes them competitively successful in establishing tumorous clones. These results show that the major constitutively expressed Hsps, but not the stress-inducible Hsp70, are involved in early as well as late stages of epithelial tumors and their elevated expression in lgl- clones co-overexpressing Yorkie is independent of HSF.
Collapse
Affiliation(s)
- Gunjan Singh
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| | - Saptomee Chakraborty
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
- Present Address: Department of Biosciences & Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Subhash C. Lakhotia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
7
|
Shih SR, Bach DM, Rondeau NC, Sam J, Lovinger NL, Lopatkin AJ, Snow JW. Honey bee sHSP are responsive to diverse proteostatic stresses and potentially promising biomarkers of honey bee stress. Sci Rep 2021; 11:22087. [PMID: 34764357 PMCID: PMC8586346 DOI: 10.1038/s41598-021-01547-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 10/29/2021] [Indexed: 11/09/2022] Open
Abstract
The pollination services provided by the honey bee are critical in both natural and agricultural ecosystems. Honey bee colonies in the United States have suffered from an increased rate of die-off in recent years, stemming from a complex set of interacting stresses that remain poorly described. Defining specific common cellular processes and cellular stress responses impacted by multiple stressors represent a key step in understanding these synergies. Proteotoxic stresses negatively impact protein synthesis, folding, and degradation. Diverse proteotoxic stresses induce expression of genes encoding small heat shock proteins (sHSP) of the expanded lethal (2) essential for life (l(2)efl) gene family. In addition to upregulation by the Integrated Stress Response (ISR), the Heat Shock Response (HSR), and the Oxidative Stress Response (OSR), our data provide first evidence that sHSP genes are upregulated by the Unfolded Protein Response (UPR). As these genes appear to be part of a core stress response that could serve as a useful biomarker for cellular stress in honey bees, we designed and tested an RT-LAMP assay to detect increased l(2)efl gene expression in response to heat-stress. While this assay provides a powerful proof of principle, further work will be necessary to link changes in sHSP gene expression to colony-level outcomes, to adapt our preliminary assay into a Point of Care Testing (POCT) assay appropriate for use as a diagnostic tool for use in the field, and to couple assay results to management recommendations.
Collapse
Affiliation(s)
- Samantha R Shih
- Biology Department, Barnard College, New York, NY, 10027, USA
| | - Dunay M Bach
- Biology Department, Barnard College, New York, NY, 10027, USA
| | | | - Jessica Sam
- Biology Department, Barnard College, New York, NY, 10027, USA
| | | | | | - Jonathan W Snow
- Biology Department, Barnard College, New York, NY, 10027, USA.
| |
Collapse
|
8
|
Bach DM, Holzman MA, Wague F, Miranda JL, Lopatkin AJ, Mansfield JH, Snow JW. Thermal stress induces tissue damage and a broad shift in regenerative signaling pathways in the honey bee digestive tract. J Exp Biol 2021; 224:272039. [PMID: 34477881 DOI: 10.1242/jeb.242262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/19/2021] [Indexed: 11/20/2022]
Abstract
Honey bee colonies in the USA have suffered from increased die-off in the last few years with a complex set of interacting stresses playing a key role. With changing climate, an increase in the frequency of severe weather events, such as heat waves, is anticipated. Understanding how these changes may contribute to stress in honey bees is crucial. Individual honey bees appear to have a high capacity to endure thermal stress. One reason for this high-level endurance is likely their robust heat shock response (HSR), which contributes to thermotolerance at the cellular level. However, less is known about other mechanisms of thermotolerance, especially those operating at the tissue level. To elucidate other determinants of resilience in this species, we used thermal stress coupled with RNAseq and identified broad transcriptional remodeling of a number of key signaling pathways in the honey bee, including those pathways known to be involved in digestive tract regeneration in the fruit fly such as the Hippo and JAK/STAT pathways. We also observed cell death and shedding of epithelial cells, which likely leads to induction of this regenerative transcriptional program. We found that thermal stress affects many of these pathways in other tissues, suggesting a shared program of damage response. This study provides important foundational characterization of the tissue damage response program in this key pollinating species. In addition, our data suggest that a robust regeneration program may also be a critical contributor to thermotolerance at the tissue level, a possibility which warrants further exploration in this and other species.
Collapse
Affiliation(s)
- Dunay M Bach
- Biology Department, Barnard College, New York, NY 10027, USA
| | | | - Fatoumata Wague
- Biology Department, Barnard College, New York, NY 10027, USA
| | - Jj L Miranda
- Biology Department, Barnard College, New York, NY 10027, USA
| | - Allison J Lopatkin
- Biology Department, Barnard College, New York, NY 10027, USA.,Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY 10027, USA.,Data Science Institute , Columbia University, New York, NY 10027, USA
| | | | - Jonathan W Snow
- Biology Department, Barnard College, New York, NY 10027, USA
| |
Collapse
|
9
|
The transcriptomic response of adult salmon lice (Lepeophtheirus salmonis) to reduced salinity. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 37:100778. [PMID: 33271493 DOI: 10.1016/j.cbd.2020.100778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 11/22/2022]
Abstract
Salmon lice (Lepeophtheirus salmonis) are marine parasitic copepods living on salmonids and are challenging for salmon aquaculture. One of several treatment methods is the application of freshwater to the fish which can lead to lice loss. However, lab experiments have shown that salmon lice, acclimated to seawater, are capable of surviving for several weeks in freshwater, when attached to a host. If not attached to a host, they die within a few hours in freshwater but can survive a longer time in brackish water. The molecular mechanisms involved in the adaptation to low salinity of the louse have not been identified yet. In this study we incubated salmon lice, being attached to a host, or detached, in seawater, brackish water and freshwater for 4 h and 1 d, sampled the animals and used RNA-Seq to identify genes involved in these mechanisms. Freshwater incubation led to a much stronger regulatory response than brackish water and a longer incubation time gave a stronger effect than a short incubation. Among the most interesting genes, upregulated in low salinity water are in addition to several transporters, several enzymes involved in amino acid metabolism and especially in the proline biosynthesis. A strong upregulation of these enzymes might lead to an accumulation of proline which is known to be used as an osmolyte in other species. While the RNA-Seq experiment was performed with female samples, qPCR showed that at least 10 genes regulated in females, were also regulated in males.
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
|
Joutsen J, Sistonen L. Tailoring of Proteostasis Networks with Heat Shock Factors. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034066. [PMID: 30420555 DOI: 10.1101/cshperspect.a034066] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heat shock factors (HSFs) are the main transcriptional regulators of the heat shock response and indispensable for maintaining cellular proteostasis. HSFs mediate their protective functions through diverse genetic programs, which are composed of genes encoding molecular chaperones and other genes crucial for cell survival. The mechanisms that are used to tailor HSF-driven proteostasis networks are not yet completely understood, but they likely comprise from distinct combinations of both genetic and proteomic determinants. In this review, we highlight the versatile HSF-mediated cellular functions that extend from cellular stress responses to various physiological and pathological processes, and we underline the key advancements that have been achieved in the field of HSF research during the last decade.
Collapse
Affiliation(s)
- Jenny Joutsen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| |
Collapse
|
12
|
Abstract
Protein homeostasis, or proteostasis, is required for proper cell function and thus must be
under tight maintenance in all circumstances. In crowded cell conditions, protein folding is sometimes
unfavorable, and this condition is worsened during stress situations. Cells cope with such stress
through the use of a Protein Quality Control system, which uses molecular chaperones and heat shock
proteins as its major players. This system aids with folding, avoiding misfolding and/or reversing aggregation.
A pivotal regulator of the response to heat stress is Heat Shock Factor, which is recruited to
the promoters of the chaperone genes, inducting their expression. This mini review aims to cover our
general knowledge on the structure and function of this factor.
Collapse
Affiliation(s)
- Natália Galdi Quel
- Institute of Chemistry and Institute of Biology, University of Campinas - UNICAMP, Campinas, Brazil
| | - Carlos H.I. Ramos
- Institute of Chemistry and Institute of Biology, University of Campinas - UNICAMP, Campinas, Brazil
| |
Collapse
|
13
|
Rolandi C, Lighton JRB, de la Vega GJ, Schilman PE, Mensch J. Genetic variation for tolerance to high temperatures in a population of Drosophila melanogaster. Ecol Evol 2018; 8:10374-10383. [PMID: 30464811 PMCID: PMC6238130 DOI: 10.1002/ece3.4409] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 07/01/2018] [Accepted: 07/02/2018] [Indexed: 12/26/2022] Open
Abstract
The range of thermal tolerance is one of the main factors influencing the geographic distribution of species. Climate change projections predict increases in average and extreme temperatures over the coming decades; hence, the ability of living beings to resist these changes will depend on physiological and adaptive responses. On an evolutionary scale, changes will occur as the result of selective pressures on individual heritable differences. In this work, we studied the genetic basis of tolerance to high temperatures in the fly Drosophila melanogaster and whether this species presents sufficient genetic variability to allow expansion of its upper thermo-tolerance limit. To do so, we used adult flies derived from a natural population belonging to the Drosophila Genetic Reference Panel, for which genomic sequencing data are available. We characterized the phenotypic variation of the upper thermal limit in 34 lines by measuring knockdown temperature (i.e., critical thermal maximum [CTmax]) by exposing flies to a ramp of increasing temperature (0.25°C/min). Fourteen percent of the variation in CTmax is explained by the genetic variation across lines, without a significant sexual dimorphism. Through a genomewide association study, 12 single nucleotide polymorphisms associated with the CTmax were identified. In most of these SNPs, the less frequent allele increased the upper thermal limit suggesting that this population harbors raw genetic variation capable of expanding its heat tolerance. This potential upper thermal tolerance increase has implications under the global warming scenario. Past climatic records show a very low incidence of days above CTmax (10 days over 25 years); however, future climate scenarios predict 243 days with extreme high temperature above CTmax from 2045 to 2070. Thus, in the context of the future climate warming, rising temperatures might drive the evolution of heat tolerance in this population by increasing the frequency of the alleles associated with higher CTmax.
Collapse
Affiliation(s)
- Carmen Rolandi
- IBBEA‐CONICET‐UBA. DBBEAFacultad de Ciencias Exactas y NaturalesUniversidad de Buenos AiresBuenos AiresArgentina
| | | | - Gerardo J. de la Vega
- Grupo de Ecología de Poblaciones de Insectos (GEPI)INTA EEA BarilocheBarilocheArgentina
| | - Pablo E. Schilman
- IBBEA‐CONICET‐UBA. DBBEAFacultad de Ciencias Exactas y NaturalesUniversidad de Buenos AiresBuenos AiresArgentina
| | - Julián Mensch
- IEGEBA‐CONICET‐UBADEGEFacultad de Ciencias Exactas y NaturalesUniversidad de Buenos AiresBuenos AiresArgentina
| |
Collapse
|
14
|
Barna J, Csermely P, Vellai T. Roles of heat shock factor 1 beyond the heat shock response. Cell Mol Life Sci 2018; 75:2897-2916. [PMID: 29774376 PMCID: PMC11105406 DOI: 10.1007/s00018-018-2836-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/07/2018] [Indexed: 01/09/2023]
Abstract
Various stress factors leading to protein damage induce the activation of an evolutionarily conserved cell protective mechanism, the heat shock response (HSR), to maintain protein homeostasis in virtually all eukaryotic cells. Heat shock factor 1 (HSF1) plays a central role in the HSR. HSF1 was initially known as a transcription factor that upregulates genes encoding heat shock proteins (HSPs), also called molecular chaperones, which assist in refolding or degrading injured intracellular proteins. However, recent accumulating evidence indicates multiple additional functions for HSF1 beyond the activation of HSPs. Here, we present a nearly comprehensive list of non-HSP-related target genes of HSF1 identified so far. Through controlling these targets, HSF1 acts in diverse stress-induced cellular processes and molecular mechanisms, including the endoplasmic reticulum unfolded protein response and ubiquitin-proteasome system, multidrug resistance, autophagy, apoptosis, immune response, cell growth arrest, differentiation underlying developmental diapause, chromatin remodelling, cancer development, and ageing. Hence, HSF1 emerges as a major orchestrator of cellular stress response pathways.
Collapse
Affiliation(s)
- János Barna
- Department of Genetics, Eötvös Loránd University, Pázmány Péter Stny. 1/C, Budapest, 1117, Hungary
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Péter Csermely
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter Stny. 1/C, Budapest, 1117, Hungary.
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
15
|
|
16
|
Liaw GJ. Pits, a protein interacting with Ttk69 and Sin3A, has links to histone deacetylation. Sci Rep 2016; 6:33388. [PMID: 27622813 PMCID: PMC5020733 DOI: 10.1038/srep33388] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/25/2016] [Indexed: 01/09/2023] Open
Abstract
Histone deacetylation plays an important role in transcriptional repression. Previous results showed that the genetic interaction between ttk and rpd3, which encodes a class I histone deacetylase, is required for tll repression. This study investigated the molecular mechanism by which Ttk69 recruits Rpd3. Using yeast two-hybrid screening and datamining, one novel protein was found that weakly interacts with Ttk69 and Sin3A, designated as Protein interacting with Ttk69 and Sin3A (Pits). Pits protein expressed in the early stages of embryos and bound to the region of the tor response element in vivo. Expanded tll expression patterns were observed in embryos lacking maternal pits activity and the expansion was not widened by reducing either maternal ttk or sin3A activity. However, in embryos with simultaneously reduced maternal pits and sin3A activities or maternal pits, sin3A and ttk activities, the proportions of the embryos with expanded tll expression were significantly increased. These results indicate that all three gene activities are involved in tll repression. Level of histone H3 acetylation in the tll proximal region was found to be elevated in embryo with reduced these three gene activities. In conclusion, Ttk69 causes the histone deacetylation-mediated repression of tll via the interaction of Pits and Sin3A.
Collapse
Affiliation(s)
- Gwo-Jen Liaw
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, 112-22, Taiwan, Republic of China
| |
Collapse
|
17
|
The heat shock response restricts virus infection in Drosophila. Sci Rep 2015; 5:12758. [PMID: 26234525 PMCID: PMC4522674 DOI: 10.1038/srep12758] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/09/2015] [Indexed: 01/02/2023] Open
Abstract
Innate immunity is the first line of defence against pathogens and is essential for survival of the infected host. The fruit fly Drosophila melanogaster is an emerging model to study viral pathogenesis, yet antiviral defence responses remain poorly understood. Here, we describe the heat shock response, a cellular mechanism that prevents proteotoxicity, as a component of the antiviral immune response in Drosophila. Transcriptome analyses of Drosophila S2 cells and adult flies revealed strong induction of the heat shock response upon RNA virus infection. Dynamic induction patterns of heat shock pathway components were characterized in vitro and in vivo following infection with different classes of viruses. The heat shock transcription factor (Hsf), as well as active viral replication, were necessary for the induction of the response. Hsf-deficient adult flies were hypersensitive to virus infection, indicating a role of the heat shock response in antiviral defence. In accordance, transgenic activation of the heat shock response prolonged survival time after infection and enabled long-term control of virus replication to undetectable levels. Together, our results establish the heat shock response as an important constituent of innate antiviral immunity in Drosophila.
Collapse
|
18
|
Vydra N, Toma A, Widlak W. Pleiotropic role of HSF1 in neoplastic transformation. Curr Cancer Drug Targets 2015; 14:144-55. [PMID: 24467529 PMCID: PMC4435066 DOI: 10.2174/1568009614666140122155942] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 01/06/2014] [Accepted: 01/22/2014] [Indexed: 01/13/2023]
Abstract
HSF1 (Heat Shock transcription Factor 1) is the main transcription factor activated in response to proteotoxic stress. Once activated, it induces an expression of heat shock proteins (HSPs) which enables cells to survive in suboptimal conditions. HSF1 could be also activated by altered kinase signaling characteristic for cancer cells, which is a probable reason for its high activity found in a broad range of tumors. There is rapidly growing evidence that HSF1 supports tumor initiation and growth, as well as metastasis and angiogenesis. It also modulates the sensitivity of cancer cells to therapy. Functions of HSF1 in cancer are connected with HSPs’ activity, which generally protects cells from apoptosis, but also are independent of its classical targets. HSF1-dependent regulation of non-HSPs genes plays a role in cell cycle
progression, glucose metabolism, autophagy and drug efflux. HSF1 affects the key cell-survival and regulatory pathways, including p53, RAS/MAPK, cAMP/PKA, mTOR and insulin signaling. Although the exact mechanism of HSF1 action is still somewhat obscure, HSF1 is becoming an attractive target in anticancer therapies, whose inhibition could enhance the effects of other treatments.
Collapse
Affiliation(s)
| | | | - Wieslawa Widlak
- Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-101 Gliwice, Poland.
| |
Collapse
|
19
|
Drosophila Small Heat Shock Proteins: An Update on Their Features and Functions. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_25] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Magbanua JP, Runneburger E, Russell S, White R. A variably occupied CTCF binding site in the ultrabithorax gene in the Drosophila bithorax complex. Mol Cell Biol 2015; 35:318-30. [PMID: 25368383 PMCID: PMC4295388 DOI: 10.1128/mcb.01061-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/10/2014] [Accepted: 10/25/2014] [Indexed: 11/20/2022] Open
Abstract
Although the majority of genomic binding sites for the insulator protein CCCTC-binding factor (CTCF) are constitutively occupied, a subset show variable occupancy. Such variable sites provide an opportunity to assess context-specific CTCF functions in gene regulation. Here, we have identified a variably occupied CTCF site in the Drosophila Ultrabithorax (Ubx) gene. This site is occupied in tissues where Ubx is active (third thoracic leg imaginal disc) but is not bound in tissues where the Ubx gene is repressed (first thoracic leg imaginal disc). Using chromatin conformation capture, we show that this site preferentially interacts with the Ubx promoter region in the active state. The site lies close to Ubx enhancer elements and is also close to the locations of several gypsy transposon insertions that disrupt Ubx expression, leading to the bx mutant phenotype. gypsy insertions carry the Su(Hw)-dependent gypsy insulator and were found to affect both CTCF binding at the variable site and the chromatin topology. This suggests that insertion of the gypsy insulator in this region interferes with CTCF function and supports a model for the normal function of the variable CTCF site as a chromatin loop facilitator, promoting interaction between Ubx enhancers and the Ubx transcription start site.
Collapse
Affiliation(s)
- Jose Paolo Magbanua
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Estelle Runneburger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Steven Russell
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Robert White
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Telonis-Scott M, Clemson AS, Johnson TK, Sgrò CM. Spatial analysis of gene regulation reveals new insights into the molecular basis of upper thermal limits. Mol Ecol 2014; 23:6135-51. [PMID: 25401770 DOI: 10.1111/mec.13000] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/06/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022]
Abstract
The cellular stress response has long been the primary model for studying the molecular basis of thermal adaptation, yet the link between gene expression, RNA metabolism and physiological responses to thermal stress remains largely unexplored. We address this by comparing the transcriptional and physiological responses of three geographically distinct populations of Drosophila melanogaster from eastern Australia in response to, and recovery from, a severe heat stress with and without a prestress hardening treatment. We focus on starvin (stv), recently identified as an important thermally responsive gene. Intriguingly, stv encodes seven transcripts from alternative transcription sites and alternative splicing, yet appears to be rapidly heat inducible. First, we show genetic differences in upper thermal limits of the populations tested. We then demonstrate that the stv locus does not ubiquitously respond to thermal stress but is expressed as three distinct thermal and temporal RNA phenotypes (isoforms). The shorter transcript isoforms are rapidly upregulated under stress in all populations and show similar molecular signatures to heat-shock proteins. Multiple stress exposures seem to generate a reserve of pre-mRNAs, effectively 'priming' the cells for subsequent stress. Remarkably, we demonstrate a bypass in the splicing blockade in these isoforms, suggesting an essential role for these transcripts under heat stress. Temporal profiles for the weakly heat responsive stv isoform subset show opposing patterns in the two most divergent populations. Innate and induced transcriptome responses to hyperthermia are complex, and warrant moving beyond gene-level analyses.
Collapse
Affiliation(s)
- Marina Telonis-Scott
- School of Biological Sciences, Monash University, Clayton, Vic., 3800, Australia
| | | | | | | |
Collapse
|
22
|
New levels of transcriptome complexity at upper thermal limits in wild Drosophila revealed by exon expression analysis. Genetics 2013; 195:809-30. [PMID: 24002645 DOI: 10.1534/genetics.113.156224] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
While the cellular heat-shock response has been a paradigm for studying the impact of thermal stress on RNA metabolism and gene expression, the genome-wide response to thermal stress and its connection to physiological stress resistance remain largely unexplored. Here, we address this issue using an array-based exon expression analysis to interrogate the transcriptome in recently established Drosophila melanogaster stocks during severe thermal stress and recovery. We first demonstrated the efficacy of exon-level analyses to reveal a level of thermally induced transcriptome complexity extending well beyond gene-level analyses. Next, we showed that the upper range of both the cellular and physiological thermal stress response profoundly affected message expression and processing in D. melanogaster, limiting expression to a small subset of transcripts, many that share features of known rapidly responding stress genes. As predicted from cellular heat-shock research, constitutive splicing was blocked in a set of novel genes; we did not detect changes to alternative splicing during heat stress, but rather induction of intronless isoforms of known heat-responsive genes. We observed transcriptome plasticity in the form of differential isoform expression during recovery from heat shock, mediated by multiple mechanisms including alternative transcription and alternative splicing. This affected genes involved in DNA regulation, immune response, and thermotolerance. These patterns highlight the complex nature of innate transcriptome responses under stress and potential for adaptive shifts through plasticity and evolved genetic responses at different hierarchical levels.
Collapse
|
23
|
Piñeyro D, Blanch M, Badal M, Kosoy A, Bernués J. GAGA factor repression of transcription is a rare event but the negative regulation of Trl is conserved in Drosophila species. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1056-65. [PMID: 23860261 DOI: 10.1016/j.bbagrm.2013.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/02/2013] [Accepted: 07/08/2013] [Indexed: 10/26/2022]
Abstract
GAGA is a highly conserved Drosophila transcription factor encoded by the Trithorax-like (Trl) gene. While GAGA usually activates transcription, it represses its own promoter. Here we show that GAGA-mediated repression of Trl is conserved between two distant Drosophila species. A detailed promoter study showed that GAGA repressive activity can't be attributed to any discrete element in the Trl promoter. Genome-wide analysis of the transcriptome in S2 cells indicated that repression of Trl is very likely unique, being GAGA factor a transactivator for all the other promoters. Taken together, our results suggest a new mechanism to explain GAGA-mediated repression that involves a dose-dependent change in the architecture of the Trl promoter.
Collapse
Affiliation(s)
- David Piñeyro
- Departament. de Genòmica Molecular, Institut de Biologia Molecular de Barcelona-CSIC, Parc Científic de Barcelona, 08028 Barcelona, Spain; Cell and Developmental Biology Programme, Institute for Research in Biomedicine, Barcelona, Spain
| | | | | | | | | |
Collapse
|
24
|
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
|
25
|
Pakravan N. Tumorigenesis: cell defense against hypoxia? Oncol Rev 2013; 7:e1. [PMID: 25992222 PMCID: PMC4419620 DOI: 10.4081/oncol.2013.e1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/02/2013] [Accepted: 04/02/2013] [Indexed: 01/01/2023] Open
Abstract
Microenvironmental elements can directly contribute to the induction and the maintenance of tumor. Oxygen is the main element in the cell microenvironment and hypoxia can affect the process of tumorigenesis. In response to hypoxia, cells change their pattern and characteristics. These changes suggest that it is not just adaptation, but some sort of cell defense against hypoxia. If hypoxia is corrected, then cell defense mechanisms are interrupted. An examination of the process of tumorigenesis helps to design better therapeutic strategies. A systematic review of the English literature was conducted by searching PubMed, Google Scholar, and ISI Web databases for studies on changes that defend and help cells to live in a hypoxic microenvironment. Cells respond to hypoxia by de-differentiation and an increase in heat shock proteins. Angiogenesis and deviation of inflammatory response in favor of hypoxic cell survival also defend and save the oxygen-starved cells from death. Finally, anti-angiogenic therapies and more hypoxia enhance metastasis, as tumors with low oxygen concentration are more malignant than tumors with high oxygen concentration. All these enable cells to migrate away from low oxygen areas and seek a more conducive microenvironment. Therapies that make the microenvironment more hypoxic need to be revised. This has been done for anti-angiogenic therapies, previously considered to be anti-tumor approaches. Effective therapies may be correcting therapies which direct the tumor microenvironment towards natural physical/chemical condition. Correcting therapies either bring back tumor cells to a normal form (correct tumor cells) or help the immune system to eradicate tumor cells which can not be corrected.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Department of Pathobiology, Medical School, Alborz University of Medical Sciences , Karaj, Iran
| |
Collapse
|
26
|
Shen SP, Aleksic J, Russell S. Identifying targets of the Sox domain protein Dichaete in the Drosophila CNS via targeted expression of dominant negative proteins. BMC DEVELOPMENTAL BIOLOGY 2013; 13:1. [PMID: 23289785 PMCID: PMC3541953 DOI: 10.1186/1471-213x-13-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 01/03/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Group B Sox domain transcription factors play important roles in metazoan central nervous system development. They are, however, difficult to study as mutations often have pleiotropic effects and other Sox family members can mask phenotypes due to functional compensation. In Drosophila melanogaster, the Sox gene Dichaete is dynamically expressed in the embryonic CNS, where it is known to have functional roles in neuroblasts and the ventral midline. In this study, we use inducible dominant negative proteins in combination with ChIP, immunohistochemistry and genome-wide expression profiling to further dissect the role of Dichaete in these two tissues. RESULTS We generated two dominant negative Dichaete constructs, one lacking a DNA binding domain and the other fused to the Engrailed transcriptional repressor domain. We expressed these tissue-specifically in the midline and in neuroblasts using the UAS/GAL4 system, validating their use at the phenotypic level and with known target genes. Using ChIP and immunohistochemistry, we identified two new likely direct Dichaete target genes, commisureless in the midline and asense in the neuroectoderm. We performed genome-wide expression profiling in stage 8-9 embryos, identifying almost a thousand potential tissue-specific Dichaete targets, with half of these genes showing evidence of Dichaete binding in vivo. These include a number of genes with known roles in CNS development, including several components of the Notch, Wnt and EGFR signalling pathways. CONCLUSIONS As well as identifying commisureless as a target, our data indicate that Dichaete helps establish its expression during early midline development but has less effect on its established later expression, highlighting Dichaete action on tissue specific enhancers. An analysis of the broader range of candidate Dichaete targets indicates that Dichaete plays diverse roles in CNS development, with the 500 or so Dichaete-bound putative targets including a number of transcription factors, signalling pathway components and terminal differentiation genes. In the early neurectoderm we implicate Dichaete in the lateral inhibition pathway and show that Dichaete acts to repress the proneural gene asense. Our analysis also reveals that dominant negatives cause off-target effects, highlighting the need to use other experimental data for validating findings from dominant negative studies.
Collapse
Affiliation(s)
- Shih Pei Shen
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
27
|
Dai C, Dai S, Cao J. Proteotoxic stress of cancer: implication of the heat-shock response in oncogenesis. J Cell Physiol 2012; 227:2982-7. [PMID: 22105155 DOI: 10.1002/jcp.24017] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Organisms frequently encounter a wide variety of proteotoxic stressors. The heat-shock response, an ancient cytoprotective mechanism, has evolved to augment organismal survival and longevity in the face of proteotoxic stress from without and within. These broadly recognized beneficial effects, ironically, contrast sharply with its emerging role as a culprit in the pathogenesis of cancers. Here, we present an overview of the normal biology of the heat-shock response and highlight its implications in oncogenic processes, including the proteotoxic stress phenotype of cancer; the function of this stress response in helping cancer survive and adapt to proteotoxic stress; and perturbation of proteome homeostasis in cancer as a potential therapeutic avenue.
Collapse
Affiliation(s)
- Chengkai Dai
- The Jackson Laboratory, Bar Harbor, ME 04609, USA.
| | | | | |
Collapse
|
28
|
Riva L, Koeva M, Yildirim F, Pirhaji L, Dinesh D, Mazor T, Duennwald ML, Fraenkel E. Poly-glutamine expanded huntingtin dramatically alters the genome wide binding of HSF1. J Huntingtons Dis 2012; 1:33-45. [PMID: 23293686 PMCID: PMC3537492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
In Huntington's disease (HD), polyglutamine expansions in the huntingtin (Htt) protein cause subtle changes in cellular functions that, over-time, lead to neurodegeneration and death. Studies have indicated that activation of the heat shock response can reduce many of the effects of mutant Htt in disease models, suggesting that the heat shock response is impaired in the disease. To understand the basis for this impairment, we have used genome-wide chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-Seq) to examine the effects of mutant Htt on the master regulator of the heat shock response, HSF1. We find that, under normal conditions, HSF1 function is highly similar in cells carrying either wild-type or mutant Htt. However, polyQ-expanded Htt severely blunts the HSF1-mediated stress response. Surprisingly, we find that the HSF1 targets most affected upon stress are not directly associated with proteostasis, but with cytoskeletal binding, focal adhesion and GTPase activity. Our data raise the intriguing hypothesis that the accumulated damage from life-long impairment in these stress responses may contribute significantly to the etiology of Huntington's disease.
Collapse
Affiliation(s)
- Laura Riva
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Martina Koeva
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
- The Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Ferah Yildirim
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Leila Pirhaji
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Deepika Dinesh
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Tali Mazor
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | | | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| |
Collapse
|
29
|
Tucker NR, Middleton RC, Le QP, Shelden EA. HSF1 is essential for the resistance of zebrafish eye and brain tissues to hypoxia/reperfusion injury. PLoS One 2011; 6:e22268. [PMID: 21814572 PMCID: PMC3141033 DOI: 10.1371/journal.pone.0022268] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 06/21/2011] [Indexed: 12/21/2022] Open
Abstract
Ischemia and subsequent reperfusion (IR) produces injury to brain, eye and other tissues, contributing to the progression of important clinical pathologies. The response of cells to IR involves activation of several signaling pathways including those activating hypoxia and heat shock responsive transcription factors. However, specific roles of these responses in limiting cell damage and preventing cell death after IR have not been fully elucidated. Here, we have examined the role of heat shock factor 1 (HSF1) in the response of zebrafish embryos to hypoxia and subsequent return to normoxic conditions (HR) as a model for IR. Heat shock preconditioning elevated heat shock protein expression and protected zebrafish embryo eye and brain tissues against HR-induced apoptosis. These effects were inhibited by translational suppression of HSF1 expression. Reduced expression of HSF1 also increased cell death in brain and eye tissues of embryos subjected to hypoxia and reperfusion without prior heat shock. Surprisingly, reduced expression of HSF1 had only a modest effect on hypoxia-induced expression of Hsp70 and no effect on hypoxia-induced expression of Hsp27. These results establish the zebrafish embryo as a model for the study of ischemic injury in the brain and eye and reveal a critical role for HSF1 in the response of these tissues to HR. Our results also uncouple the role of HSF1 expression from that of Hsp27, a well characterized heat shock protein considered essential for cell survival after hypoxia. Alternative roles for HSF1 are considered.
Collapse
Affiliation(s)
- Nathan R. Tucker
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Ryan C. Middleton
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Quynh P. Le
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Eric A. Shelden
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
- Center for Reproductive Biology, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
30
|
Liu Z, Lin X, Cai Z, Zhang Z, Han C, Jia S, Meng A, Wang Q. Global identification of SMAD2 target genes reveals a role for multiple co-regulatory factors in zebrafish early gastrulas. J Biol Chem 2011; 286:28520-32. [PMID: 21669877 PMCID: PMC3151094 DOI: 10.1074/jbc.m111.236307] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nodal and Smad2/3 signals play pivotal roles in mesendoderm induction and axis determination during late blastulation and early gastrulation in vertebrate embryos. However, Smad2/3 direct target genes during those critical developmental stages have not been systematically identified. Here, through ChIP-chip assay, we show that the promoter/enhancer regions of 679 genes are bound by Smad2 in the zebrafish early gastrulas. Expression analyses confirm that a significant proportion of Smad2 targets are indeed subjected to Nodal/Smad2 regulation at the onset of gastrulation. The co-existence of DNA-binding sites of other transcription factors in the Smad2-bound regions allows the identification of well known Smad2-binding partners, such as FoxH1 and Lef1/β-catenin, as well as many previously unknown Smad2 partners, including Oct1 and Gata6, during embryogenesis. We demonstrate that Oct1 physically associates with and enhances the transcription and mesendodermal induction activity of Smad2, whereas Gata6 exerts an inhibitory role in Smad2 signaling and mesendodermal induction. Thus, our study systemically uncovers a large number of Smad2 targets in early gastrulas and suggests cooperative roles of Smad2 and other transcription factors in controlling target gene transcription, which will be valuable for studying regulatory cascades during germ layer formation and patterning of vertebrate embryos.
Collapse
Affiliation(s)
- Zhaoting Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Choo SW, White R, Russell S. Genome-wide analysis of the binding of the Hox protein Ultrabithorax and the Hox cofactor Homothorax in Drosophila. PLoS One 2011; 6:e14778. [PMID: 21483667 PMCID: PMC3071696 DOI: 10.1371/journal.pone.0014778] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/15/2011] [Indexed: 12/15/2022] Open
Abstract
Hox genes encode a family of transcription factors that are key developmental regulators with a highly conserved role in specifying segmental diversity along the metazoan body axis. Although they have been shown to regulate a wide variety of downstream processes, direct transcriptional targets have been difficult to identify and this has been a major obstacle to our understanding of Hox gene function. We report the identification of genome-wide binding sites for the Hox protein Ultrabithorax (Ubx) using a YFP-tagged Drosophila protein-trap line together with chromatin immunoprecipitation and microarray analysis. We identify 1,147 genes bound by Ubx at high confidence in chromatin from the haltere imaginal disc, a prominent site of Ubx function where it specifies haltere versus wing development. The functional relevance of these genes is supported by their overlap with genes differentially expressed between wing and haltere imaginal discs. The Ubx-bound gene set is highly enriched in genes involved in developmental processes and contains both high-level regulators as well as genes involved in more basic cellular functions. Several signalling pathways are highly enriched in the Ubx target gene set and our analysis supports the view that Hox genes regulate many levels of developmental pathways and have targets distributed throughout the gene network. We also performed genome-wide analysis of the binding sites for the Hox cofactor Homothorax (Hth), revealing a striking similarity with the Ubx binding profile. We suggest that these binding profiles may be strongly influenced by chromatin accessibility and provide evidence of a link between Ubx/Hth binding and chromatin state at genes regulated by Polycomb silencing. Overall, we define a set of direct Ubx targets in the haltere imaginal disc and suggest that chromatin accessibility has important implications for Hox target selection and for transcription factor binding in general.
Collapse
Affiliation(s)
- Siew Woh Choo
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Robert White
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Steven Russell
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
32
|
Gonsalves SE, Moses AM, Razak Z, Robert F, Westwood JT. Whole-genome analysis reveals that active heat shock factor binding sites are mostly associated with non-heat shock genes in Drosophila melanogaster. PLoS One 2011; 6:e15934. [PMID: 21264254 PMCID: PMC3021535 DOI: 10.1371/journal.pone.0015934] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 12/01/2010] [Indexed: 11/19/2022] Open
Abstract
During heat shock (HS) and other stresses, HS gene transcription in eukaryotes is up-regulated by the transcription factor heat shock factor (HSF). While the identities of the major HS genes have been known for more than 30 years, it has been suspected that HSF binds to numerous other genes and potentially regulates their transcription. In this study, we have used a chromatin immunoprecipitation and microarray (ChIP-chip) approach to identify 434 regions in the Drosophila genome that are bound by HSF. We have also performed a transcript analysis of heat shocked Kc167 cells and third instar larvae and compared them to HSF binding sites. The heat-induced transcription profiles were quite different between cells and larvae and surprisingly only about 10% of the genes associated with HSF binding sites show changed transcription. There were also genes that showed changes in transcript levels that did not appear to correlate with HSF binding sites. Analysis of the locations of the HSF binding sites revealed that 57% were contained within genes with approximately 2/3rds of these sites being in introns. We also found that the insulator protein, BEAF, has enriched binding prior to HS to promoters of genes that are bound by HSF upon HS but that are not transcriptionally induced during HS. When the genes associated with HSF binding sites in promoters were analyzed for gene ontology terms, categories such as stress response and transferase activity were enriched whereas analysis of genes having HSF binding sites in introns identified those categories plus ones related to developmental processes and reproduction. These results suggest that Drosophila HSF may be regulating many genes besides the known HS genes and that some of these genes may be regulated during non-stress conditions.
Collapse
Affiliation(s)
- Sarah E. Gonsalves
- Department of Cell and Systems Biology, University of Toronto, Mississauga, Canada
| | - Alan M. Moses
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Canada
| | - Zak Razak
- Department of Cell and Systems Biology, University of Toronto, Mississauga, Canada
| | - Francois Robert
- Institut de Recherches Cliniques de Montréal, Montréal, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - J. Timothy Westwood
- Department of Cell and Systems Biology, University of Toronto, Mississauga, Canada
- * E-mail:
| |
Collapse
|
33
|
Fox RM, Hanlon CD, Andrew DJ. The CrebA/Creb3-like transcription factors are major and direct regulators of secretory capacity. ACTA ACUST UNITED AC 2010; 191:479-92. [PMID: 21041443 PMCID: PMC3003312 DOI: 10.1083/jcb.201004062] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CrebA up-regulates expression of both the general protein machinery required in all cells for secretion and genes encoding cell type–specific secreted components. Secretion occurs in all cells, with relatively low levels in most cells and extremely high levels in specialized secretory cells, such as those of the pancreas, salivary, and mammary glands. How secretory capacity is selectively up-regulated in specialized secretory cells is unknown. Here, we find that the CrebA/Creb3-like family of bZip transcription factors functions to up-regulate expression of both the general protein machinery required in all cells for secretion and of cell type–specific secreted proteins. Drosophila CrebA directly binds the enhancers of secretory pathway genes and is both necessary and sufficient to activate expression of every secretory pathway component gene examined thus far. Microarray profiling reveals that CrebA also up-regulates expression of genes encoding cell type–specific secreted components. Finally, we found that the human CrebA orthologues, Creb3L1 and Creb3L2, have the ability to up-regulate the secretory pathway in nonsecretory cell types.
Collapse
Affiliation(s)
- Rebecca M Fox
- Department of Cell Biology, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
34
|
Abstract
The heat shock response was originally characterized as the induction of a set of major heat shock proteins encoded by heat shock genes. Because heat shock proteins act as molecular chaperones that facilitate protein folding and suppress protein aggregation, this response plays a major role in maintaining protein homeostasis. The heat shock response is regulated mainly at the level of transcription by heat shock factors (HSFs) in eukaryotes. HSF1 is a master regulator of the heat shock genes in mammalian cells, as is HSF3 in avian cells. HSFs play a significant role in suppressing protein misfolding in cells and in ameliorating the progression of Caenorhabditis elegans, Drosophila and mouse models of protein-misfolding disorders, by inducing the expression of heat shock genes. Recently, numerous HSF target genes were identified, such as the classical heat shock genes and other heat-inducible genes, called nonclassical heat shock genes in this study. Importance of the expression of the nonclassical heat shock genes was evidenced by the fact that mouse HSF3 and chicken HSF1 play a substantial role in the protection of cells from heat shock without inducing classical heat shock genes. Furthermore, HSF2 and HSF4, as well as HSF1, shown to have roles in development, were also revealed to be necessary for the expression of certain nonclassical heat shock genes. Thus, the heat shock response regulated by the HSF family should consist of the induction of classical as well as of nonclassical heat shock genes, both of which might be required to maintain protein homeostasis.
Collapse
|
35
|
Hayashida N, Fujimoto M, Tan K, Prakasam R, Shinkawa T, Li L, Ichikawa H, Takii R, Nakai A. Heat shock factor 1 ameliorates proteotoxicity in cooperation with the transcription factor NFAT. EMBO J 2010; 29:3459-69. [PMID: 20834230 DOI: 10.1038/emboj.2010.225] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 08/17/2010] [Indexed: 01/06/2023] Open
Abstract
Heat shock transcription factor 1 (HSF1) is an important regulator of protein homeostasis (proteostasis) by controlling the expression of major heat shock proteins (Hsps) that facilitate protein folding. However, it is unclear whether other proteostasis pathways are mediated by HSF1. Here, we identified novel targets of HSF1 in mammalian cells, which suppress the aggregation of polyglutamine (polyQ) protein. Among them, we show that one of the nuclear factor of activated T cells (NFAT) proteins, NFATc2, significantly inhibits polyQ aggregation in cells and is required for HSF1-mediated suppression of polyQ aggregation. NFAT deficiency accelerated disease progression including aggregation of a mutant polyQ-huntingtin protein and shortening of lifespan in R6/2 Huntington's disease mice. Furthermore, we found that HSF1 and NFAT cooperatively induce the expression of the scaffold protein PDZK3 and αB-crystallin, which facilitate the degradation of polyQ protein. These results show the first mechanistic basis for the observation that HSF1 has a much more profound effect on proteostasis than individual Hsp or combination of different Hsps, and suggest a new pathway for ameliorating protein-misfolding diseases.
Collapse
Affiliation(s)
- Naoki Hayashida
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Akerfelt M, Vihervaara A, Laiho A, Conter A, Christians ES, Sistonen L, Henriksson E. Heat shock transcription factor 1 localizes to sex chromatin during meiotic repression. J Biol Chem 2010; 285:34469-76. [PMID: 20802198 PMCID: PMC2966061 DOI: 10.1074/jbc.m110.157552] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heat shock factor 1 (HSF1) is an important transcription factor in cellular stress responses, cancer, aging, and developmental processes including gametogenesis. Disruption of Hsf1, together with another HSF family member, Hsf2, causes male sterility and complete lack of mature sperm in mice, but the specific role of HSF1 in spermatogenesis has remained unclear. Here, we show that HSF1 is transiently expressed in meiotic spermatocytes and haploid round spermatids in mouse testis. The Hsf1(-/-) male mice displayed regions of seminiferous tubules containing only spermatogonia and increased morphological abnormalities in sperm heads. In search for HSF1 target genes, we identified 742 putative promoters in mouse testis. Among them, the sex chromosomal multicopy genes that are expressed in postmeiotic cells were occupied by HSF1. Given that the sex chromatin mostly is repressed during and after meiosis, it is remarkable that HSF1 directly regulates the transcription of sex-linked multicopy genes during postmeiotic repression. In addition, our results show that HSF1 localizes to the sex body prior to the meiotic divisions and to the sex chromocenter after completed meiosis. To the best of our knowledge, HSF1 is the first known transcription factor found at the repressed sex chromatin during meiosis.
Collapse
Affiliation(s)
- Malin Akerfelt
- Department of Biosciences, Åbo Akademi University, FI-20521 Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
37
|
Ball RW, Warren-Paquin M, Tsurudome K, Liao EH, Elazzouzi F, Cavanagh C, An BS, Wang TT, White JH, Haghighi AP. Retrograde BMP signaling controls synaptic growth at the NMJ by regulating trio expression in motor neurons. Neuron 2010; 66:536-49. [PMID: 20510858 DOI: 10.1016/j.neuron.2010.04.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2010] [Indexed: 10/19/2022]
Abstract
Retrograde signaling is essential for coordinating the growth of synaptic structures; however, it is not clear how it can lead to modulation of cytoskeletal dynamics and structural changes at presynaptic terminals. We show that loss of retrograde bone morphogenic protein (BMP) signaling at the Drosophila larval neuromuscular junction (NMJ) leads to a significant reduction in levels of Rac GEF Trio and a diminution of transcription at the trio locus. We further find that Trio is required in motor neurons for normal structural growth. Finally, we show that transgenic expression of Trio in motor neurons can partially restore NMJ defects in larvae mutant for BMP signaling. Based on our findings, we propose a model in which a retrograde BMP signal from the muscle modulates GTPase activity through transcriptional regulation of Rac GEF trio, thereby regulating the homeostasis of synaptic growth at the NMJ.
Collapse
Affiliation(s)
- Robin W Ball
- Department of Physiology, McGill University, Montréal, QC H3G 1Y6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang S, Zhao X, Suran R, Vogt VM, Lis JT, Shi H. Knocking down gene function with an RNA aptamer expressed as part of an intron. Nucleic Acids Res 2010; 38:e154. [PMID: 20542918 PMCID: PMC2926621 DOI: 10.1093/nar/gkq529] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We developed a powerful expression system to produce aptamers and other types of functional RNA in yeast to examine their effects. Utilizing the intron homing process, the aptamer-coding sequences were integrated into hundreds of rRNA genes, and the aptamers were transcribed at high levels by RNA polymerase I without any additional promoter being introduced into the cell. We used this system to express an aptamer against the heat shock factor 1 (HSF1), a conserved transcription factor responsible for mobilizing specific genomic expression programs in response to stressful conditions such as elevated temperature. We observed a temperature sensitive growth retardation phenotype and specific decrease of heat shock gene expression. As HSF1 enables and promotes malignant growth and metastasis in mammals, and this aptamer binds yeast HSF1 and its mammalian ortholog with equal affinity, the results presented here attest to the potential of this aptamer as a specific and effective inhibitor of HSF1 activity.
Collapse
Affiliation(s)
- Shengchun Wang
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Xiaoching Zhao
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Robert Suran
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Volker M. Vogt
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - John T. Lis
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Hua Shi
- Department of Biological Sciences and Institute for RNA Science and Technology, University at Albany, State University of New York, Albany, NY 12222 and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
- *To whom correspondence should be addressed. Tel: +1 518 591 8840; Fax: +1 518 442 4767;
| |
Collapse
|
39
|
Tian S, Haney RA, Feder ME. Phylogeny disambiguates the evolution of heat-shock cis-regulatory elements in Drosophila. PLoS One 2010; 5:e10669. [PMID: 20498853 PMCID: PMC2871787 DOI: 10.1371/journal.pone.0010669] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 04/23/2010] [Indexed: 11/19/2022] Open
Abstract
Heat-shock genes have a well-studied control mechanism for their expression that is mediated through cis-regulatory motifs known as heat-shock elements (HSEs). The evolution of important features of this control mechanism has not been investigated in detail, however. Here we exploit the genome sequencing of multiple Drosophila species, combined with a wealth of available information on the structure and function of HSEs in D. melanogaster, to undertake this investigation. We find that in single-copy heat shock genes, entire HSEs have evolved or disappeared 14 times, and the phylogenetic approach bounds the timing and direction of these evolutionary events in relation to speciation. In contrast, in the multi-copy gene Hsp70, the number of HSEs is nearly constant across species. HSEs evolve in size, position, and sequence within heat-shock promoters. In turn, functional significance of certain features is implicated by preservation despite this evolutionary change; these features include tail-to-tail arrangements of HSEs, gapped HSEs, and the presence or absence of entire HSEs. The variation among Drosophila species indicates that the cis-regulatory encoding of responsiveness to heat and other stresses is diverse. The broad dimensions of variation uncovered are particularly important as they suggest a substantial challenge for functional studies.
Collapse
Affiliation(s)
- Sibo Tian
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, Illinois, United States of America
| | - Robert A. Haney
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, Illinois, United States of America
| | - Martin E. Feder
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
40
|
Aleksic J, Russell S. ChIPing away at the genome: the new frontier travel guide. MOLECULAR BIOSYSTEMS 2010; 5:1421-8. [PMID: 19617957 DOI: 10.1039/b906179g] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chromatin immunoprecipitation (ChIP) is a powerful technique for obtaining in vivo data on protein-DNA binding, providing an invaluable tool for elucidating gene regulation at a molecular level. Combined with high-throughput methods such as microarrays (ChIP-array) and second generation sequencing (ChIP-seq), the technique is now commonly used for answering questions about protein binding on a genome-wide level. This review focuses on the use of microarrays and sequencing for ChIP studies, provides a critical comparison of the currently used platforms and an overview of the computational methods available, and offers recommendations for optimal use of the techniques in a research context.
Collapse
Affiliation(s)
- Jelena Aleksic
- Department of Genetics and Cambridge Systems Biology Centre, University of Cambridge, Downing Street, Cambridge, UK
| | | |
Collapse
|
41
|
Johnson ML, Nagengast AA, Salz HK. PPS, a large multidomain protein, functions with sex-lethal to regulate alternative splicing in Drosophila. PLoS Genet 2010; 6:e1000872. [PMID: 20221253 PMCID: PMC2832672 DOI: 10.1371/journal.pgen.1000872] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 02/03/2010] [Indexed: 12/25/2022] Open
Abstract
Alternative splicing controls the expression of many genes, including the Drosophila sex determination gene Sex-lethal (Sxl). Sxl expression is controlled via a negative regulatory mechanism where inclusion of the translation-terminating male exon is blocked in females. Previous studies have shown that the mechanism leading to exon skipping is autoregulatory and requires the SXL protein to antagonize exon inclusion by interacting with core spliceosomal proteins, including the U1 snRNP protein Sans-fille (SNF). In studies begun by screening for proteins that interact with SNF, we identified PPS, a previously uncharacterized protein, as a novel component of the machinery required for Sxl male exon skipping. PPS encodes a large protein with four signature motifs, PHD, BRK, TFS2M, and SPOC, typically found in proteins involved in transcription. We demonstrate that PPS has a direct role in Sxl male exon skipping by showing first that loss of function mutations have phenotypes indicative of Sxl misregulation and second that the PPS protein forms a complex with SXL and the unspliced Sxl RNA. In addition, we mapped the recruitment of PPS, SXL, and SNF along the Sxl gene using chromatin immunoprecipitation (ChIP), which revealed that, like many other splicing factors, these proteins bind their RNA targets while in close proximity to the DNA. Interestingly, while SNF and SXL are specifically recruited to their predicted binding sites, PPS has a distinct pattern of accumulation along the Sxl gene, associating with a region that includes, but is not limited to, the SxlPm promoter. Together, these data indicate that PPS is different from other splicing factors involved in male-exon skipping and suggest, for the first time, a functional link between transcription and SXL-mediated alternative splicing. Loss of zygotic PPS function, however, is lethal to both sexes, indicating that its role may be of broad significance.
Collapse
Affiliation(s)
- Matthew L. Johnson
- Department of Genetics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
| | - Alexis A. Nagengast
- Department of Genetics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
| | - Helen K. Salz
- Department of Genetics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
42
|
Abstract
Analysis of the relationship between gene copy number and gene expression in aneuploid male Drosophila cells reveals a global compensation mechanism in addition to X chromosome-specific dosage compensation. Extensive departures from balanced gene dose in aneuploids are highly deleterious. However, we know very little about the relationship between gene copy number and expression in aneuploid cells. We determined copy number and transcript abundance (expression) genome-wide in Drosophila S2 cells by DNA-Seq and RNA-Seq. We found that S2 cells are aneuploid for >43 Mb of the genome, primarily in the range of one to five copies, and show a male genotype (∼ two X chromosomes and four sets of autosomes, or 2X;4A). Both X chromosomes and autosomes showed expression dosage compensation. X chromosome expression was elevated in a fixed-fold manner regardless of actual gene dose. In engineering terms, the system “anticipates” the perturbation caused by X dose, rather than responding to an error caused by the perturbation. This feed-forward regulation resulted in precise dosage compensation only when X dose was half of the autosome dose. Insufficient compensation occurred at lower X chromosome dose and excessive expression occurred at higher doses. RNAi knockdown of the Male Specific Lethal complex abolished feed-forward regulation. Both autosome and X chromosome genes show Male Specific Lethal–independent compensation that fits a first order dose-response curve. Our data indicate that expression dosage compensation dampens the effect of altered DNA copy number genome-wide. For the X chromosome, compensation includes fixed and dose-dependent components. While it is widely recognized that mutations in protein coding genes can have harmful consequences, one can also have too much or too little of a good thing. Except for the sex chromosomes, genes come in sets of two in diploid organisms. Extra or missing copies of genes or chromosomes result in an imbalance that can lead to cancers, miscarriages, and disease susceptibility. We have examined what happens to gene expression in Drosophila cells with the types of gross copy number changes that are typical of cancers. We have compared the response of autosomes and sex chromosomes and show that there is some compensation for copy number change in both cases. One response is universal and acts to correct copy number changes by changing transcript abundance. The other is specific to the X chromosome and acts to increase expression regardless of gene dose. Our data highlight how important gene expression balance is for cell function.
Collapse
|
43
|
Zhang Y, Oliver B. An evolutionary consequence of dosage compensation on Drosophila melanogaster female X-chromatin structure? BMC Genomics 2010; 11:6. [PMID: 20051121 PMCID: PMC2820458 DOI: 10.1186/1471-2164-11-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 01/05/2010] [Indexed: 01/07/2023] Open
Abstract
Background X chromosomes are subject to dosage compensation in Drosophila males. Dosage compensation requires cis sequence features of the X chromosome that are present in both sexes by definition and trans acting factors that target chromatin modifying machinery to the X specifically in males. The evolution of this system could result in neutral X chromatin changes that will be apparent in females. Results We find that the general chromatin structure of female X chromosomes is distinct from autosomes. Additionally, specific histone marks associated with dosage compensation and active chromatin marks on the male X chromosome are also enriched on the X chromosomes of females, albeit to a lesser degree. Conclusions Our data indicate that X chromatin structure is fundamentally different from autosome structure in both sexes. We suggest that the differences between the X chromosomes and autosomes in females are a consequence of mechanisms that have evolved to ensure sufficient X chromosome expression in the soma of males.
Collapse
Affiliation(s)
- Yu Zhang
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-8028, USA.
| | | |
Collapse
|
44
|
Norry FM, Larsen PF, Liu Y, Loeschcke V. Combined expression patterns of QTL-linked candidate genes best predict thermotolerance in Drosophila melanogaster. JOURNAL OF INSECT PHYSIOLOGY 2009; 55:1050-1057. [PMID: 19651134 DOI: 10.1016/j.jinsphys.2009.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 07/22/2009] [Accepted: 07/23/2009] [Indexed: 05/28/2023]
Abstract
Knockdown resistance to high temperature (KRHT) is a thermal adaptation trait in Drosophila melanogaster. Here we used quantitative real-time PCR (qRT-PCR) to test for possible associations between KRHT and the expression of candidate genes within quantitative trait loci (QTL) in eight recombinant inbred lines (RIL). hsp60 and hsc70-3 map within an X-linked QTL, while CG10383, catsup, ddc, trap1, and cyp6a13 are linked in a KRHT-QTL on chromosome 2. hsc70-3 expression increased by heat-hardening. Principal Components analysis revealed that catsup, ddc and trap1 were either co-expressed or combined in their expression levels. This composite expression variable (e-PC1) was positively associated to KRHT in non-hardened RIL. In heat-hardened flies, hsp60 was negatively related to hsc70-3 on e-PC2, with effects on KRHT. These results are consistent with the notion that QTL can be shaped by expression variation in combined candidate loci. We found composite variables of gene expression (e-PCs) that best correlated to KRHT. Network effects with other untested linked loci are apparent because, in spite of their associations with KRHT phenotypes, e-PCs were sometimes uncorrelated with their QTL genotype.
Collapse
Affiliation(s)
- Fabian M Norry
- Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
45
|
Fujimoto M, Hayashida N, Katoh T, Oshima K, Shinkawa T, Prakasam R, Tan K, Inouye S, Takii R, Nakai A. A novel mouse HSF3 has the potential to activate nonclassical heat-shock genes during heat shock. Mol Biol Cell 2009; 21:106-16. [PMID: 19864465 PMCID: PMC2801703 DOI: 10.1091/mbc.e09-07-0639] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
HSF1 is a master regulator of the heat-shock response in mammalian cells, whereas in avian cells, HSF3, which was considered as an avian-specific factor, is required for the expression of classical heat-shock genes. Here, the authors identify mouse HSF3, and demonstrate that it has the potential to activate only nonclassical heat-shock genes. The heat-shock response is characterized by the expression of a set of classical heat-shock genes, and is regulated by heat-shock transcription factor 1 (HSF1) in mammals. However, comprehensive analyses of gene expression have revealed very large numbers of inducible genes in cells exposed to heat shock. It is believed that HSF1 is required for the heat-inducible expression of these genes although HSF2 and HSF4 modulate some of the gene expression. Here, we identified a novel mouse HSF3 (mHSF3) translocated into the nucleus during heat shock. However, mHSF3 did not activate classical heat-shock genes such as Hsp70. Remarkably, overexpression of mHSF3 restored the expression of nonclassical heat-shock genes such as PDZK3 and PROM2 in HSF1-null mouse embryonic fibroblasts (MEFs). Although down-regulation of mHSF3 expression had no effect on gene expression or cell survival in wild-type MEF cells, it abolished the moderate expression of PDZK3 mRNA and reduced cell survival in HSF1-null MEF cells during heat shock. We propose that mHSF3 represents a unique HSF that has the potential to activate only nonclassical heat-shock genes to protect cells from detrimental stresses.
Collapse
Affiliation(s)
- Mitsuaki Fujimoto
- Department of Biochemistry, Yamaguchi University School of Medicine, Ube, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Whitesell L, Lindquist S. Inhibiting the transcription factor HSF1 as an anticancer strategy. Expert Opin Ther Targets 2009; 13:469-78. [PMID: 19335068 DOI: 10.1517/14728220902832697] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND In mammals, the cytoprotective heat-shock response is regulated primarily by heat shock factor 1 (HSF1). Unfortunately, the effects of HSF1 also support the ability of cancer cells to accommodate imbalances in signaling and alterations in DNA, protein and energy metabolism associated with oncogenesis. The malignant lifestyle confers dependence on this 'non-oncogene', suggesting a therapeutic role for HSF1 inhibitors. OBJECTIVE/METHODS We begin with an overview of how HSF1 affects cancer biology and how its activity is regulated. We then summarize progress in discovery and development of HSF1 inhibitors, their current limitations and potential as anticancer agents with a fundamentally different scope of action from other clinically validated modulators of protein homeostasis. RESULTS/CONCLUSIONS It is likely that within the next 5 years usable inhibitors of HSF1 will be identified and in early pre-clinical evaluation.
Collapse
Affiliation(s)
- Luke Whitesell
- Whitehead Institute, 9 Cambridge Center, Cambridge, MA 02142, USA.
| | | |
Collapse
|
47
|
Chen Q, Yu Q, Song Y, Li P, Chang Y, Wang Z, Liu L, Wu W, Lin J. Cloning of human XAF1 gene promoter and assay of its transcription activity in a variety of cell lines. FRONTIERS OF MEDICINE IN CHINA 2009; 3:148-152. [DOI: 10.1007/s11684-009-0032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
Mahalingam D, Swords R, Carew JS, Nawrocki ST, Bhalla K, Giles FJ. Targeting HSP90 for cancer therapy. Br J Cancer 2009; 100:1523-9. [PMID: 19401686 PMCID: PMC2696754 DOI: 10.1038/sj.bjc.6605066] [Citation(s) in RCA: 246] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Heat-shock proteins (HSPs) are molecular chaperones that regulate protein folding to ensure correct conformation and translocation and to avoid protein aggregation. Heat-shock proteins are increased in many solid tumours and haematological malignancies. Many oncogenic proteins responsible for the transformation of cells to cancerous forms are client proteins of HSP90. Targeting HSP90 with chemical inhibitors would degrade these oncogenic proteins, and thus serve as useful anticancer agents. This review provides an overview of the HSP chaperone machinery and the structure and function of HSP90. We also highlight the key oncogenic proteins that are regulated by HSP90 and describe how inhibition of HSP90 could alter the activity of multiple signalling proteins, receptors and transcriptional factors implicated in carcinogenesis.
Collapse
Affiliation(s)
- D Mahalingam
- Institute for Drug Development, Cancer Research and Therapy Centre at the University of Texas Health Science Centre, San Antonio, TX 78229, USA
| | | | | | | | | | | |
Collapse
|
49
|
Kwong C, Adryan B, Bell I, Meadows L, Russell S, Manak JR, White R. Stability and dynamics of polycomb target sites in Drosophila development. PLoS Genet 2008; 4:e1000178. [PMID: 18773083 PMCID: PMC2525605 DOI: 10.1371/journal.pgen.1000178] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 07/18/2008] [Indexed: 11/25/2022] Open
Abstract
Polycomb-group (PcG) and Trithorax-group proteins together form a maintenance machinery that is responsible for stable heritable states of gene activity. While the best-studied target genes are the Hox genes of the Antennapedia and Bithorax complexes, a large number of key developmental genes are also Polycomb (Pc) targets, indicating a widespread role for this maintenance machinery in cell fate determination. We have studied the linkage between the binding of PcG proteins and the developmental regulation of gene expression using whole-genome mapping to identify sites bound by the PcG proteins, Pc and Pleiohomeotic (Pho), in the Drosophila embryo and in a more restricted tissue, the imaginal discs of the third thoracic segment. Our data provide support for the idea that Pho is a general component of the maintenance machinery, since the majority of Pc targets are also associated with Pho binding. We find, in general, considerable developmental stability of Pc and Pho binding at target genes and observe that Pc/Pho binding can be associated with both expressed and inactive genes. In particular, at the Hox complexes, both active and inactive genes have significant Pc and Pho binding. However, in comparison to inactive genes, the active Hox genes show reduced and altered binding profiles. During development, Pc target genes are not simply constantly associated with Pc/Pho binding, and we identify sets of genes with clear differential binding between embryo and imaginal disc. Using existing datasets, we show that for specific fate-determining genes of the haemocyte lineage, the active state is characterised by lack of Pc binding. Overall, our analysis suggests a dynamic relationship between Pc/Pho binding and gene transcription. Pc/Pho binding does not preclude transcription, but levels of Pc/Pho binding change during development, and loss of Pc/Pho binding can be associated with both stable gene activity and inactivity. Cells make fate decisions as they progressively differentiate into specific cell types during development. The stability of these decisions is important and is achieved, in part, by changes to the chromatin that packages DNA in the nucleus. A key set of protein complexes that together constitute the Polycomb-group/Trithorax-group (PcG/TrxG) machinery is involved in chromatin modification and is known to operate at a large number of genes involved in developmental decisions. The PcG proteins establish stable gene repression, whereas the TrxG counteract the PcG to enable gene activation. How this PcG/TrxG balance works is not understood. By mapping PcG protein binding to chromatin in vivo, we show, in general, a relatively constant association of PcG protein at target genes during development. However, we also find changes in binding at specific genes. While some of these changes are consistent with a loss of PcG proteins associated with gene expression, we also find examples where PcG proteins are present at active genes and not present at inactive genes. Our analysis supports the idea that simply the presence of PcG proteins at a target gene does not necessarily result in gene repression and suggests a more dynamic balance between PcG protein binding and gene expression.
Collapse
Affiliation(s)
- Camilla Kwong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Boris Adryan
- Theoretical and Computational Biology Group, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Ian Bell
- Affymetrix Inc., Affy Labs–Transcriptome, Santa Clara, California, United States of America
| | - Lisa Meadows
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Steven Russell
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - J. Robert Manak
- Affymetrix Inc., Affy Labs–Transcriptome, Santa Clara, California, United States of America
| | - Robert White
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Liu HT, Gao F, Li GL, Han JL, Liu DL, Sun DY, Zhou RG. The calmodulin-binding protein kinase 3 is part of heat-shock signal transduction in Arabidopsis thaliana. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2008; 55:760-73. [PMID: 18466301 DOI: 10.1111/j.1365-313x.2008.03544.x] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Based on our previous findings, we proposed a pathway for the participation of Ca(2+)/calmodulin (CaM) in heat-shock (HS) signal transduction. The specific mechanism by which CaM regulates activation of heat-shock transcription factors (HSFs) is not known. CaM-binding protein kinases (CBK) are the most poorly understood of the CaM target proteins in plants. In this study, using a yeast two-hybrid assay, we found that AtCBK3 interacts with AtHSFA1a. Fluorescence resonance energy transfer was used to confirm the interaction between AtCBK3-YFP and AtHSFA1a-CFP. Furthermore, we demonstrate that purified recombinant AtCBK3 phosphorylated recombinant AtHSFA1a in vitro. We also describe the results of both downregulation of AtCBK3 expression and ectopic overexpression in Arabidopsis thaliana. The T-DNA insertion AtCBK3 knockout lines had impaired basal thermotolerance, which could be complemented by transformation of plants with the native gene. Overexpression of AtCBK3 resulted in plants with increased basal thermotolerance. Results from real-time quantitative PCR and protein gel-blot analyses suggest that AtCBK3 regulates transcription of heat-shock protein (HSP) genes and synthesis of HSPs. The binding activity of HSF to the heat-shock element (HSE), the mRNA level of HSP genes and synthesis of HSPs were upregulated in AtCBK3-overexpressing lines after HS, but downregulated in AtCBK3 null lines. These results indicate that AtCBK3 controls the binding activity of HSFs to HSEs by phosphorylation of AtHSFA1a, and is an important component of the HS signal transduction pathway.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Institute of Genetics and Physiology, Hebei Academy of Agricultural Sciences, Shijiazhuang 050051, China
| | | | | | | | | | | | | |
Collapse
|