251
|
Horibata S, Rice EJ, Zheng H, Mukai C, Chu T, Marks BA, Coonrod SA, Danko CG. A bi-stable feedback loop between GDNF, EGR1, and ERα contribute to endocrine resistant breast cancer. PLoS One 2018; 13:e0194522. [PMID: 29614078 PMCID: PMC5882141 DOI: 10.1371/journal.pone.0194522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/05/2018] [Indexed: 01/06/2023] Open
Abstract
Discovering regulatory interactions between genes that specify the behavioral properties of cells remains an important challenge. We used the dynamics of transcriptional changes resolved by PRO-seq to identify a regulatory network responsible for endocrine resistance in breast cancer. We show that GDNF leads to endocrine resistance by switching the active state in a bi-stable feedback loop between GDNF, EGR1, and the master transcription factor ERα. GDNF stimulates MAP kinase, activating the transcription factors SRF and AP-1. SRF initiates an immediate transcriptional response, activating EGR1 and suppressing ERα. Newly translated EGR1 protein activates endogenous GDNF, leading to constitutive GDNF and EGR1 up-regulation, and the sustained down-regulation of ERα. Endocrine resistant MCF-7 cells are constitutively in the GDNF-high/ ERα-low state, suggesting that the state in the bi-stable feedback loop may provide a 'memory' of endocrine resistance. Thus, we identified a regulatory network switch that contributes to drug resistance in breast cancer.
Collapse
Affiliation(s)
- Sachi Horibata
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Edward J. Rice
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Hui Zheng
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Chinatsu Mukai
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Tinyi Chu
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Graduate Field of Computational Biology, Cornell University, Ithaca, NY, United States of America
| | - Brooke A. Marks
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Scott A. Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Charles G. Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
252
|
Horibata S, Rice EJ, Mukai C, Marks BA, Sams K, Zheng H, Anguish LJ, Coonrod SA, Danko CG. ER-positive breast cancer cells are poised for RET-mediated endocrine resistance. PLoS One 2018; 13:e0194023. [PMID: 29608602 PMCID: PMC5880349 DOI: 10.1371/journal.pone.0194023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/22/2018] [Indexed: 01/13/2023] Open
Abstract
The RET tyrosine kinase signaling pathway is involved in the development of endocrine resistant ER+ breast cancer. However, we know little about how ER+ cells activate RET signaling and initiate an endocrine resistant phenotype. Here we show that both ER+ endocrine resistant and sensitive breast cancers have a functional RET tyrosine kinase signaling pathway, but that endocrine sensitive breast cancer cells lack RET ligands that are necessary to drive endocrine resistance. Transcription of one RET ligand, GDNF, is necessary and sufficient to confer resistance in the ER+ MCF-7 cell line. Endogenous GDNF produced by endocrine resistant cells is translated, secreted into the media, and activates RET signaling in nearby cells. In patients, RET ligand expression predicts responsiveness to endocrine therapies and correlates with survival. Collectively, our findings show that ER+ tumor cells are "poised" for RET mediated endocrine resistance, expressing all components of the RET signaling pathway, but endocrine sensitive cells lack high expression of RET ligands that are necessary to initiate the resistance phenotype.
Collapse
Affiliation(s)
- Sachi Horibata
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Edward J. Rice
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Chinatsu Mukai
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Brooke A. Marks
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Kelly Sams
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Hui Zheng
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Lynne J. Anguish
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Scott A. Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Charles G. Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
253
|
Fujimoto H, Saito Y, Ohuchida K, Kawakami E, Fujiki S, Watanabe T, Ono R, Kaneko A, Takagi S, Najima Y, Hijikata A, Cui L, Ueki T, Oda Y, Hori S, Ohara O, Nakamura M, Saito T, Ishikawa F. Deregulated Mucosal Immune Surveillance through Gut-Associated Regulatory T Cells and PD-1 + T Cells in Human Colorectal Cancer. THE JOURNAL OF IMMUNOLOGY 2018; 200:3291-3303. [PMID: 29581358 DOI: 10.4049/jimmunol.1701222] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Disturbed balance between immune surveillance and tolerance may lead to poor clinical outcomes in some malignancies. In paired analyses of adenocarcinoma and normal mucosa from 142 patients, we found a significant increase of the CD4/CD8 ratio and accumulation of regulatory T cells (Tregs) within the adenocarcinoma. The increased frequency of Tregs correlated with the local infiltration and extension of the tumor. There was concurrent maturation arrest, upregulation of programmed death-1 expression, and functional impairment in CD8+ T cells (CTLs) isolated from the adenocarcinoma. Adenocarcinoma-associated Tregs directly inhibit the function of normal human CTLs in vitro. With histopathological analysis, Foxp3+ Tregs were preferentially located in stroma. Concurrent transcriptome analysis of epithelial cells, stromal cells, and T cell subsets obtained from carcinomatous and normal intestinal samples from patients revealed a distinct gene expression signature in colorectal adenocarcinoma-associated Tregs, with overexpression of CCR1, CCR8, and TNFRSF9, whereas their ligands CCL4 and TNFSF9 were found upregulated in cancerous epithelium. Overexpression of WNT2 and CADM1, associated with carcinogenesis and metastasis, in cancer-associated stromal cells suggests that both cancer cells and stromal cells play important roles in the development and progression of colorectal cancer through the formation of a tumor microenvironment. The identification of CTL anergy by Tregs and the unique gene expression signature of human Tregs and stromal cells in colorectal cancer patients may facilitate the development of new therapeutics against malignancies.
Collapse
Affiliation(s)
- Hanae Fujimoto
- Department of Immune Regulation Research, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba 260-0856, Japan.,Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Yoriko Saito
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Eiryo Kawakami
- RIKEN Medical Sciences Innovation Hub Program, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Saera Fujiki
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Rintaro Ono
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Akiko Kaneko
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Shinsuke Takagi
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Yuho Najima
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Atsushi Hijikata
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Lin Cui
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ueki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,Department of Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shohei Hori
- Laboratory for Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; and
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.,Department of Human Genome Research, Kazusa DNA Research Institute, Kisarazu 292-0818, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Fumihiko Ishikawa
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan;
| |
Collapse
|
254
|
Schofield JA, Duffy EE, Kiefer L, Sullivan MC, Simon MD. TimeLapse-seq: adding a temporal dimension to RNA sequencing through nucleoside recoding. Nat Methods 2018; 15:221-225. [PMID: 29355846 PMCID: PMC5831505 DOI: 10.1038/nmeth.4582] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022]
Abstract
RNA sequencing (RNA-seq) offers a snapshot of cellular RNA populations, but not temporal information about the sequenced RNA. Here we report TimeLapse-seq, which uses oxidative-nucleophilic-aromatic substitution to convert 4-thiouridine into cytidine analogs, yielding apparent U-to-C mutations that mark new transcripts upon sequencing. TimeLapse-seq is a single-molecule approach that is adaptable to many applications and reveals RNA dynamics and induced differential expression concealed in traditional RNA-seq.
Collapse
Affiliation(s)
- Jeremy A Schofield
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, USA
- Chemical Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Erin E Duffy
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, USA
- Chemical Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Lea Kiefer
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, USA
- Chemical Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Meaghan C Sullivan
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, USA
- Chemical Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Matthew D Simon
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, USA
- Chemical Biology Institute, Yale University, West Haven, Connecticut, USA
| |
Collapse
|
255
|
Booth GT, Parua PK, Sansó M, Fisher RP, Lis JT. Cdk9 regulates a promoter-proximal checkpoint to modulate RNA polymerase II elongation rate in fission yeast. Nat Commun 2018; 9:543. [PMID: 29416031 PMCID: PMC5803247 DOI: 10.1038/s41467-018-03006-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/12/2018] [Indexed: 12/25/2022] Open
Abstract
Post-translational modifications of the transcription elongation complex provide mechanisms to fine-tune gene expression, yet their specific impacts on RNA polymerase II regulation remain difficult to ascertain. Here, in Schizosaccharomyces pombe, we examine the role of Cdk9, and related Mcs6/Cdk7 and Lsk1/Cdk12 kinases, on transcription at base-pair resolution with Precision Run-On sequencing (PRO-seq). Within a minute of Cdk9 inhibition, phosphorylation of Pol II-associated factor, Spt5 is undetectable. The effects of Cdk9 inhibition are more severe than inhibition of Cdk7 and Cdk12, resulting in a shift of Pol II toward the transcription start site (TSS). A time course of Cdk9 inhibition reveals that early transcribing Pol II can escape promoter-proximal regions, but with a severely reduced elongation rate of only ~400 bp/min. Our results in fission yeast suggest the existence of a conserved global regulatory checkpoint that requires Cdk9 kinase activity.
Collapse
Affiliation(s)
- Gregory T Booth
- Department of Molecular Biology and Genetics, Cornell University, 107 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853-2703, USA
| | - Pabitra K Parua
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Miriam Sansó
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert P Fisher
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, 107 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853-2703, USA.
| |
Collapse
|
256
|
Krakowiak J, Zheng X, Patel N, Feder ZA, Anandhakumar J, Valerius K, Gross DS, Khalil AS, Pincus D. Hsf1 and Hsp70 constitute a two-component feedback loop that regulates the yeast heat shock response. eLife 2018; 7:31668. [PMID: 29393852 PMCID: PMC5809143 DOI: 10.7554/elife.31668] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/01/2018] [Indexed: 01/29/2023] Open
Abstract
Models for regulation of the eukaryotic heat shock response typically invoke a negative feedback loop consisting of the transcriptional activator Hsf1 and a molecular chaperone. Previously we identified Hsp70 as the chaperone responsible for Hsf1 repression and constructed a mathematical model that recapitulated the yeast heat shock response (Zheng et al., 2016). The model was based on two assumptions: dissociation of Hsp70 activates Hsf1, and transcriptional induction of Hsp70 deactivates Hsf1. Here we validate these assumptions. First, we severed the feedback loop by uncoupling Hsp70 expression from Hsf1 regulation. As predicted by the model, Hsf1 was unable to efficiently deactivate in the absence of Hsp70 transcriptional induction. Next, we mapped a discrete Hsp70 binding site on Hsf1 to a C-terminal segment known as conserved element 2 (CE2). In vitro, CE2 binds to Hsp70 with low affinity (9 µM), in agreement with model requirements. In cells, removal of CE2 resulted in increased basal Hsf1 activity and delayed deactivation during heat shock, while tandem repeats of CE2 sped up Hsf1 deactivation. Finally, we uncovered a role for the N-terminal domain of Hsf1 in negatively regulating DNA binding. These results reveal the quantitative control mechanisms underlying the heat shock response.
Collapse
Affiliation(s)
- Joanna Krakowiak
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Xu Zheng
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Nikit Patel
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States
| | - Zoë A Feder
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Jayamani Anandhakumar
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States
| | - Kendra Valerius
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - David S Gross
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, United States
| | - Ahmad S Khalil
- Whitehead Institute for Biomedical Research, Cambridge, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, United States
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, United States
| |
Collapse
|
257
|
Nguyen PH, Greene E, Kong BW, Bottje W, Anthony N, Dridi S. Acute Heat Stress Alters the Expression of Orexin System in Quail Muscle. Front Physiol 2017; 8:1079. [PMID: 29311994 PMCID: PMC5742252 DOI: 10.3389/fphys.2017.01079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/07/2017] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidences indicate that the hypothalamic neuropeptide orexins or hypocretins are involved in stress-induced responses in mammals. Recently, we found that orexin is expressed and secreted from avian muscle cells, however its regulation is still unknown. In this study, we investigated the effect of heat and oxidative stress, the most challenging stressors in poultry production, on the expression of orexin system in quail muscle tissues and myoblast cell lines. Four week-old genetically selected susceptible and resistant Japanese quail (Coturnix coturnix Japonica) lines were exposed to acute heat stress (HS, 37°C for 1.5 h) or maintained at thermoneutral conditions (24°C). Quail myoblast (QM7) cell line was exposed to heat stress (45°C) for 0.5, 1, 2, or 4 h. The control cells were maintained at 37°C. The cells were also treated with several doses of hydrogen peroxide (H2O2, 10-200 μM) or 4-Hydroxynonenal (4-HNE, 10-30 μM) as oxidative stress. Untreated cells were used as controls. Acute HS significantly induced the expression of HSP70 and down-regulated orexin system in both quail muscle tissue and QM7 cells. Similarly, H2O2 but not 4-HNE treatment significantly increased HSP70 protein levels and dysregulated the expression of orexin and its related receptors in a dose-dependent manner in QM7 cells. Transient overexpression of HSP70 down-regulated the expression of orexin system in QM7 cells. Taken together, these data indicate that orexin may be a key player in stress response in avian muscle by demonstrating that heat and oxidative stress alter the expression of orexin system in quail muscle. This effect might be mediated through HSP70. Unraveling the upstream regulators and downstream effectors of orexin in avian muscle merits further in depth investigations.
Collapse
Affiliation(s)
- Phuong H Nguyen
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Elisabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Byung-Whi Kong
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Walter Bottje
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Nicholas Anthony
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
258
|
Pomatto LCD, Davies KJA. The role of declining adaptive homeostasis in ageing. J Physiol 2017; 595:7275-7309. [PMID: 29028112 PMCID: PMC5730851 DOI: 10.1113/jp275072] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Adaptive homeostasis is "the transient expansion or contraction of the homeostatic range for any given physiological parameter in response to exposure to sub-toxic, non-damaging, signalling molecules or events, or the removal or cessation of such molecules or events" (Davies, 2016). Adaptive homeostasis enables biological systems to make continuous short-term adjustments for optimal functioning despite ever-changing internal and external environments. Initiation of adaptation in response to an appropriate signal allows organisms to successfully cope with much greater, normally toxic, stresses. These short-term responses are initiated following effective signals, including hypoxia, cold shock, heat shock, oxidative stress, exercise-induced adaptation, caloric restriction, osmotic stress, mechanical stress, immune response, and even emotional stress. There is now substantial literature detailing a decline in adaptive homeostasis that, unfortunately, appears to manifest with ageing, especially in the last third of the lifespan. In this review, we present the hypothesis that one hallmark of the ageing process is a significant decline in adaptive homeostasis capacity. We discuss the mechanistic importance of diminished capacity for short-term (reversible) adaptive responses (both biochemical and signal transduction/gene expression-based) to changing internal and external conditions, for short-term survival and for lifespan and healthspan. Studies of cultured mammalian cells, worms, flies, rodents, simians, apes, and even humans, all indicate declining adaptive homeostasis as a potential contributor to age-dependent senescence, increased risk of disease, and even mortality. Emerging work points to Nrf2-Keap1 signal transduction pathway inhibitors, including Bach1 and c-Myc, both of whose tissue concentrations increase with age, as possible major causes for age-dependent loss of adaptive homeostasis.
Collapse
Affiliation(s)
- Laura C. D. Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
| | - Kelvin J. A. Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
- Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of LettersArts & Sciences: the University of Southern CaliforniaLos AngelesCA 90089‐0191USA
| |
Collapse
|
259
|
Dhanani KCH, Samson WJ, Edkins AL. Fibronectin is a stress responsive gene regulated by HSF1 in response to geldanamycin. Sci Rep 2017; 7:17617. [PMID: 29247221 PMCID: PMC5732156 DOI: 10.1038/s41598-017-18061-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/02/2017] [Indexed: 01/16/2023] Open
Abstract
Fibronectin is an extracellular matrix glycoprotein with key roles in cell adhesion and migration. Hsp90 binds directly to fibronectin and Hsp90 depletion regulates fibronectin matrix stability. Where inhibition of Hsp90 with a C-terminal inhibitor, novobiocin, reduced the fibronectin matrix, treatment with an N-terminal inhibitor, geldanamycin, increased fibronectin levels. Geldanamycin treatment induced a stress response and a strong dose and time dependent increase in fibronectin mRNA via activation of the fibronectin promoter. Three putative heat shock elements (HSEs) were identified in the fibronectin promoter. Loss of two of these HSEs reduced both basal and geldanamycin-induced promoter activity, as did inhibition of the stress-responsive transcription factor HSF1. Binding of HSF1 to one of the putative HSE was confirmed by ChIP under basal conditions, and occupancy shown to increase with geldanamycin treatment. These data support the hypothesis that fibronectin is stress-responsive and a functional HSF1 target gene. COLA42 and LAMB3 mRNA levels were also increased with geldanamycin indicating that regulation of extracellular matrix (ECM) genes by HSF1 may be a wider phenomenon. Taken together, these data have implications for our understanding of ECM dynamics in stress-related diseases in which HSF1 is activated, and where the clinical application of N-terminal Hsp90 inhibitors is intended.
Collapse
Affiliation(s)
- Karim Colin Hassan Dhanani
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, 6140, South Africa
| | - William John Samson
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, 6140, South Africa
| | - Adrienne Lesley Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, 6140, South Africa.
| |
Collapse
|
260
|
Global unleashing of transcription elongation waves in response to genotoxic stress restricts somatic mutation rate. Nat Commun 2017; 8:2076. [PMID: 29233992 PMCID: PMC5727188 DOI: 10.1038/s41467-017-02145-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/08/2017] [Indexed: 01/11/2023] Open
Abstract
Complex molecular responses preserve gene expression accuracy and genome integrity in the face of environmental perturbations. Here we report that, in response to UV irradiation, RNA polymerase II (RNAPII) molecules are dynamically and synchronously released from promoter-proximal regions into elongation to promote uniform and accelerated surveillance of the whole transcribed genome. The maximised influx of de novo released RNAPII correlates with increased damage-sensing, as confirmed by RNAPII progressive accumulation at dipyrimidine sites and by the average slow-down of elongation rates in gene bodies. In turn, this transcription elongation ‘safe’ mode guarantees efficient DNA repair regardless of damage location, gene size and transcription level. Accordingly, we detect low and homogenous rates of mutational signatures associated with UV exposure or cigarette smoke across all active genes. Our study reveals a novel advantage for transcription regulation at the promoter-proximal level and provides unanticipated insights into how active transcription shapes the mutagenic landscape of cancer genomes. Precise orchestration of gene expression regulation upon DNA damage is essential for genome integrity. Here the authors identify a novel widespread stress-triggered defence mechanism that promotes rapid transcription-driven genomic surveillance thus limiting mutagenesis and shaping cancer genomes.
Collapse
|
261
|
Molecular Genetic Analysis of Human Endometrial Mesenchymal Stem Cells That Survived Sublethal Heat Shock. Stem Cells Int 2017; 2017:2362630. [PMID: 29375621 PMCID: PMC5742502 DOI: 10.1155/2017/2362630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
High temperature is a critical environmental and personal factor. Although heat shock is a well-studied biological phenomenon, hyperthermia response of stem cells is poorly understood. Previously, we demonstrated that sublethal heat shock induced premature senescence in human endometrial mesenchymal stem cells (eMSC). This study aimed to investigate the fate of eMSC-survived sublethal heat shock (SHS) with special emphasis on their genetic stability and possible malignant transformation using methods of classic and molecular karyotyping, next-generation sequencing, and transcriptome functional analysis. G-banding revealed random chromosome breakages and aneuploidy in the SHS-treated eMSC. Molecular karyotyping found no genomic imbalance in these cells. Gene module and protein interaction network analysis of mRNA sequencing data showed that compared to untreated cells, SHS-survived progeny revealed some difference in gene expression. However, no hallmarks of cancer were found. Our data identified downregulation of oncogenic signaling, upregulation of tumor-suppressing and prosenescence signaling, induction of mismatch, and excision DNA repair. The common feature of heated eMSC is the silence of MYC, AKT1/PKB oncogenes, and hTERT telomerase. Overall, our data indicate that despite genetic instability, SHS-survived eMSC do not undergo transformation. After long-term cultivation, these cells like their unheated counterparts enter replicative senescence and die.
Collapse
|
262
|
The HSF1-PARP13-PARP1 complex facilitates DNA repair and promotes mammary tumorigenesis. Nat Commun 2017; 8:1638. [PMID: 29158484 PMCID: PMC5696371 DOI: 10.1038/s41467-017-01807-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/13/2017] [Indexed: 01/23/2023] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is involved in DNA repair, chromatin structure, and transcription. However, the mechanisms that regulate PARP1 distribution on DNA are poorly understood. Here, we show that heat shock transcription factor 1 (HSF1) recruits PARP1 through the scaffold protein PARP13. In response to DNA damage, activated and auto-poly-ADP-ribosylated PARP1 dissociates from HSF1–PARP13, and redistributes to DNA lesions and DNA damage-inducible gene loci. Histone deacetylase 1 maintains PARP1 in the ternary complex by inactivating PARP1 through deacetylation. Blocking ternary complex formation impairs redistribution of PARP1 during DNA damage, which reduces gene expression and DNA repair. Furthermore, ternary complex formation and PARP1 redistribution protect cells from DNA damage by promoting DNA repair, and support growth of BRCA1-null mammary tumors, which are sensitive to PARP inhibitors. Our findings identify HSF1 as a regulator of genome integrity and define this function as a guarding mechanism for a specific type of mammary tumorigenesis. PARP1 recruitment to DNA lesions is critical for DNA damage repair but how DNA damage induces PARP1 redistribution is largely unknown. Here, the authors provide evidence that PARP1 redistribution and DNA repair in tumor cells depend on the formation of a HSF1–PARP13–PARP1 complex.
Collapse
|
263
|
Dukler N, Booth GT, Huang YF, Tippens N, Waters CT, Danko CG, Lis JT, Siepel A. Nascent RNA sequencing reveals a dynamic global transcriptional response at genes and enhancers to the natural medicinal compound celastrol. Genome Res 2017; 27:1816-1829. [PMID: 29025894 PMCID: PMC5668940 DOI: 10.1101/gr.222935.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/13/2017] [Indexed: 12/16/2022]
Abstract
Most studies of responses to transcriptional stimuli measure changes in cellular mRNA concentrations. By sequencing nascent RNA instead, it is possible to detect changes in transcription in minutes rather than hours and thereby distinguish primary from secondary responses to regulatory signals. Here, we describe the use of PRO-seq to characterize the immediate transcriptional response in human cells to celastrol, a compound derived from traditional Chinese medicine that has potent anti-inflammatory, tumor-inhibitory, and obesity-controlling effects. Celastrol is known to elicit a cellular stress response resembling the response to heat shock, but the transcriptional basis of this response remains unclear. Our analysis of PRO-seq data for K562 cells reveals dramatic transcriptional effects soon after celastrol treatment at a broad collection of both coding and noncoding transcription units. This transcriptional response occurred in two major waves, one within 10 min, and a second 40-60 min after treatment. Transcriptional activity was generally repressed by celastrol, but one distinct group of genes, enriched for roles in the heat shock response, displayed strong activation. Using a regression approach, we identified key transcription factors that appear to drive these transcriptional responses, including members of the E2F and RFX families. We also found sequence-based evidence that particular transcription factors drive the activation of enhancers. We observed increased polymerase pausing at both genes and enhancers, suggesting that pause release may be widely inhibited during the celastrol response. Our study demonstrates that a careful analysis of PRO-seq time-course data can disentangle key aspects of a complex transcriptional response, and it provides new insights into the activity of a powerful pharmacological agent.
Collapse
Affiliation(s)
- Noah Dukler
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York 10065, USA
| | - Gregory T Booth
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850, USA
| | - Yi-Fei Huang
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Nathaniel Tippens
- Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York 10065, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850, USA
| | - Colin T Waters
- Program in Biological and Biomedical Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Charles G Danko
- Baker Institute for Animal Health, Cornell University, Ithaca, New York 14850, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850, USA
| | - Adam Siepel
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
264
|
Abstract
HSF1 is the supposed master regulator of the heat shock response. In this issue of Molecular Cell, Solís et al. reveal that it has a much narrower job description: organizing a small team of molecular chaperones that keep the proteome moving.
Collapse
Affiliation(s)
| | - D Allan Drummond
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
265
|
Cortijo S, Charoensawan V, Brestovitsky A, Buning R, Ravarani C, Rhodes D, van Noort J, Jaeger KE, Wigge PA. Transcriptional Regulation of the Ambient Temperature Response by H2A.Z Nucleosomes and HSF1 Transcription Factors in Arabidopsis. MOLECULAR PLANT 2017; 10:1258-1273. [PMID: 28893714 PMCID: PMC6175055 DOI: 10.1016/j.molp.2017.08.014] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 05/18/2023]
Abstract
Temperature influences the distribution, range, and phenology of plants. The key transcriptional activators of heat shock response in eukaryotes, the heat shock factors (HSFs), have undergone large-scale gene amplification in plants. While HSFs are central in heat stress responses, their role in the response to ambient temperature changes is less well understood. We show here that the warm ambient temperature transcriptome is dependent upon the HSFA1 clade of Arabidopsis HSFs, which cause a rapid and dynamic eviction of H2A.Z nucleosomes at target genes. A transcriptional cascade results in the activation of multiple downstream stress-responsive transcription factors, triggering large-scale changes to the transcriptome in response to elevated temperature. H2A.Z nucleosomes are enriched at temperature-responsive genes at non-inducible temperature, and thus likely confer inducibility of gene expression and higher responsive dynamics. We propose that the antagonistic effects of H2A.Z and HSF1 provide a mechanism to activate gene expression rapidly and precisely in response to temperature, while preventing leaky transcription in the absence of an activation signal.
Collapse
Affiliation(s)
- Sandra Cortijo
- The Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK
| | - Varodom Charoensawan
- The Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK; Department of Biochemistry, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi District, Bangkok 10400, Thailand; Integrative Computational BioScience (ICBS) Center, Mahidol University, 999 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand.
| | - Anna Brestovitsky
- The Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK
| | - Ruth Buning
- Biological and Soft Matter Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA Leiden, the Netherlands
| | - Charles Ravarani
- Medical Research Council Laboratory for Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Daniela Rhodes
- Medical Research Council Laboratory for Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Institute of Structural Biology, Nanyang Technical University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - John van Noort
- Biological and Soft Matter Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA Leiden, the Netherlands
| | - Katja E Jaeger
- The Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK
| | - Philip A Wigge
- The Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK.
| |
Collapse
|
266
|
Neueder A, Gipson TA, Batterton S, Lazell HJ, Farshim PP, Paganetti P, Housman DE, Bates GP. HSF1-dependent and -independent regulation of the mammalian in vivo heat shock response and its impairment in Huntington's disease mouse models. Sci Rep 2017; 7:12556. [PMID: 28970536 PMCID: PMC5624871 DOI: 10.1038/s41598-017-12897-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/30/2017] [Indexed: 01/20/2023] Open
Abstract
The heat shock response (HSR) is a mechanism to cope with proteotoxic stress by inducing the expression of molecular chaperones and other heat shock response genes. The HSR is evolutionarily well conserved and has been widely studied in bacteria, cell lines and lower eukaryotic model organisms. However, mechanistic insights into the HSR in higher eukaryotes, in particular in mammals, are limited. We have developed an in vivo heat shock protocol to analyze the HSR in mice and dissected heat shock factor 1 (HSF1)-dependent and -independent pathways. Whilst the induction of proteostasis-related genes was dependent on HSF1, the regulation of circadian function related genes, indicating that the circadian clock oscillators have been reset, was independent of its presence. Furthermore, we demonstrate that the in vivo HSR is impaired in mouse models of Huntington's disease but we were unable to corroborate the general repression of transcription that follows a heat shock in lower eukaryotes.
Collapse
Affiliation(s)
- Andreas Neueder
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom.
| | - Theresa A Gipson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
| | - Sophie Batterton
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Hayley J Lazell
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Pamela P Farshim
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Paolo Paganetti
- Neuroscience Discovery, Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
- Laboratory for Biomedical Neuroscience, Neurocenter of Southern Switzerland, EOC, c/o SIRM, Torricella-Taverne, Switzerland
| | - David E Housman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
| | - Gillian P Bates
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
267
|
Vilaboa N, Boré A, Martin-Saavedra F, Bayford M, Winfield N, Firth-Clark S, Kirton SB, Voellmy R. New inhibitor targeting human transcription factor HSF1: effects on the heat shock response and tumor cell survival. Nucleic Acids Res 2017; 45:5797-5817. [PMID: 28369544 PMCID: PMC5449623 DOI: 10.1093/nar/gkx194] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/13/2017] [Indexed: 12/20/2022] Open
Abstract
Comparative modeling of the DNA-binding domain of human HSF1 facilitated the prediction of possible binding pockets for small molecules and definition of corresponding pharmacophores. In silico screening of a large library of lead-like compounds identified a set of compounds that satisfied the pharmacophoric criteria, a selection of which compounds was purchased to populate a biased sublibrary. A discriminating cell-based screening assay identified compound 001, which was subjected to systematic analysis of structure–activity relationships, resulting in the development of compound 115 (IHSF115). IHSF115 bound to an isolated HSF1 DNA-binding domain fragment. The compound did not affect heat-induced oligomerization, nuclear localization and specific DNA binding but inhibited the transcriptional activity of human HSF1, interfering with the assembly of ATF1-containing transcription complexes. IHSF115 was employed to probe the human heat shock response at the transcriptome level. In contrast to earlier studies of differential regulation in HSF1-naïve and -depleted cells, our results suggest that a large majority of heat-induced genes is positively regulated by HSF1. That IHSF115 effectively countermanded repression in a significant fraction of heat-repressed genes suggests that repression of these genes is mediated by transcriptionally active HSF1. IHSF115 is cytotoxic for a variety of human cancer cell lines, multiple myeloma lines consistently exhibiting high sensitivity.
Collapse
Affiliation(s)
- Nuria Vilaboa
- Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain.,CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Alba Boré
- Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain.,CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Francisco Martin-Saavedra
- Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain.,CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Melanie Bayford
- Domainex Ltd, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Natalie Winfield
- Domainex Ltd, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Stuart Firth-Clark
- Domainex Ltd, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Stewart B Kirton
- University of Hertfordshire, Hatfield, Hertfordshire, AL10 9AB, UK
| | | |
Collapse
|
268
|
Comparative analysis reveals genomic features of stress-induced transcriptional readthrough. Proc Natl Acad Sci U S A 2017; 114:E8362-E8371. [PMID: 28928151 DOI: 10.1073/pnas.1711120114] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Transcription is a highly regulated process, and stress-induced changes in gene transcription have been shown to play a major role in stress responses and adaptation. Genome-wide studies reveal prevalent transcription beyond known protein-coding gene loci, generating a variety of RNA classes, most of unknown function. One such class, termed downstream of gene-containing transcripts (DoGs), was reported to result from transcriptional readthrough upon osmotic stress in human cells. However, how widespread the readthrough phenomenon is, and what its causes and consequences are, remain elusive. Here we present a genome-wide mapping of transcriptional readthrough, using nuclear RNA-Seq, comparing heat shock, osmotic stress, and oxidative stress in NIH 3T3 mouse fibroblast cells. We observe massive induction of transcriptional readthrough, both in levels and length, under all stress conditions, with significant, yet not complete, overlap of readthrough-induced loci between different conditions. Importantly, our analyses suggest that stress-induced transcriptional readthrough is not a random failure process, but is rather differentially induced across different conditions. We explore potential regulators and find a role for HSF1 in the induction of a subset of heat shock-induced readthrough transcripts. Analysis of public datasets detected increases in polymerase II occupancy in DoG regions after heat shock, supporting our findings. Interestingly, DoGs tend to be produced in the vicinity of neighboring genes, leading to a marked increase in their antisense-generating potential. Finally, we examine genomic features of readthrough transcription and observe a unique chromatin signature typical of DoG-producing regions, suggesting that readthrough transcription is associated with the maintenance of an open chromatin state.
Collapse
|
269
|
Li J, Labbadia J, Morimoto RI. Rethinking HSF1 in Stress, Development, and Organismal Health. Trends Cell Biol 2017; 27:895-905. [PMID: 28890254 DOI: 10.1016/j.tcb.2017.08.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 11/29/2022]
Abstract
The heat shock response (HSR) was originally discovered as a transcriptional response to elevated temperature shock and led to the identification of heat shock proteins and heat shock factor 1 (HSF1). Since then HSF1 has been shown to be important for combating other forms of environmental perturbations as well as genetic variations that cause proteotoxic stress. The HSR has long been thought to be an absolute response to conditions of cell stress and the primary mechanism by which HSF1 promotes organismal health by preventing protein aggregation and subsequent proteome imbalance. Accumulating evidence now shows that HSF1, the central player in the HSR, is regulated according to specific cellular requirements through cell-autonomous and non-autonomous signals, and directs transcriptional programs distinct from the HSR during development and in carcinogenesis. We discuss here these 'non-canonical' roles of HSF1, its regulation in diverse conditions of development, reproduction, metabolism, and aging, and posit that HSF1 serves to integrate diverse biological and pathological responses.
Collapse
Affiliation(s)
- Jian Li
- Department of Molecular Biosciences, Rice Institute for Biomedical Research Northwestern University, Evanston, IL 60208, USA; Present address: Functional and Chemical Genomics Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Johnathan Labbadia
- Department of Molecular Biosciences, Rice Institute for Biomedical Research Northwestern University, Evanston, IL 60208, USA; Present address: Institute of Healthy Ageing, Genetics, Evolution and Environment, University College London, WC1E 6BT, UK
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
270
|
Budzyński MA, Crul T, Himanen SV, Toth N, Otvos F, Sistonen L, Vigh L. Chaperone co-inducer BGP-15 inhibits histone deacetylases and enhances the heat shock response through increased chromatin accessibility. Cell Stress Chaperones 2017; 22:717-728. [PMID: 28474205 PMCID: PMC5573690 DOI: 10.1007/s12192-017-0798-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/22/2017] [Accepted: 04/07/2017] [Indexed: 01/20/2023] Open
Abstract
Defects in cellular protein homeostasis are associated with many severe and prevalent pathological conditions such as neurodegenerative diseases, muscle dystrophies, and metabolic disorders. One way to counteract these defects is to improve the protein homeostasis capacity through induction of the heat shock response. Despite numerous attempts to develop strategies for chemical activation of the heat shock response by heat shock transcription factor 1 (HSF1), the underlying mechanisms of drug candidates' mode of action are poorly understood. To lower the threshold for the heat shock response activation, we used the chaperone co-inducer BGP-15 that was previously shown to have beneficial effects on several proteinopathic disease models. We found that BGP-15 treatment combined with heat stress caused a substantial increase in HSF1-dependent heat shock protein 70 (HSPA1A/B) expression already at a febrile range of temperatures. Moreover, BGP-15 alone inhibited the activity of histone deacetylases (HDACs), thereby increasing chromatin accessibility at multiple genomic loci including the stress-inducible HSPA1A. Intriguingly, treatment with well-known potent HDAC inhibitors trichostatin A and valproic acid enhanced the heat shock response and improved cytoprotection. These results present a new pharmacological strategy for restoring protein homeostasis by inhibiting HDACs, increasing chromatin accessibility, and lowering the threshold for heat shock response activation.
Collapse
Affiliation(s)
- Marek A Budzyński
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20520, Turku, Finland
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| | - Tim Crul
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary
| | - Samu V Himanen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20520, Turku, Finland
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| | - Noemi Toth
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary
| | - Ferenc Otvos
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20520, Turku, Finland.
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland.
| | - Laszlo Vigh
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary.
| |
Collapse
|
271
|
Chen FX, Xie P, Collings CK, Cao K, Aoi Y, Marshall SA, Rendleman EJ, Ugarenko M, Ozark PA, Zhang A, Shiekhattar R, Smith ER, Zhang MQ, Shilatifard A. PAF1 regulation of promoter-proximal pause release via enhancer activation. Science 2017; 357:1294-1298. [PMID: 28860207 DOI: 10.1126/science.aan3269] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 08/22/2017] [Indexed: 12/14/2022]
Abstract
Gene expression in metazoans is regulated by RNA polymerase II (Pol II) promoter-proximal pausing and its release. Previously, we showed that Pol II-associated factor 1 (PAF1) modulates the release of paused Pol II into productive elongation. Here, we found that PAF1 occupies transcriptional enhancers and restrains hyperactivation of a subset of these enhancers. Enhancer activation as the result of PAF1 loss releases Pol II from paused promoters of nearby PAF1 target genes. Knockout of PAF1-regulated enhancers attenuates the release of paused Pol II on PAF1 target genes without major interference in the establishment of pausing at their cognate promoters. Thus, a subset of enhancers can primarily modulate gene expression by controlling the release of paused Pol II in a PAF1-dependent manner.
Collapse
Affiliation(s)
- Fei Xavier Chen
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peng Xie
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Clayton K Collings
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kaixiang Cao
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yuki Aoi
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Stacy A Marshall
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Emily J Rendleman
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michal Ugarenko
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patrick A Ozark
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anda Zhang
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ramin Shiekhattar
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Edwin R Smith
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA.,MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST, Tsinghua University, Beijing 100084, China
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
272
|
A Model of Exposure to Extreme Environmental Heat Uncovers the Human Transcriptome to Heat Stress. Sci Rep 2017; 7:9429. [PMID: 28842615 PMCID: PMC5573409 DOI: 10.1038/s41598-017-09819-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 07/31/2017] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms by which individuals subjected to environmental heat stress either recover or develop heat-related complications are not well understood. We analysed the changes in blood mononuclear gene expression patterns in human volunteers exposed to extreme heat in a sauna (temperature of 75.7 ± 0.86 °C). Our analysis reveals that expression changes occur rapidly with no significant increase in core temperature and continue to amplify one hour after the end of heat stress. The reprogramed transcriptome was predominantly inhibitory, as more than two-thirds of the expressed genes were suppressed. The differentially expressed genes encoded proteins that function in stress-associated pathways; including proteostasis, energy metabolism, cell growth and proliferation, and cell death, and survival. The transcriptome also included mitochondrial dysfunction, altered protein synthesis, and reduced expression of genes -related to immune function. The findings reveal the human transcriptomic response to heat and highlight changes that might underlie the health outcomes observed during heat waves.
Collapse
|
273
|
McKinstry M, Chung C, Truong H, Johnston BA, Snow JW. The heat shock response and humoral immune response are mutually antagonistic in honey bees. Sci Rep 2017; 7:8850. [PMID: 28821863 PMCID: PMC5562734 DOI: 10.1038/s41598-017-09159-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/21/2017] [Indexed: 11/24/2022] Open
Abstract
The honey bee is of paramount importance to humans in both agricultural and ecological settings. Honey bee colonies have suffered from increased attrition in recent years, stemming from complex interacting stresses. Defining common cellular stress responses elicited by these stressors represents a key step in understanding potential synergies. The proteostasis network is a highly conserved network of cellular stress responses involved in maintaining the homeostasis of protein production and function. Here, we have characterized the Heat Shock Response (HSR), one branch of this network, and found that its core components are conserved. In addition, exposing bees to elevated temperatures normally encountered by honey bees during typical activities results in robust HSR induction with increased expression of specific heat shock proteins that was variable across tissues. Surprisingly, we found that heat shock represses multiple immune genes in the abdomen and additionally showed that wounding the cuticle of the abdomen results in decreased expression of multiple HSR genes in proximal and distal tissues. This mutually antagonistic relationship between the HSR and immune activation is unique among invertebrates studied to date and may promote understanding of potential synergistic effects of disparate stresses in this critical pollinator and social insects more broadly.
Collapse
Affiliation(s)
- Mia McKinstry
- Biology Department, Barnard College, New York, NY, 10027, USA
| | - Charlie Chung
- Natural Sciences Department, LaGuardia Community College-CUNY, Long Island City, NY, 11101, USA
| | - Henry Truong
- Biology Department, Barnard College, New York, NY, 10027, USA
| | - Brittany A Johnston
- Biology Department, The City College of New York-CUNY, New York, NY, 10031, USA
| | - Jonathan W Snow
- Biology Department, Barnard College, New York, NY, 10027, USA.
| |
Collapse
|
274
|
Vihervaara A, Mahat DB, Guertin MJ, Chu T, Danko CG, Lis JT, Sistonen L. Transcriptional response to stress is pre-wired by promoter and enhancer architecture. Nat Commun 2017; 8:255. [PMID: 28811569 PMCID: PMC5557961 DOI: 10.1038/s41467-017-00151-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/05/2017] [Indexed: 12/29/2022] Open
Abstract
Programs of gene expression are executed by a battery of transcription factors that coordinate divergent transcription from a pair of tightly linked core initiation regions of promoters and enhancers. Here, to investigate how divergent transcription is reprogrammed upon stress, we measured nascent RNA synthesis at nucleotide-resolution, and profiled histone H4 acetylation in human cells. Our results globally show that the release of promoter-proximal paused RNA polymerase into elongation functions as a critical switch at which a gene's response to stress is determined. Highly transcribed and highly inducible genes display strong transcriptional directionality and selective assembly of general transcription factors on the core sense promoter. Heat-induced transcription at enhancers, instead, correlates with prior binding of cell-type, sequence-specific transcription factors. Activated Heat Shock Factor 1 (HSF1) binds to transcription-primed promoters and enhancers, and CTCF-occupied, non-transcribed chromatin. These results reveal chromatin architectural features that orient transcription at divergent regulatory elements and prime transcriptional responses genome-wide.Heat Shock Factor 1 (HSF1) is a regulator of stress-induced transcription. Here, the authors investigate changes to transcription and chromatin organization upon stress and find that activated HSF1 binds to transcription-primed promoters and enhancers, and to CTCF occupied, untranscribed chromatin.
Collapse
Affiliation(s)
- Anniina Vihervaara
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, 20520, Finland
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA
| | - Dig Bijay Mahat
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA
| | - Michael J Guertin
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Tinyi Chu
- Department of Biomedical Sciences, The Baker Institute for Animal Health, Cornell University, Ithaca, New York, 14853, USA
- Graduate Field of Computational Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Charles G Danko
- Department of Biomedical Sciences, The Baker Institute for Animal Health, Cornell University, Ithaca, New York, 14853, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA.
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, 20520, Finland.
| |
Collapse
|
275
|
Abstract
During aging, the mechanisms that normally maintain health and stress resistance strikingly decline, resulting in decrepitude, frailty, and ultimately death. Exactly when and how this decline occurs is unknown. Changes in transcriptional networks and chromatin state lie at the heart of age-dependent decline. These epigenomic changes are not only observed during aging but also profoundly affect cellular function and stress resistance, thereby contributing to the progression of aging. We propose that the dysregulation of transcriptional and chromatin networks is a crucial component of aging. Understanding age-dependent epigenomic changes will yield key insights into how aging begins and progresses and should lead to the development of new therapeutics that delay or even reverse aging and age-related diseases.
Collapse
Affiliation(s)
- Lauren N Booth
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA.
| |
Collapse
|
276
|
Multisubunit DNA-Dependent RNA Polymerases from Vaccinia Virus and Other Nucleocytoplasmic Large-DNA Viruses: Impressions from the Age of Structure. Microbiol Mol Biol Rev 2017; 81:81/3/e00010-17. [PMID: 28701329 DOI: 10.1128/mmbr.00010-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The past 17 years have been marked by a revolution in our understanding of cellular multisubunit DNA-dependent RNA polymerases (MSDDRPs) at the structural level. A parallel development over the past 15 years has been the emerging story of the giant viruses, which encode MSDDRPs. Here we link the two in an attempt to understand the specialization of multisubunit RNA polymerases in the domain of life encompassing the large nucleocytoplasmic DNA viruses (NCLDV), a superclade that includes the giant viruses and the biochemically well-characterized poxvirus vaccinia virus. The first half of this review surveys the recently determined structural biology of cellular RNA polymerases for a microbiology readership. The second half discusses a reannotation of MSDDRP subunits from NCLDV families and the apparent specialization of these enzymes by virus family and by subunit with regard to subunit or domain loss, subunit dissociability, endogenous control of polymerase arrest, and the elimination/customization of regulatory interactions that would confer higher-order cellular control. Some themes are apparent in linking subunit function to structure in the viral world: as with cellular RNA polymerases I and III and unlike cellular RNA polymerase II, the viral enzymes seem to opt for speed and processivity and seem to have eliminated domains associated with higher-order regulation. The adoption/loss of viral RNA polymerase proofreading functions may have played a part in matching intrinsic mutability to genome size.
Collapse
|
277
|
Genome-wide characterization of mammalian promoters with distal enhancer functions. Nat Genet 2017; 49:1073-1081. [PMID: 28581502 DOI: 10.1038/ng.3884] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
Gene expression in mammals is precisely regulated by the combination of promoters and gene-distal regulatory regions, known as enhancers. Several studies have suggested that some promoters might have enhancer functions. However, the extent of this type of promoters and whether they actually function to regulate the expression of distal genes have remained elusive. Here, by exploiting a high-throughput enhancer reporter assay, we unravel a set of mammalian promoters displaying enhancer activity. These promoters have distinct genomic and epigenomic features and frequently interact with other gene promoters. Extensive CRISPR-Cas9 genomic manipulation demonstrated the involvement of these promoters in the cis regulation of expression of distal genes in their natural loci. Our results have important implications for the understanding of complex gene regulation in normal development and disease.
Collapse
|
278
|
Spanier KI, Jansen M, Decaestecker E, Hulselmans G, Becker D, Colbourne JK, Orsini L, De Meester L, Aerts S. Conserved Transcription Factors Steer Growth-Related Genomic Programs in Daphnia. Genome Biol Evol 2017; 9:1821-1842. [PMID: 28854641 PMCID: PMC5569996 DOI: 10.1093/gbe/evx127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2017] [Indexed: 02/06/2023] Open
Abstract
Ecological genomics aims to understand the functional association between environmental gradients and the genes underlying adaptive traits. Many genes that are identified by genome-wide screening in ecologically relevant species lack functional annotations. Although gene functions can be inferred from sequence homology, such approaches have limited power. Here, we introduce ecological regulatory genomics by presenting an ontology-free gene prioritization method. Specifically, our method combines transcriptome profiling with high-throughput cis-regulatory sequence analysis in the water fleas Daphnia pulex and Daphnia magna. It screens coexpressed genes for overrepresented DNA motifs that serve as transcription factor binding sites, thereby providing insight into conserved transcription factors and gene regulatory networks shaping the expression profile. We first validated our method, called Daphnia-cisTarget, on a D. pulex heat shock data set, which revealed a network driven by the heat shock factor. Next, we performed RNA-Seq in D. magna exposed to the cyanobacterium Microcystis aeruginosa. Daphnia-cisTarget identified coregulated gene networks that associate with the moulting cycle and potentially regulate life history changes in growth rate and age at maturity. These networks are predicted to be regulated by evolutionary conserved transcription factors such as the homologues of Drosophila Shavenbaby and Grainyhead, nuclear receptors, and a GATA family member. In conclusion, our approach allows prioritising candidate genes in Daphnia without bias towards prior knowledge about functional gene annotation and represents an important step towards exploring the molecular mechanisms of ecological responses in organisms with poorly annotated genomes.
Collapse
Affiliation(s)
- Katina I. Spanier
- Department of Biology, Laboratory of Aquatic Ecology, Evolution and Conservation, KU Leuven, Belgium
- Department of Human Genetics, Laboratory of Computational Biology, KU Leuven, Belgium
- VIB Center for Brain and Disease Research, KU Leuven, Belgium
| | - Mieke Jansen
- Department of Biology, Laboratory of Aquatic Ecology, Evolution and Conservation, KU Leuven, Belgium
| | - Ellen Decaestecker
- Department of Biology, Laboratory of Aquatic Biology, Science and Technology, KU Leuven Campus Kulak, Kortrjik, Belgium
| | - Gert Hulselmans
- Department of Human Genetics, Laboratory of Computational Biology, KU Leuven, Belgium
- VIB Center for Brain and Disease Research, KU Leuven, Belgium
| | - Dörthe Becker
- Environmental Genomics Group, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, United Kingdom
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, United Kingdom
| | - John K. Colbourne
- Environmental Genomics Group, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, United Kingdom
| | - Luisa Orsini
- Environmental Genomics Group, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, United Kingdom
| | - Luc De Meester
- Department of Biology, Laboratory of Aquatic Ecology, Evolution and Conservation, KU Leuven, Belgium
| | - Stein Aerts
- Department of Human Genetics, Laboratory of Computational Biology, KU Leuven, Belgium
- VIB Center for Brain and Disease Research, KU Leuven, Belgium
| |
Collapse
|
279
|
Iyengar BR, Pillai B, Venkatesh KV, Gadgil CJ. Systematic comparison of the response properties of protein and RNA mediated gene regulatory motifs. MOLECULAR BIOSYSTEMS 2017; 13:1235-1245. [PMID: 28485414 DOI: 10.1039/c6mb00808a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We present a framework enabling the dissection of the effects of motif structure (feedback or feedforward), the nature of the controller (RNA or protein), and the regulation mode (transcriptional, post-transcriptional or translational) on the response to a step change in the input. We have used a common model framework for gene expression where both motif structures have an activating input and repressing regulator, with the same set of parameters, to enable a comparison of the responses. We studied the global sensitivity of the system properties, such as steady-state gain, overshoot, peak time, and peak duration, to parameters. We find that, in all motifs, overshoot correlated negatively whereas peak duration varied concavely with peak time. Differences in the other system properties were found to be mainly dependent on the nature of the controller rather than the motif structure. Protein mediated motifs showed a higher degree of adaptation i.e. a tendency to return to baseline levels; in particular, feedforward motifs exhibited perfect adaptation. RNA mediated motifs had a mild regulatory effect; they also exhibited a lower peaking tendency and mean overshoot. Protein mediated feedforward motifs showed higher overshoot and lower peak time compared to the corresponding feedback motifs.
Collapse
|
280
|
Zovoilis A, Cifuentes-Rojas C, Chu HP, Hernandez AJ, Lee JT. Destabilization of B2 RNA by EZH2 Activates the Stress Response. Cell 2017; 167:1788-1802.e13. [PMID: 27984727 DOI: 10.1016/j.cell.2016.11.041] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022]
Abstract
More than 98% of the mammalian genome is noncoding, and interspersed transposable elements account for ∼50% of noncoding space. Here, we demonstrate that a specific interaction between the polycomb protein EZH2 and RNA made from B2 SINE retrotransposons controls stress-responsive genes in mouse cells. In the heat-shock model, B2 RNA binds stress genes and suppresses their transcription. Upon stress, EZH2 is recruited and triggers cleavage of B2 RNA. B2 degradation in turn upregulates stress genes. Evidence indicates that B2 RNA operates as a "speed bump" against advancement of RNA polymerase II, and temperature stress releases the brakes on transcriptional elongation. These data attribute a new function to EZH2 that is independent of its histone methyltransferase activity and reconcile how EZH2 can be associated with both gene repression and activation. Our study reveals that EZH2 and B2 together control activation of a large network of genes involved in thermal stress.
Collapse
Affiliation(s)
- Athanasios Zovoilis
- Howard Hughes Medical Institute; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | - Catherine Cifuentes-Rojas
- Howard Hughes Medical Institute; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | - Hsueh-Ping Chu
- Howard Hughes Medical Institute; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | - Alfredo J Hernandez
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeannie T Lee
- Howard Hughes Medical Institute; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
281
|
Niskanen EA, Palvimo JJ. Chromatin SUMOylation in heat stress: To protect, pause and organise?: SUMO stress response on chromatin. Bioessays 2017; 39. [PMID: 28440894 DOI: 10.1002/bies.201600263] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Post-translational modifications, e.g. SUMO modifications (SUMOylation), provide a mechanism for swiftly changing a protein's activity. Various stress conditions trigger a SUMO stress response (SSR) - a stress-induced rapid change in the conjugation of SUMO to multiple proteins, which predominantly targets nuclear proteins. The SSR has been postulated to protect stressed cells by preserving the functionality of crucial proteins. However, it is unclear how it exerts its protective functions. Interestingly, heat stress (HS) increases SUMOylation of proteins at active promoters and enhancers. In promoters, HS-induced SUMOylation correlates with gene transcription and stress-induced RNA polymerase II (Pol2) pausing. Conversely, a disappearance of SUMOylation in HS occurs at chromatin anchor points that maintain chromatin-looping structures and the spatial organisation of chromatin. In reviewing the literature, we hypothesise that the SSR regulates Pol2 pausing by modulating the interactions of pausing-regulating proteins, whereas deSUMOylation alters the function of chromatin anchors.
Collapse
Affiliation(s)
- Einari A Niskanen
- University of Eastern Finland, Institute of Biomedicine, Kuopio, Finland
| | - Jorma J Palvimo
- University of Eastern Finland, Institute of Biomedicine, Kuopio, Finland
| |
Collapse
|
282
|
Shining a light on early stress responses and late-onset disease vulnerability. Proc Natl Acad Sci U S A 2017; 114:2109-2111. [PMID: 28179564 DOI: 10.1073/pnas.1700323114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
283
|
Abstract
The leap from simple unicellularity to complex multicellularity remains one of life's major enigmas. The origins of metazoan developmental gene regulatory mechanisms are sought by analyzing gene regulation in extant eumetazoans, sponges, and unicellular organisms. The main hypothesis of this manuscript is that, developmental enhancers evolved from unicellular inducible promoters that diversified the expression of regulatory genes during metazoan evolution. Promoters and enhancers are functionally similar; both can regulate the transcription of distal promoters and both direct local transcription. Additionally, enhancers have experimentally characterized structural features that reveal their origin from inducible promoters. The distal co-operative regulation among promoters identified in unicellular opisthokonts possibly represents the precursor of distal regulation of promoters by enhancers. During metazoan evolution, constitutive-type promoters of regulatory genes would have acquired novel receptivity to distal regulatory inputs from promoters of inducible genes that eventually specialized as enhancers. The novel regulatory interactions would have caused constitutively expressed genes controlling differential gene expression in unicellular organisms to become themselves differentially expressed. The consequence of the novel regulatory interactions was that regulatory pathways of unicellular organisms became interlaced and ultimately evolved into the intricate developmental gene regulatory networks (GRNs) of extant metazoans.
Collapse
Affiliation(s)
- César Arenas-Mena
- Department of Biology, College of Staten Island and Graduate Center, The City University of New York (CUNY), Staten Island, NY 10314, USA
| |
Collapse
|
284
|
Mahat DB, Lis JT. Use of conditioned media is critical for studies of regulation in response to rapid heat shock. Cell Stress Chaperones 2017; 22:155-162. [PMID: 27812889 PMCID: PMC5225056 DOI: 10.1007/s12192-016-0737-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022] Open
Abstract
Heat shock response (HSR) maintains and restores protein homeostasis when cells are exposed to proteotoxic heat stress. Heat shock (HS) triggers a rapid and robust change in genome-wide transcription, protein synthesis, and chaperone activity; and therefore, the HSR has been widely used as a model system in these studies. The conventional method of performing instantaneous HS in the laboratory uses heated fresh media to induce HSR when added to cells. However, addition of fresh media to cells may evoke additional cellular responses and signaling pathways. Here, we compared the change in global transcription profile when HS is performed with either heated fresh media or heated conditioned media. We found that the use of heated fresh media induces transcription of hundreds of genes that HS alone does not induce, and masks or partially masks HS-mediated downregulation of thousands of genes. The fresh-media-dependent upregulated genes encode ribosomal subunit proteins involved in translation and RNA processing factors. More importantly, fresh media also induce transcription of several heat shock protein genes (Hsps) in a heat shock factor 1 (HSF1)-independent manner. Thus, we conclude that a conventional method of HS with heated fresh media causes changes in transcription regulation that confound the actual change caused solely by elevated temperature of cells.
Collapse
Affiliation(s)
- Dig B Mahat
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14850, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
285
|
Zheng X, Krakowiak J, Patel N, Beyzavi A, Ezike J, Khalil AS, Pincus D. Dynamic control of Hsf1 during heat shock by a chaperone switch and phosphorylation. eLife 2016; 5. [PMID: 27831465 PMCID: PMC5127643 DOI: 10.7554/elife.18638] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023] Open
Abstract
Heat shock factor (Hsf1) regulates the expression of molecular chaperones to maintain protein homeostasis. Despite its central role in stress resistance, disease and aging, the mechanisms that control Hsf1 activity remain unresolved. Here we show that in budding yeast, Hsf1 basally associates with the chaperone Hsp70 and this association is transiently disrupted by heat shock, providing the first evidence that a chaperone repressor directly regulates Hsf1 activity. We develop and experimentally validate a mathematical model of Hsf1 activation by heat shock in which unfolded proteins compete with Hsf1 for binding to Hsp70. Surprisingly, we find that Hsf1 phosphorylation, previously thought to be required for activation, in fact only positively tunes Hsf1 and does so without affecting Hsp70 binding. Our work reveals two uncoupled forms of regulation - an ON/OFF chaperone switch and a tunable phosphorylation gain - that allow Hsf1 to flexibly integrate signals from the proteostasis network and cell signaling pathways. DOI:http://dx.doi.org/10.7554/eLife.18638.001 Proteins are strings of amino acids that carry out crucial activities inside cells, such as harvesting energy and generating the building blocks that cells need to grow. In order to carry out their specific roles inside the cell, the proteins need to “fold” into precise three-dimensional shapes. Protein folding is critical for life, and cells don’t leave it up to chance. Cells employ “molecular chaperones” to help proteins to fold properly. However, under some conditions – such as high temperature – proteins are more difficult to fold and the chaperones can become overwhelmed. In these cases, unfolded proteins can pile up in the cell. This leads not only to the cell being unable to work properly, but also to the formation of toxic “aggregates”. These aggregates are tangles of unfolded proteins that are hallmarks of many neurodegenerative diseases such as Alzheimer’s, Parkinson’s and amyotrophic lateral sclerosis (ALS). Protein aggregates can be triggered by high temperature in a condition termed “heat shock”. A sensor named heat shock factor 1 (Hsf1 for short) increases the amount of chaperones following heat shock. But what controls the activity of Hsf1? To answer this question, Zheng, Krakowiak et al. combined mathematical modelling and experiments in yeast cells. The most important finding is that the ‘on/off switch’ that controls Hsf1 is based on whether Hsf1 is itself bound to a chaperone. When bound to the chaperone, Hsf1 is turned ‘off’; when the chaperone falls off, Hsf1 turns ‘on’ and makes more chaperones; when there are enough chaperones, they once again bind to Hsf1 and turn it back ‘off’. In this way, Hsf1 and the chaperones form a feedback loop that ensures that there are always enough chaperones to keep the cell’s proteins folded. Now that we know how Hsf1 is controlled, can we harness this understanding to tune the activity of Hsf1 without disrupting how the chaperones work? If we can activate Hsf1, we can provide cells with more chaperones. This could be a therapeutic strategy to combat neurodegenerative diseases. DOI:http://dx.doi.org/10.7554/eLife.18638.002
Collapse
Affiliation(s)
- Xu Zheng
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Joanna Krakowiak
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Nikit Patel
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States
| | - Ali Beyzavi
- Department of Mechanical Engineering, Boston University, Boston, United States
| | - Jideofor Ezike
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Ahmad S Khalil
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, United States
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, United States
| |
Collapse
|
286
|
Zhao Y, Liu Q, Acharya P, Stengel KR, Sheng Q, Zhou X, Kwak H, Fischer MA, Bradner JE, Strickland SA, Mohan SR, Savona MR, Venters BJ, Zhou MM, Lis JT, Hiebert SW. High-Resolution Mapping of RNA Polymerases Identifies Mechanisms of Sensitivity and Resistance to BET Inhibitors in t(8;21) AML. Cell Rep 2016; 16:2003-16. [PMID: 27498870 PMCID: PMC4996374 DOI: 10.1016/j.celrep.2016.07.032] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 12/17/2015] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Bromodomain and extra-terminal domain (BET) family inhibitors offer an approach to treating hematological malignancies. We used precision nuclear run-on transcription sequencing (PRO-seq) to create high-resolution maps of active RNA polymerases across the genome in t(8;21) acute myeloid leukemia (AML), as these polymerases are exceptionally sensitive to BET inhibitors. PRO-seq identified over 1,400 genes showing impaired release of promoter-proximal paused RNA polymerases, including the stem cell factor receptor tyrosine kinase KIT that is mutated in t(8;21) AML. PRO-seq also identified an enhancer 3' to KIT. Chromosome conformation capture confirmed contacts between this enhancer and the KIT promoter, while CRISPRi-mediated repression of this enhancer impaired cell growth. PRO-seq also identified microRNAs, including MIR29C and MIR29B2, that target the anti-apoptotic factor MCL1 and were repressed by BET inhibitors. MCL1 protein was upregulated, and inhibition of BET proteins sensitized t(8:21)-containing cells to MCL1 inhibition, suggesting a potential mechanism of resistance to BET-inhibitor-induced cell death.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Azepines/pharmacology
- Cell Line, Tumor
- Chromosomes, Human, Pair 21
- Chromosomes, Human, Pair 8
- Clustered Regularly Interspaced Short Palindromic Repeats
- DNA-Directed RNA Polymerases/genetics
- DNA-Directed RNA Polymerases/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enhancer Elements, Genetic
- Gene Expression Regulation, Leukemic
- High-Throughput Nucleotide Sequencing/methods
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Multigene Family
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Promoter Regions, Genetic
- Protein Isoforms/antagonists & inhibitors
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Proteins/antagonists & inhibitors
- Proteins/genetics
- Proteins/metabolism
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-kit/metabolism
- Transcription, Genetic
- Translocation, Genetic
- Triazoles/pharmacology
Collapse
Affiliation(s)
- Yue Zhao
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pankaj Acharya
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristy R Stengel
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Quanhu Sheng
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Xiaofan Zhou
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37212, USA
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Melissa A Fischer
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Stephen A Strickland
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sanjay R Mohan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael R Savona
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bryan J Venters
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ming-Ming Zhou
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Scott W Hiebert
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
287
|
Pincus D. Size doesn't matter in the heat shock response. Curr Genet 2016; 63:175-178. [PMID: 27502399 DOI: 10.1007/s00294-016-0638-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 11/25/2022]
Abstract
Heat shock factor 1 (Hsf1) is a transcription factor that is often described as the master regulator of the heat shock response in all eukaryotes. However, due to its essentiality in yeast, Hsf1's contribution to the transcriptome under basal and heat shock conditions has never been directly determined. Using a chemical genetics approach that allowed rapid Hsf1 inactivation, my colleagues and I have recently shown that the bulk of the heat shock response is Hsf1 independent. Rather than inducing genes responsible for carrying out the various cellular processes required for adaptation to thermal stress, Hsf1 controls a dedicated set of chaperone protein genes devoted to restoring protein-folding homeostasis. The limited scope of the Hsf1 regulon belies its outsize importance in cellular fitness.
Collapse
Affiliation(s)
- David Pincus
- Nine Cambridge Center, Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.
| |
Collapse
|
288
|
Mahat DB, Kwak H, Booth GT, Jonkers IH, Danko CG, Patel RK, Waters CT, Munson K, Core LJ, Lis JT. Base-pair-resolution genome-wide mapping of active RNA polymerases using precision nuclear run-on (PRO-seq). Nat Protoc 2016; 11:1455-76. [PMID: 27442863 PMCID: PMC5502525 DOI: 10.1038/nprot.2016.086] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We provide a protocol for precision nuclear run-on sequencing (PRO-seq) and its variant, PRO-cap, which map the location of active RNA polymerases (PRO-seq) or transcription start sites (TSSs) (PRO-cap) genome-wide at high resolution. The density of RNA polymerases at a particular genomic locus directly reflects the level of nascent transcription at that region. Nuclei are isolated from cells and, under nuclear run-on conditions, transcriptionally engaged RNA polymerases incorporate one or, at most, a few biotin-labeled nucleotide triphosphates (biotin-NTPs) into the 3' end of nascent RNA. The biotin-labeled nascent RNA is used to prepare sequencing libraries, which are sequenced from the 3' end to provide high-resolution positional information for the RNA polymerases. PRO-seq provides much higher sensitivity than ChIP-seq, and it generates a much larger fraction of usable sequence reads than ChIP-seq or NET-seq (native elongating transcript sequencing). Similarly to NET-seq, PRO-seq maps the RNA polymerase at up to base-pair resolution with strand specificity, but unlike NET-seq it does not require immunoprecipitation. With the protocol provided here, PRO-seq (or PRO-cap) libraries for high-throughput sequencing can be generated in 4-5 working days. The method has been applied to human, mouse, Drosophila melanogaster and Caenorhabditis elegans cells and, with slight modifications, to yeast.
Collapse
Affiliation(s)
- Dig Bijay Mahat
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Gregory T Booth
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Iris H Jonkers
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Charles G Danko
- The Baker Institute of Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Ravi K Patel
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Colin T Waters
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Katie Munson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Leighton J Core
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| |
Collapse
|
289
|
Duarte FM, Fuda NJ, Mahat DB, Core LJ, Guertin MJ, Lis JT. Transcription factors GAF and HSF act at distinct regulatory steps to modulate stress-induced gene activation. Genes Dev 2016; 30:1731-46. [PMID: 27492368 PMCID: PMC5002978 DOI: 10.1101/gad.284430.116] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/11/2016] [Indexed: 12/24/2022]
Abstract
The coordinated regulation of gene expression at the transcriptional level is fundamental to development and homeostasis. Inducible systems are invaluable when studying transcription because the regulatory process can be triggered instantaneously, allowing the tracking of ordered mechanistic events. Here, we use precision run-on sequencing (PRO-seq) to examine the genome-wide heat shock (HS) response in Drosophila and the function of two key transcription factors on the immediate transcription activation or repression of all genes regulated by HS. We identify the primary HS response genes and the rate-limiting steps in the transcription cycle that GAGA-associated factor (GAF) and HS factor (HSF) regulate. We demonstrate that GAF acts upstream of promoter-proximally paused RNA polymerase II (Pol II) formation (likely at the step of chromatin opening) and that GAF-facilitated Pol II pausing is critical for HS activation. In contrast, HSF is dispensable for establishing or maintaining Pol II pausing but is critical for the release of paused Pol II into the gene body at a subset of highly activated genes. Additionally, HSF has no detectable role in the rapid HS repression of thousands of genes.
Collapse
Affiliation(s)
- Fabiana M Duarte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14835, USA
| | - Nicholas J Fuda
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14835, USA
| | - Dig B Mahat
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14835, USA
| | - Leighton J Core
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14835, USA
| | - Michael J Guertin
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia 22908, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14835, USA
| |
Collapse
|
290
|
Solís EJ, Pandey JP, Zheng X, Jin DX, Gupta PB, Airoldi EM, Pincus D, Denic V. Defining the Essential Function of Yeast Hsf1 Reveals a Compact Transcriptional Program for Maintaining Eukaryotic Proteostasis. Mol Cell 2016; 63:60-71. [PMID: 27320198 PMCID: PMC4938784 DOI: 10.1016/j.molcel.2016.05.014] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/22/2016] [Accepted: 05/11/2016] [Indexed: 11/18/2022]
Abstract
Despite its eponymous association with the heat shock response, yeast heat shock factor 1 (Hsf1) is essential even at low temperatures. Here we show that engineered nuclear export of Hsf1 results in cytotoxicity associated with massive protein aggregation. Genome-wide analysis revealed that Hsf1 nuclear export immediately decreased basal transcription and mRNA expression of 18 genes, which predominately encode chaperones. Strikingly, rescuing basal expression of Hsp70 and Hsp90 chaperones enabled robust cell growth in the complete absence of Hsf1. With the exception of chaperone gene induction, the vast majority of the heat shock response was Hsf1 independent. By comparative analysis of mammalian cell lines, we found that only heat shock-induced but not basal expression of chaperones is dependent on the mammalian Hsf1 homolog (HSF1). Our work reveals that yeast chaperone gene expression is an essential housekeeping mechanism and provides a roadmap for defining the function of HSF1 as a driver of oncogenesis.
Collapse
Affiliation(s)
- Eric J Solís
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, MA 02138, USA; Systems Biology PhD Program, Harvard University, Cambridge, MA 02138, USA
| | - Jai P Pandey
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Xu Zheng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Dexter X Jin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Piyush B Gupta
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Edoardo M Airoldi
- Department of Statistics, Harvard University, Cambridge, MA 02138, USA
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Vladimir Denic
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|