1
|
Waldron R, Rodriguez MDLAB, Williams JM, Ning Z, Ahmed A, Lindsay A, Moore T. JRK binds satellite III DNA and is necessary for the heat shock response. Cell Biol Int 2024; 48:1212-1222. [PMID: 38946594 DOI: 10.1002/cbin.12216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
JRK is a DNA-binding protein of the pogo superfamily of transposons, which includes the well-known centromere binding protein B (CENP-B). Jrk null mice exhibit epilepsy, and growth and reproductive disorders, consistent with its relatively high expression in the brain and reproductive tissues. Human JRK DNA variants and gene expression levels are implicated in cancers and neuropsychiatric disorders. JRK protein modulates β-catenin-TCF activity but little is known of its cellular functions. Based on its homology to CENP-B, we determined whether JRK binds centromeric or other satellite DNAs. We show that human JRK binds satellite III DNA, which is abundant at the chromosome 9q12 juxtacentromeric region and on Yq12, both sites of nuclear stress body assembly. Human JRK-GFP overexpressed in HeLa cells strongly localises to 9q12. Using an anti-JRK antiserum we show that endogenous JRK co-localises with a subset of centromeres in non-stressed cells, and with heat shock factor 1 following heat shock. Knockdown of JRK in HeLa cells proportionately reduces heat shock protein gene expression in heat-shocked cells. A role for JRK in regulating the heat shock response is consistent with the mouse Jrk null phenotype and suggests that human JRK may act as a modifier of diseases with a cellular stress component.
Collapse
Affiliation(s)
- Rosalie Waldron
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | | | - John M Williams
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Zhenfei Ning
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Abrar Ahmed
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Andrew Lindsay
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Tom Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Kim H, Gomez-Pastor R. HSF1 and Its Role in Huntington's Disease Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:35-95. [PMID: 36396925 PMCID: PMC12001818 DOI: 10.1007/5584_2022_742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW Heat shock factor 1 (HSF1) is the master transcriptional regulator of the heat shock response (HSR) in mammalian cells and is a critical element in maintaining protein homeostasis. HSF1 functions at the center of many physiological processes like embryogenesis, metabolism, immune response, aging, cancer, and neurodegeneration. However, the mechanisms that allow HSF1 to control these different biological and pathophysiological processes are not fully understood. This review focuses on Huntington's disease (HD), a neurodegenerative disease characterized by severe protein aggregation of the huntingtin (HTT) protein. The aggregation of HTT, in turn, leads to a halt in the function of HSF1. Understanding the pathways that regulate HSF1 in different contexts like HD may hold the key to understanding the pathomechanisms underlying other proteinopathies. We provide the most current information on HSF1 structure, function, and regulation, emphasizing HD, and discussing its potential as a biological target for therapy. DATA SOURCES We performed PubMed search to find established and recent reports in HSF1, heat shock proteins (Hsp), HD, Hsp inhibitors, HSF1 activators, and HSF1 in aging, inflammation, cancer, brain development, mitochondria, synaptic plasticity, polyglutamine (polyQ) diseases, and HD. STUDY SELECTIONS Research and review articles that described the mechanisms of action of HSF1 were selected based on terms used in PubMed search. RESULTS HSF1 plays a crucial role in the progression of HD and other protein-misfolding related neurodegenerative diseases. Different animal models of HD, as well as postmortem brains of patients with HD, reveal a connection between the levels of HSF1 and HSF1 dysfunction to mutant HTT (mHTT)-induced toxicity and protein aggregation, dysregulation of the ubiquitin-proteasome system (UPS), oxidative stress, mitochondrial dysfunction, and disruption of the structural and functional integrity of synaptic connections, which eventually leads to neuronal loss. These features are shared with other neurodegenerative diseases (NDs). Currently, several inhibitors against negative regulators of HSF1, as well as HSF1 activators, are developed and hold promise to prevent neurodegeneration in HD and other NDs. CONCLUSION Understanding the role of HSF1 during protein aggregation and neurodegeneration in HD may help to develop therapeutic strategies that could be effective across different NDs.
Collapse
Affiliation(s)
- Hyuck Kim
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Kovács D, Kovács M, Ahmed S, Barna J. Functional diversification of heat shock factors. Biol Futur 2022; 73:427-439. [PMID: 36402935 DOI: 10.1007/s42977-022-00138-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
Abstract
Heat shock transcription factors (HSFs) are widely known as master regulators of the heat shock response. In invertebrates, a single heat shock factor, HSF1, is responsible for the maintenance of protein homeostasis. In vertebrates, seven members of the HSF family have been identified, namely HSF1, HSF2, HSF3, HSF4, HSF5, HSFX, and HSFY, of which HSF1 and HSF2 are clearly associated with heat shock response, while HSF4 is involved in development. Other members of the family have not yet been studied as extensively. Besides their role in cellular proteostasis, HSFs influence a plethora of biological processes such as aging, development, cell proliferation, and cell differentiation, and they are implicated in several pathologies such as neurodegeneration and cancer. This is achieved by regulating the expression of a great variety of genes including chaperones. Here, we review our current knowledge on the function of HSF family members and important aspects that made possible the functional diversification of HSFs.
Collapse
Affiliation(s)
- Dániel Kovács
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - Márton Kovács
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - Saqib Ahmed
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary
| | - János Barna
- Department of Genetics, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary. .,ELKH-ELTE Genetics Research Group, Pázmány Péter sétány 1/c, Budapest, H-1117, Hungary.
| |
Collapse
|
4
|
Roos-Mattjus P, Sistonen L. Interplay between mammalian heat shock factors 1 and 2 in physiology and pathology. FEBS J 2022; 289:7710-7725. [PMID: 34478606 DOI: 10.1111/febs.16178] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/17/2021] [Accepted: 09/02/2021] [Indexed: 01/14/2023]
Abstract
The heat-shock factors (HSFs) belong to an evolutionary conserved family of transcription factors that were discovered already over 30 years ago. The HSFs have been shown to a have a broad repertoire of target genes, and they also have crucial functions during normal development. Importantly, HSFs have been linked to several disease states, such as neurodegenerative disorders and cancer, highlighting their importance in physiology and pathology. However, it is still unclear how HSFs are regulated and how they choose their specific target genes under different conditions. Posttranslational modifications and interplay among the HSF family members have been shown to be key regulatory mechanisms for these transcription factors. In this review, we focus on the mammalian HSF1 and HSF2, including their interplay, and provide an updated overview of the advances in understanding how HSFs are regulated and how they function in multiple processes of development, aging, and disease. We also discuss HSFs as therapeutic targets, especially the recently reported HSF1 inhibitors.
Collapse
Affiliation(s)
- Pia Roos-Mattjus
- Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Lea Sistonen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| |
Collapse
|
5
|
Sahebnasagh A, Eghbali S, Saghafi F, Sureda A, Avan R. Neurohormetic phytochemicals in the pathogenesis of neurodegenerative diseases. Immun Ageing 2022; 19:36. [PMID: 35953850 PMCID: PMC9367062 DOI: 10.1186/s12979-022-00292-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/24/2022] [Indexed: 12/02/2022]
Abstract
The world population is progressively ageing, assuming an enormous social and health challenge. As the world ages, neurodegenerative diseases are on the rise. Regarding the progressive nature of these diseases, none of the neurodegenerative diseases are curable at date, and the existing treatments can only help relieve the symptoms or slow the progression. Recently, hormesis has increased attention in the treatment of age-related neurodegenerative diseases. The concept of hormesis refers to a biphasic dose-response phenomenon, where low levels of the drug or stress exert protective of beneficial effects and high doses deleterious or toxic effects. Neurohormesis, as the adaptive aspect of hormetic dose responses in neurons, has been shown to slow the onset of neurodegenerative diseases and reduce the damages caused by aging, stroke, and traumatic brain injury. Hormesis was also observed to modulate anxiety, stress, pain, and the severity of seizure. Thus, neurohormesis can be considered as a potentially innovative approach in the treatment of neurodegenerative and other neurologic disorders. Herbal medicinal products and supplements are often considered health resources with many applications. The hormesis phenomenon in medicinal plants is valuable and several studies have shown that hormetic mechanisms of bioactive compounds can prevent or ameliorate the neurodegenerative pathogenesis in animal models of Alzheimer’s and Parkinson’s diseases. Moreover, the hormesis activity of phytochemicals has been evaluated in other neurological disorders such as Autism and Huntington’s disease. In this review, the neurohormetic dose–response concept and the possible underlying neuroprotection mechanisms are discussed. Different neurohormetic phytochemicals used for the better management of neurodegenerative diseases, the rationale for using them, and the key findings of their studies are also reviewed.
Collapse
|
6
|
Liu C, Fu Z, Wu S, Wang X, Zhang S, Chu C, Hong Y, Wu W, Chen S, Jiang Y, Wu Y, Song Y, Liu Y, Guo X. Mitochondrial HSF1 triggers mitochondrial dysfunction and neurodegeneration in Huntington's disease. EMBO Mol Med 2022; 14:e15851. [PMID: 35670111 PMCID: PMC9260212 DOI: 10.15252/emmm.202215851] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 12/18/2022] Open
Affiliation(s)
- Chunyue Liu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Zixing Fu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shanshan Wu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xiaosong Wang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengrong Zhang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Wenbo Wu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengqi Chen
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yueqing Jiang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Key Laboratory of Magnetic Resonance in Biological Systems Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences Wuhan China
| | - Yongbo Song
- Department of Pharmacology Shenyang Pharmaceutical University Shenyang China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- Department of Endocrinology Sir Run Run Hospital Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
7
|
Mickael ME, Bhaumik S, Chakraborti A, Umfress AA, van Groen T, Macaluso M, Totenhagen J, Sorace AG, Bibb JA, Standaert DG, Basu R. RORγt-Expressing Pathogenic CD4 + T Cells Cause Brain Inflammation during Chronic Colitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2054-2066. [PMID: 35379749 PMCID: PMC10103644 DOI: 10.4049/jimmunol.2100869] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/11/2022] [Indexed: 01/09/2023]
Abstract
Neurobehavioral disorders and brain abnormalities have been extensively reported in both Crohn's disease and ulcerative colitis patients. However, the mechanism causing neuropathological disorders in inflammatory bowel disease patients remains unknown. Studies have linked the Th17 subset of CD4+ T cells to brain diseases associated with neuroinflammation and cognitive impairment, including multiple sclerosis, ischemic brain injury, and Alzheimer's disease. To better understand how CD4+ T lymphocytes contribute to brain pathology in chronic intestinal inflammation, we investigated the development of brain inflammation in the T cell transfer model of chronic colitis. Our findings demonstrate that CD4+ T cells infiltrate the brain of colitic Rag1 -/- mice in proportional levels to colitis severity. Colitic mice developed hypothalamic astrogliosis that correlated with neurobehavioral disorders. Moreover, the brain-infiltrating CD4+ T cells expressed Th17 cell transcription factor retinoic acid-related orphan receptor γt (RORγt) and displayed a pathogenic Th17 cellular phenotype similar to colonic Th17 cells. Adoptive transfer of RORγt-deficient naive CD4+ T cells failed to cause brain inflammation and neurobehavioral disorders in Rag1 -/- recipients, with significantly less brain infiltration of CD4+ T cells. The finding is mirrored in chronic dextran sulfate sodium-induced colitis in Rorcfl/fl Cd4-Cre mice that showed lower frequency of brain-infiltrating CD4+ T cells and astrogliosis despite onset of significantly more severe colitis compared with wild-type mice. These findings suggest that pathogenic RORγt+CD4+ T cells that aggravate colitis migrate preferentially into the brain, contributing to brain inflammation and neurobehavioral disorders, thereby linking colitis severity to neuroinflammation.
Collapse
Affiliation(s)
| | - Suniti Bhaumik
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Ayanabha Chakraborti
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Alan A Umfress
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Matthew Macaluso
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | - John Totenhagen
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL.,Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL; and
| | - James A Bibb
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Rajatava Basu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL;
| |
Collapse
|
8
|
Zarate N, Intihar TA, Yu D, Sawyer J, Tsai W, Syed M, Carlson L, Gomez-Pastor R. Heat Shock Factor 1 Directly Regulates Postsynaptic Scaffolding PSD-95 in Aging and Huntington's Disease and Influences Striatal Synaptic Density. Int J Mol Sci 2021; 22:13113. [PMID: 34884918 PMCID: PMC8657899 DOI: 10.3390/ijms222313113] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
PSD-95 (Dlg4) is an ionotropic glutamate receptor scaffolding protein essential in synapse stability and neurotransmission. PSD-95 levels are reduced during aging and in neurodegenerative diseases like Huntington's disease (HD), and it is believed to contribute to synaptic dysfunction and behavioral deficits. However, the mechanism responsible for PSD-95 dysregulation under these conditions is unknown. The Heat Shock transcription Factor 1 (HSF1), canonically known for its role in protein homeostasis, is also depleted in both aging and HD. Synaptic protein levels, including PSD-95, are influenced by alterations in HSF1 levels and activity, but the direct regulatory relationship between PSD-95 and HSF1 has yet to be determined. Here, we showed that HSF1 chronic or acute reduction in cell lines and mice decreased PSD-95 expression. Furthermore, Hsf1(+/-) mice had reduced PSD-95 synaptic puncta that paralleled a loss in thalamo-striatal excitatory synapses, an important circuit disrupted early in HD. We demonstrated that HSF1 binds to regulatory elements present in the PSD-95 gene and directly regulates PSD-95 expression. HSF1 DNA-binding on the PSD-95 gene was disrupted in an age-dependent manner in WT mice and worsened in HD cells and mice, leading to reduced PSD-95 levels. These results demonstrate a direct role of HSF1 in synaptic gene regulation that has important implications in synapse maintenance in basal and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.Z.); (T.A.I.); (D.Y.); (J.S.); (W.T.); (M.S.); (L.C.)
| |
Collapse
|
9
|
Li L, Li Y, He B, Li H, Ji H, Wang Y, Zhu Z, Hu Y, Zhou Y, Yang T, Sun C, Yuan Y, Wang Y. HSF1 is involved in suppressing A1 phenotype conversion of astrocytes following spinal cord injury in rats. J Neuroinflammation 2021; 18:205. [PMID: 34530848 PMCID: PMC8444373 DOI: 10.1186/s12974-021-02271-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
Background Two activation states of reactive astrocytes termed A1 and A2 subtypes emerge at the lesion sites following spinal cord injury (SCI). A1 astrocytes are known to be neurotoxic that participate in neuropathogenesis, whereas A2 astrocytes have been assigned the neuroprotective activity. Heat shock transcription factor 1 (HSF1) plays roles in protecting cells from stress-induced apoptosis and in controlling inflammatory activation. It is unknown whether HSF1 is involved in suppressing the conversion of A1 astrocytes following SCI. Methods A contusion model of the rat spinal cord was established, and the correlations between HSF1 expression and onset of A1 and A2 astrocytes were assayed by Western blot and immunohistochemistry. 17-AAG, the agonist of HSF1, was employed to treat the primary cultured astrocytes following a challenge by an A1-astrocyte-conditioned medium (ACM) containing 3 ng/ml of IL-1α, 30 ng/ml of TNF-α, and 400 ng/ml of C1q for induction of the A1 subtype. The effects of 17-AAG on the phenotype conversion of astrocytes, as well as underlying signal pathways, were examined by Western blot or immunohistochemistry. Results The protein levels of HSF1 were significantly increased at 4 days and 7 days following rat SCI, showing colocalization with astrocytes. Meanwhile, C3-positive A1 astrocytes were observed to accumulate at lesion sites with a peak at 1 day and 4 days. Distinctively, the S100A10-positive A2 subtype reached its peak at 4 days and 7 days. Incubation of the primary astrocytes with ACM markedly induced the conversion of the A1 phenotype, whereas an addition of 17-AAG significantly suppressed such inducible effects without conversion of the A2 subtype. Activation of HSF1 remarkably inhibited the activities of MAPKs and NFκB, which was responsible for the regulation of C3 expression. Administration of 17-AAG at the lesion sites of rats was able to reduce the accumulation of A1 astrocytes. Conclusion Collectively, these data reveal a novel mechanism of astrocyte phenotype conversion following SCI, and HSF1 plays key roles in suppressing excessive increase of neurotoxic A1 astrocytes. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02271-3.
Collapse
Affiliation(s)
- Lilan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Hui Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Huiyuan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.,Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Zhenjie Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yuming Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yue Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Chunshuai Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Ying Yuan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
10
|
Occhigrossi L, D’Eletto M, Barlev N, Rossin F. The Multifaceted Role of HSF1 in Pathophysiology: Focus on Its Interplay with TG2. Int J Mol Sci 2021; 22:ijms22126366. [PMID: 34198675 PMCID: PMC8232231 DOI: 10.3390/ijms22126366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022] Open
Abstract
The cellular environment needs to be strongly regulated and the maintenance of protein homeostasis is crucial for cell function and survival. HSF1 is the main regulator of the heat shock response (HSR), the master pathway required to maintain proteostasis, as involved in the expression of the heat shock proteins (HSPs). HSF1 plays numerous physiological functions; however, the main role concerns the modulation of HSPs synthesis in response to stress. Alterations in HSF1 function impact protein homeostasis and are strongly linked to diseases, such as neurodegenerative disorders, metabolic diseases, and different types of cancers. In this context, type 2 Transglutaminase (TG2), a ubiquitous enzyme activated during stress condition has been shown to promote HSF1 activation. HSF1-TG2 axis regulates the HSR and its function is evolutionary conserved and implicated in pathological conditions. In this review, we discuss the role of HSF1 in the maintenance of proteostasis with regard to the HSF1-TG2 axis and we dissect the stress response pathways implicated in physiological and pathological conditions.
Collapse
Affiliation(s)
- Luca Occhigrossi
- Department of Biology, University of Rome ‘Tor Vergata’, 00133 Rome, Italy; (L.O.); (M.D.)
| | - Manuela D’Eletto
- Department of Biology, University of Rome ‘Tor Vergata’, 00133 Rome, Italy; (L.O.); (M.D.)
| | - Nickolai Barlev
- Institute of Cytology, 194064 Saint-Petersburg, Russia;
- Moscow Institute of Physics and Technology (MIPT), 141701 Dolgoprudny, Russia
| | - Federica Rossin
- Institute of Cytology, 194064 Saint-Petersburg, Russia;
- Correspondence:
| |
Collapse
|
11
|
Syafruddin SE, Ling S, Low TY, Mohtar MA. More Than Meets the Eye: Revisiting the Roles of Heat Shock Factor 4 in Health and Diseases. Biomolecules 2021; 11:523. [PMID: 33807297 PMCID: PMC8066111 DOI: 10.3390/biom11040523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
Cells encounter a myriad of endogenous and exogenous stresses that could perturb cellular physiological processes. Therefore, cells are equipped with several adaptive and stress-response machinery to overcome and survive these insults. One such machinery is the heat shock response (HSR) program that is governed by the heat shock factors (HSFs) family in response towards elevated temperature, free radicals, oxidants, and heavy metals. HSF4 is a member of this HSFs family that could exist in two predominant isoforms, either the transcriptional repressor HSFa or transcriptional activator HSF4b. HSF4 is constitutively active due to the lack of oligomerization negative regulator domain. HSF4 has been demonstrated to play roles in several physiological processes and not only limited to regulating the classical heat shock- or stress-responsive transcriptional programs. In this review, we will revisit and delineate the recent updates on HSF4 molecular properties. We also comprehensively discuss the roles of HSF4 in health and diseases, particularly in lens cell development, cataract formation, and cancer pathogenesis. Finally, we will posit the potential direction of HSF4 future research that could enhance our knowledge on HSF4 molecular networks as well as physiological and pathophysiological functions.
Collapse
|
12
|
Lee MJ, Jang Y, Han J, Kim SJ, Ju X, Lee YL, Cui J, Zhu J, Ryu MJ, Choi SY, Chung W, Heo C, Yi HS, Kim HJ, Huh YH, Chung SK, Shong M, Kweon GR, Heo JY. Endothelial-specific Crif1 deletion induces BBB maturation and disruption via the alteration of actin dynamics by impaired mitochondrial respiration. J Cereb Blood Flow Metab 2020; 40:1546-1561. [PMID: 31987007 PMCID: PMC7308523 DOI: 10.1177/0271678x19900030] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebral endothelial cells (ECs) require junctional proteins to maintain blood-brain barrier (BBB) integrity, restricting toxic substances and controlling peripheral immune cells with a higher concentration of mitochondria than ECs of peripheral capillaries. The mechanism underlying BBB disruption by defective mitochondrial oxidative phosphorylation (OxPhos) is unclear in a mitochondria-related gene-targeted animal model. To assess the role of EC mitochondrial OxPhos function in the maintenance of the BBB, we developed an EC-specific CR6-interactin factor1 (Crif1) deletion mouse. We clearly observed defects in motor behavior, uncompacted myelin and leukocyte infiltration caused by BBB maturation and disruption in this mice. Furthermore, we investigated the alteration in the actin cytoskeleton, which interacts with junctional proteins to support BBB integrity. Loss of Crif1 led to reorganization of the actin cytoskeleton and a decrease in tight junction-associated protein expression through an ATP production defect in vitro and in vivo. Based on these results, we suggest that mitochondrial OxPhos is important for the maturation and maintenance of BBB integrity by supplying ATP to cerebral ECs.
Collapse
Affiliation(s)
- Min Joung Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yunseon Jang
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jeongsu Han
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Soo J Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Xianshu Ju
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yu Lim Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jianchen Cui
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jiebo Zhu
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Song-Yi Choi
- Department of Pathology, Chungnam National University, Daejeon, Republic of Korea
| | - Woosuk Chung
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Anesthesiology and Pain Medicine, School of Medicine, Chungnam National University, Daejeon, Republic of Korea.,Department of Anesthesiology and Pain medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Chaejeong Heo
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, South Korea.,Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, South Korea
| | - Hyon-Seung Yi
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Yang H Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, Chungcheongbukdo, Republic of Korea
| | - Sookja K Chung
- Medical Faculty at Macau University of Science and Technology, Taipa, Macau
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea.,Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Gi-Ryang Kweon
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Jun Young Heo
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea.,Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
13
|
Trivedi R, Jurivich DA. A molecular perspective on age-dependent changes to the heat shock axis. Exp Gerontol 2020; 137:110969. [PMID: 32407864 DOI: 10.1016/j.exger.2020.110969] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/13/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022]
Abstract
Aging is a complex process associated with progressive damage that leads to cellular dysfunction often accompanied by frailty and age-related diseases. Coping with all types of physiologic stress declines with age. While representing a primordial, cross-species response in poikilo- and homeotherms, the age-dependent perturbation of the stress response is more complex than previously thought. This short review examines how age influences the stress axis at multiple levels that involve both activating and attenuating pathways.
Collapse
Affiliation(s)
- Rachana Trivedi
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, USA.
| | - Donald A Jurivich
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, USA.
| |
Collapse
|
14
|
Duchateau A, de Thonel A, El Fatimy R, Dubreuil V, Mezger V. The "HSF connection": Pleiotropic regulation and activities of Heat Shock Factors shape pathophysiological brain development. Neurosci Lett 2020; 725:134895. [PMID: 32147500 DOI: 10.1016/j.neulet.2020.134895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/29/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022]
Abstract
The Heat Shock Factors (HSFs) have been historically identified as a family of transcription factors that are activated and work in a stress-responsive manner, after exposure to a large variety of stimuli. However, they are also critical in normal conditions, in a life long manner, in a number of physiological processes that encompass gametogenesis, embryonic development and the integrity of adult organs and organisms. The importance of such roles is emphasized by the devastating impact of their deregulation on health, ranging from reproductive failure, neurodevelopmental disorders, cancer, and aging pathologies, including neurodegenerative disorders. Here, we provide an overview of the delicate choreography of the regulation of HSFs during neurodevelopment, at prenatal and postnatal stages. The regulation of HSFs acts at multiple layers and steps, and comprises the control of (i) HSF mRNA and protein levels, (ii) HSF activity in terms of DNA-binding and transcription, (iii) HSF homo- and hetero-oligomerization capacities, and (iv) HSF combinatory set of post-translational modifications. We also describe how these regulatory mechanisms operate in the normal developing brain and how their perturbation impact neurodevelopment under prenatal or perinatal stress conditions. In addition, we put into perspective the possible role of HSFs in the evolution of the vertebrate brains and the importance of the HSF pathway in a large variety of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Agathe Duchateau
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France; ED 562 BioSPC, Université de Paris, F-75205, Paris Cedex 13, France
| | - Aurélie de Thonel
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Rachid El Fatimy
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Véronique Dubreuil
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Valérie Mezger
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France.
| |
Collapse
|
15
|
Nanocatalytic activity of clean-surfaced, faceted nanocrystalline gold enhances remyelination in animal models of multiple sclerosis. Sci Rep 2020; 10:1936. [PMID: 32041968 PMCID: PMC7010780 DOI: 10.1038/s41598-020-58709-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/26/2022] Open
Abstract
Development of pharmacotherapies that promote remyelination is a high priority for multiple sclerosis (MS), due to their potential for neuroprotection and restoration of function through repair of demyelinated lesions. A novel preparation of clean-surfaced, faceted gold nanocrystals demonstrated robust remyelinating activity in response to demyelinating agents in both chronic cuprizone and acute lysolecithin rodent animal models. Furthermore, oral delivery of gold nanocrystals improved motor functions of cuprizone-treated mice in both open field and kinematic gait studies. Gold nanocrystal treatment of oligodendrocyte precursor cells in culture resulted in oligodendrocyte maturation and expression of myelin differentiation markers. Additional in vitro data demonstrated that these gold nanocrystals act via a novel energy metabolism pathway involving the enhancement of key indicators of aerobic glycolysis. In response to gold nanocrystals, co-cultured central nervous system cells exhibited elevated levels of the redox coenzyme nicotine adenine dinucleotide (NAD+), elevated total intracellular ATP levels, and elevated extracellular lactate levels, along with upregulation of myelin-synthesis related genes, collectively resulting in functional myelin generation. Based on these preclinical studies, clean-surfaced, faceted gold nanocrystals represent a novel remyelinating therapeutic for multiple sclerosis.
Collapse
|
16
|
Gao J, Liu J, Zhang L, Zhang Y, Guo Q, Li Y, Tong J, Wang H, Zhou J, Zhu F, Shi L, Zhao H. Heat shock transcription factor 1 regulates the fetal γ-globin expression in a stress-dependent and independent manner during erythroid differentiation. Exp Cell Res 2019; 387:111780. [PMID: 31874177 DOI: 10.1016/j.yexcr.2019.111780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 01/09/2023]
Abstract
Heat shock transcription factor 1 (HSF1) is a highly versatile transcription factor that, in addition to protecting cells against proteotoxic stress, is also critical during diverse developmental processes. Although the functions of HSF1 have received considerable attention, its potential role in β-globin gene regulation during erythropoiesis has not been fully elucidated. Here, after comparing the transcriptomes of erythrocytes differentiated from cord blood or adult peripheral blood hematopoietic progenitor CD34+ cells in vitro, we constructed the molecular regulatory network associated with β-globin genes and identified novel and putative globin gene regulators by combining the weighted gene coexpression network analysis (WGCNA) and context likelihood of relatedness (CLR) algorithms. Further investigation revealed that one of the identified regulators, HSF1, acts as a key activator of the γ-globin gene in human primary erythroid cells in both erythroid developmental stages. While during stress, HSF1 is required for heat-induced globin gene activation, and HSF1 downregulation markedly decreases globin gene induction in K562 cells. Mechanistically, HSF1 occupies DNase I hypersensitive site 3 of the locus control region upstream of β-globin genes via its canonical binding motif. Hence, HSF1 executes stress-dependent and -independent roles in fetal γ-globin regulation during erythroid differentiation.
Collapse
Affiliation(s)
- Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jinhua Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lingling Zhang
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China
| | - Yingnan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qing Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yapu Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongtao Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fan Zhu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China.
| |
Collapse
|
17
|
Joutsen J, Sistonen L. Tailoring of Proteostasis Networks with Heat Shock Factors. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034066. [PMID: 30420555 DOI: 10.1101/cshperspect.a034066] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heat shock factors (HSFs) are the main transcriptional regulators of the heat shock response and indispensable for maintaining cellular proteostasis. HSFs mediate their protective functions through diverse genetic programs, which are composed of genes encoding molecular chaperones and other genes crucial for cell survival. The mechanisms that are used to tailor HSF-driven proteostasis networks are not yet completely understood, but they likely comprise from distinct combinations of both genetic and proteomic determinants. In this review, we highlight the versatile HSF-mediated cellular functions that extend from cellular stress responses to various physiological and pathological processes, and we underline the key advancements that have been achieved in the field of HSF research during the last decade.
Collapse
Affiliation(s)
- Jenny Joutsen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| |
Collapse
|
18
|
Intihar TA, Martinez EA, Gomez-Pastor R. Mitochondrial Dysfunction in Huntington's Disease; Interplay Between HSF1, p53 and PGC-1α Transcription Factors. Front Cell Neurosci 2019; 13:103. [PMID: 30941017 PMCID: PMC6433789 DOI: 10.3389/fncel.2019.00103] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by an expanded CAG repeat in the huntingtin (HTT) gene, causing the protein to misfold and aggregate. HD progression is characterized by motor impairment and cognitive decline associated with the preferential loss of striatal medium spiny neurons (MSNs). The mechanisms that determine increased susceptibility of MSNs to mutant HTT (mHTT) are not fully understood, although there is abundant evidence demonstrating the importance of mHTT mediated mitochondrial dysfunction in MSNs death. Two main transcription factors, p53 and peroxisome proliferator co-activator PGC-1α, have been widely studied in HD for their roles in regulating mitochondrial function and apoptosis. The action of these two proteins seems to be interconnected. However, it is still open to discussion whether p53 and PGC-1α dependent responses directly influence each other or if they are connected via a third mechanism. Recently, the stress responsive transcription factor HSF1, known for its role in protein homeostasis, has been implicated in mitochondrial function and in the regulation of PGC-1α and p53 levels in different contexts. Based on previous reports and our own research, we discuss in this review the potential role of HSF1 in mediating mitochondrial dysfunction in HD and propose a unifying mechanism that integrates the responses mediated by p53 and PGC-1α in HD via HSF1.
Collapse
Affiliation(s)
- Taylor A. Intihar
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Elisa A. Martinez
- Department of Biochemistry and Molecular Biology, Dickinson College, Carlisle, PA, United States
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
19
|
Abstract
Heat shock transcription factors (Hsfs) regulate transcription of heat shock proteins as well as other genes whose promoters contain heat shock elements (HSEs). There are at least five Hsfs in mammalian cells, Hsf1, Hsf2, Hsf3, Hsf4, and Hsfy (Wu, Annu Rev Cell Dev Biol 11:441-469, 1995; Morimoto, Genes Dev 12:3788-3796, 1998; Tessari et al., Mol Hum Repord 4:253-258, 2004; Fujimoto et al., Mol Biol Cell 21:106-116, 2010; Nakai et al., Mol Cell Biol 17:469-481, 1997; Sarge et al., Genes Dev 5:1902-1911, 1991). To understand the physiological roles of Hsf1, Hsf2, and Hsf4 in vivo, we generated knockout mouse lines for these factors (Zhang et al., J Cell Biochem 86:376-393, 2002; Wang et al., Genesis 36:48-61, 2003; Min et al., Genesis 40:205-217, 2004). Numbers of other laboratories have also generated Hsf1 (Xiao et al., EMBO J 18:5943-5952, 1999; Sugahara et al., Hear Res 182:88-96, 2003), Hsf2 (McMillan et al., Mol Cell Biol 22:8005-8014, 2002; Kallio et al., EMBO J 21:2591-2601, 2002), and Hsf4 (Fujimoto et al., EMBO J 23:4297-4306, 2004) knockout mouse models. In this chapter, we describe the design of the targeting vectors, the plasmids used, and the successful generation of mice lacking the individual genes. We also briefly describe what we have learned about the physiological functions of these genes in vivo.
Collapse
Affiliation(s)
- Xiongjie Jin
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Georgia Cancer Center, 1410 Laney Walker Blvd., CN3141, Augusta, GA, 30912, USA
| | - Binnur Eroglu
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Georgia Cancer Center, 1410 Laney Walker Blvd., CN3141, Augusta, GA, 30912, USA
| | - Demetrius Moskophidis
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Georgia Cancer Center, 1410 Laney Walker Blvd., CN3141, Augusta, GA, 30912, USA
| | - Nahid F Mivechi
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Georgia Cancer Center, 1410 Laney Walker Blvd., CN3141, Augusta, GA, 30912, USA.
| |
Collapse
|
20
|
Mehta RI, Tsymbalyuk N, Ivanova S, Stokum JA, Woo K, Gerzanich V, Simard JM. α-Endosulfine (ARPP-19e) Expression in a Rat Model of Stroke. J Neuropathol Exp Neurol 2017; 76:898-907. [PMID: 28922851 DOI: 10.1093/jnen/nlx074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In nutrient restricted environments, the yeast endosulfines Igo1/2 are activated via TORC1 inhibition and function critically to initiate and coordinate the cellular stress response that promotes survival. We examined expression of αEnsa, the mammalian homolog of yeast endosulfines, in rat stroke. Prominent neuronal upregulation of αEnsa was identified in 3 patterns within the ischemic gradient: (1) neurons in GFAP-/HSF1+ cortex showed upregulation and near-complete nuclear translocation of αEnsa protein within hours of ischemic onset; (2) neurons in GFAP+/HSF1+ cortex showed upregulation in cytoplasm and nuclei that persisted for days; (3) neurons in GFAP+/HSF1- cortex showed delayed cytosolic-only upregulation that persisted for days. Findings were corroborated using in situ hybridization for ENSA mRNA. Rapamycin treatment was found to reduce infarct size and behavioral deficits and, in GFAP+/HSF1+ zones, enhance αEnsa neuronal nuclear translocation and mitigate cell death, relative to controls. Based on the conservation of TOR signaling across species, and on the finding that the Rim15-Igo1/2-PP2A module is triggered by substrate deprivation in eukaryotic yeast, we speculate that αEnsa is activated by substrate deprivation, functioning through the homologous MASTL-αEnsa/ARPP19-PP2A module to promote neuronal survival. In conjunction with recent studies suggesting a neuroprotective role, our data highlight a potential function for αEnsa within ischemic brain.
Collapse
Affiliation(s)
- Rupal I Mehta
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Natalia Tsymbalyuk
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Svetlana Ivanova
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jesse A Stokum
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kyoon Woo
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Volodymyr Gerzanich
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - J M Simard
- Department of Pathology and Laboratory Medicine; Center for Neurotherapeutics Discovery, Department of Neuroscience; Center for Translational Neuromedicine, University of Rochester, Rochester, New York; Department of Pathology; Department of Neurosurgery; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Gomez-Pastor R, Burchfiel ET, Thiele DJ. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat Rev Mol Cell Biol 2017; 19:4-19. [PMID: 28852220 DOI: 10.1038/nrm.2017.73] [Citation(s) in RCA: 540] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The heat shock transcription factors (HSFs) were discovered over 30 years ago as direct transcriptional activators of genes regulated by thermal stress, encoding heat shock proteins. The accepted paradigm posited that HSFs exclusively activate the expression of protein chaperones in response to conditions that cause protein misfolding by recognizing a simple promoter binding site referred to as a heat shock element. However, we now realize that the mammalian family of HSFs comprises proteins that independently or in concert drive combinatorial gene regulation events that activate or repress transcription in different contexts. Advances in our understanding of HSF structure, post-translational modifications and the breadth of HSF-regulated target genes have revealed exciting new mechanisms that modulate HSFs and shed new light on their roles in physiology and pathology. For example, the ability of HSF1 to protect cells from proteotoxicity and cell death is impaired in neurodegenerative diseases but can be exploited by cancer cells to support their growth, survival and metastasis. These new insights into HSF structure, function and regulation should facilitate the development tof new disease therapeutics to manipulate this transcription factor family.
Collapse
Affiliation(s)
- Rocio Gomez-Pastor
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine
| | | | - Dennis J Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine.,Department of Biochemistry, Duke University School of Medicine.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
22
|
Dayalan Naidu S, Dinkova-Kostova AT. Regulation of the mammalian heat shock factor 1. FEBS J 2017; 284:1606-1627. [PMID: 28052564 DOI: 10.1111/febs.13999] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/17/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Abstract
Living organisms are endowed with the capability to tackle various forms of cellular stress due to the presence of molecular chaperone machinery complexes that are ubiquitous throughout the cell. During conditions of proteotoxic stress, the transcription factor heat shock factor 1 (HSF1) mediates the elevation of heat shock proteins, which are crucial components of the chaperone complex machinery and function to ameliorate protein misfolding and aggregation and restore protein homeostasis. In addition, HSF1 orchestrates a versatile transcriptional programme that includes genes involved in repair and clearance of damaged macromolecules and maintenance of cell structure and metabolism, and provides protection against a broad range of cellular stress mediators, beyond heat shock. Here, we discuss the structure and function of the mammalian HSF1 and its regulation by post-translational modifications (phosphorylation, sumoylation and acetylation), proteasomal degradation, and small-molecule activators and inhibitors.
Collapse
Affiliation(s)
- Sharadha Dayalan Naidu
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, UK
| | - Albena T Dinkova-Kostova
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, UK
- Department of Pharmacology and Molecular Sciences, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Ingenwerth M, Estrada V, Stahr A, Müller HW, von Gall C. HSF1-deficiency affects gait coordination and cerebellar calbindin levels. Behav Brain Res 2016; 310:103-8. [DOI: 10.1016/j.bbr.2016.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/02/2016] [Accepted: 05/06/2016] [Indexed: 12/27/2022]
|
24
|
The Activators of Cyclin-Dependent Kinase 5 p35 and p39 Are Essential for Oligodendrocyte Maturation, Process Formation, and Myelination. J Neurosci 2016; 36:3024-37. [PMID: 26961956 DOI: 10.1523/jneurosci.2250-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The regulation of oligodendrocyte development and myelin formation in the CNS is poorly defined. Multiple signals influence the rate and extent of CNS myelination, including the noncanonical cyclin-dependent kinase 5 (Cdk5) whose functions are regulated by its activators p35 and p39. Here we show that selective loss of either p35 or p39 perturbed specific aspects of oligodendrocyte development, whereas loss of both p35 and p39 completely inhibited the development of mature oligodendrocytes and myelination. In the absence of p35, oligodendrocyte differentiation was delayed, process outgrowth was truncated in vitro, and the patterning and extent of myelination were perturbed in the CNS of p35(-/-) mice. In the absence of p39, oligodendrocyte maturation was transiently affected both in vitro and in vivo. However, loss of both p35 and p39 in oligodendrocyte lineage cells completely inhibited oligodendrocyte progenitor cell differentiation and myelination both in vitro and after transplantation into shiverer slice cultures. Loss of p35 and p39 had a more profound effect on oligodendrocyte development than simply the loss of Cdk5 and could not be rescued by Cdk5 overexpression. These data suggest p35 and p39 have specific and overlapping roles in oligodendrocyte development, some of which may be independent of Cdk5 activation.
Collapse
|
25
|
Ingenwerth M, Noichl E, Stahr A, Korf HW, Reinke H, von Gall C. Heat Shock Factor 1 Deficiency Affects Systemic Body Temperature Regulation. Neuroendocrinology 2016; 103:605-15. [PMID: 26513256 DOI: 10.1159/000441947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/22/2015] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Heat shock factor 1 (HSF1) is a ubiquitous heat-sensitive transcription factor that mediates heat shock protein transcription in response to cellular stress, such as increased temperature, in order to protect the organism against misfolded proteins. In this study, we analysed the effect of HSF1 deficiency on core body temperature regulation. MATERIALS AND METHODS Body temperature, locomotor activity, and food consumption of wild-type mice and HSF1-deficient mice were recorded. Prolactin and thyroid-stimulating hormone levels were measured by ELISA. Gene expression in brown adipose tissue was analysed by quantitative real-time PCR. Hypothalamic HSF1 and its co-localisation with tyrosine hydroxylase was analysed using confocal laser scanning microscopy. RESULTS HSF1-deficient mice showed an increase in core body temperature (hyperthermia), decreased overall locomotor activity, and decreased levels of prolactin in pituitary and blood plasma reminiscent of cold adaptation. HSF1 could be detected in various hypothalamic regions involved in temperature regulation, suggesting a potential role of HSF1 in hypothalamic thermoregulation. Moreover, HSF1 co-localises with tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis, suggesting a potential role of HSF1 in the hypothalamic control of prolactin release. In brown adipose tissue, levels of prolactin receptor and uncoupled protein 1 were increased in HSF1-deficient mice, consistent with an up-regulation of heat production. CONCLUSION Our data suggest a role of HSF1 in systemic thermoregulation.
Collapse
|
26
|
Analysis of the heat shock factor complex in mammalian HSP70 promoter. Methods Mol Biol 2015; 1292:53-65. [PMID: 25804747 DOI: 10.1007/978-1-4939-2522-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The heat shock response is characterized by the induction of heat shock proteins (HSPs) and is one of prominent mechanisms that regulate proteostasis capacity in the cell. In mammals, heat shock factor 1 (HSF1) regulates the expression of HSPs transcriptionally in both unstressed and stressed cells. Recent reports show that the HSF1-RPA complex constitutively gains access to nucleosomal DNA in part by recruiting a histone chaperone and a chromatin-remodeling component. Here, we describe the strategies to substitute endogenous HSF1 with ectopically expressed HSF1 or its mutant and to detect the occupancy of HSF1 transcription complex including RPA in vivo on two heat shock response elements located close together in the human or mouse HSP70 promoters by chromatin immunoprecipitation assay with high sensitivity and specificity.
Collapse
|
27
|
Liu DJ, Hammer D, Komlos D, Chen KY, Firestein BL, Liu AYC. SIRT1 knockdown promotes neural differentiation and attenuates the heat shock response. J Cell Physiol 2014; 229:1224-35. [PMID: 24435709 DOI: 10.1002/jcp.24556] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 01/14/2014] [Indexed: 02/06/2023]
Abstract
Neurons have a limited capacity for heat shock protein (HSP) induction and are vulnerable to the pathogenic consequence of protein misfolding and aggregation as seen in age-related neurodegenerative diseases. Sirtuin 1 (SIRT1), an NAD(+) -dependent lysine deacetylase with important biological functions, has been shown to sustain the DNA-binding state of HSF1 for HSP induction. Here we show that differentiation and maturation of embryonic cortical neurons and N2a neuroprogenitor cells is associated with decreases in SIRT1 expression and heat shock-dependent induction of HSP70 protein. Tests of a pharmacological activator and an inhibitor of SIRT1 affirm the regulatory role of SIRT1 in HSP70 induction. Protein cross-linking studies show that nuclear SIRT1 and HSF1 form a co-migrating high molecular weight complex upon stress. The use of retroviral vectors to manipulate SIRT1 expression in N2a cells show that shRNA-mediated knock down of SIRT1 causes spontaneous neurite outgrowth coincident with reduced growth rate and decreased induction of hsp70-reporter gene, whereas SIRT1 over-expression blocks the induced neural differentiation of N2a cells. Our results suggest that decreased SIRT1 expression is conducive to neuronal differentiation and this decrease contributes to the attenuated induction of HSPs in neurons.
Collapse
Affiliation(s)
- Diana J Liu
- Department of Cell Biology and Neuroscience, Rutgers State University of New Jersey, Piscataway, New Jersey
| | | | | | | | | | | |
Collapse
|
28
|
The systemic amyloid precursor transthyretin (TTR) behaves as a neuronal stress protein regulated by HSF1 in SH-SY5Y human neuroblastoma cells and APP23 Alzheimer's disease model mice. J Neurosci 2014; 34:7253-65. [PMID: 24849358 DOI: 10.1523/jneurosci.4936-13.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Increased neuronal synthesis of transthyretin (TTR) may favorably impact on Alzheimer's disease (AD) because TTR has been shown to inhibit Aβ aggregation and detoxify cell-damaging conformers. The mechanism whereby hippocampal and cortical neurons from AD patients and APP23 AD model mice produce more TTR is unknown. We now show that TTR expression in SH-SY5Y human neuroblastoma cells, primary hippocampal neurons and the hippocampus of APP23 mice, is significantly enhanced by heat shock factor 1 (HSF1). Chromatin immunoprecipitation (ChIP) assays demonstrated occupation of TTR promoter heat shock elements by HSF1 in APP23 hippocampi, primary murine hippocampal neurons, and SH-SY5Y cells, but not in mouse liver, cultured human hepatoma (HepG2) cells, or AC16 cultured human cardiomyocytes. Treating SH-SY5Y human neuroblastoma cells with heat shock or the HSF1 stimulator celastrol increased TTR transcription in parallel with that of HSP40, HSP70, and HSP90. With both treatments, ChIP showed increased occupancy of heat shock elements in the TTR promoter by HSF1. In vivo celastrol increased the HSF1 ChIP signal in hippocampus but not in liver. Transfection of a human HSF1 construct into SH-SY5Y cells increased TTR transcription and protein production, which could be blocked by shHSF1 antisense. The effect is neuron specific. In cultured HepG2 cells, HSF1 was either suppressive or had no effect on TTR expression confirming the differential effects of HSF1 on TTR transcription in different cell types.
Collapse
|
29
|
Eroglu B, Kimbler DE, Pang J, Choi J, Moskophidis D, Yanasak N, Dhandapani KM, Mivechi NF. Therapeutic inducers of the HSP70/HSP110 protect mice against traumatic brain injury. J Neurochem 2014; 130:626-41. [PMID: 24903326 DOI: 10.1111/jnc.12781] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/09/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022]
Abstract
Traumatic brain injury (TBI) induces severe harm and disability in many accident victims and combat-related activities. The heat-shock proteins Hsp70/Hsp110 protect cells against death and ischemic damage. In this study, we used mice deficient in Hsp110 or Hsp70 to examine their potential requirement following TBI. Data indicate that loss of Hsp110 or Hsp70 increases brain injury and death of neurons. One of the mechanisms underlying the increased cell death observed in the absence of Hsp110 and Hsp70 following TBI is the increased expression of reactive oxygen species-induced p53 target genes Pig1, Pig8, and Pig12. To examine whether drugs that increase the levels of Hsp70/Hsp110 can protect cells against TBI, we subjected mice to TBI and administered Celastrol or BGP-15. In contrast to Hsp110- or Hsp70i-deficient mice that were not protected following TBI and Celastrol treatment, there was a significant improvement of wild-type mice following administration of these drugs during the first week following TBI. In addition, assessment of neurological injury shows significant improvement in contextual and cued fear conditioning tests and beam balance in wild-type mice that were treated with Celastrol or BGP-15 following TBI compared to TBI-treated mice. These studies indicate a significant role of Hsp70/Hsp110 in neuronal survival following TBI and the beneficial effects of Hsp70/Hsp110 inducers toward reducing the pathological consequences of TBI. Our data indicate that loss of Hsp110 or Hsp70 in mice increases brain injury following TBI. (a) One of the mechanisms underlying the increased cell death observed in the absence of these Hsps following TBI is the increased expression of ROS-induced p53 target genes known as Pigs. In addition, (b) using drugs (Celastrol or BGP-15) to increase Hsp70/Hsp110 levels protect cells against TBI, suggesting the beneficial effects of Hsp70/Hsp110 inducers to reduce the pathological consequences of TBI.
Collapse
Affiliation(s)
- Binnur Eroglu
- Charlie Norwood VA Medical Center (CNVAMC), Augusta, Georgia, USA; Molecular Chaperone Biology, Georgia Regents University, Augusta, Georgia, USA; Cancer Center, Georgia Regents University, Augusta, Georgia, USA; Georgia Regents University (GRU) and Medical College of Georgia, Augusta, Georgia, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Roles of heat shock factor 1 in neuronal response to fetal environmental risks and its relevance to brain disorders. Neuron 2014; 82:560-72. [PMID: 24726381 DOI: 10.1016/j.neuron.2014.03.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 12/25/2022]
Abstract
Prenatal exposure of the developing brain to various environmental challenges increases susceptibility to late onset of neuropsychiatric dysfunction; still, the underlying mechanisms remain obscure. Here we show that exposure of embryos to a variety of environmental factors such as alcohol, methylmercury, and maternal seizure activates HSF1 in cerebral cortical cells. Furthermore, Hsf1 deficiency in the mouse cortex exposed in utero to subthreshold levels of these challenges causes structural abnormalities and increases seizure susceptibility after birth. In addition, we found that human neural progenitor cells differentiated from induced pluripotent stem cells derived from schizophrenia patients show higher variability in the levels of HSF1 activation induced by environmental challenges compared to controls. We propose that HSF1 plays a crucial role in the response of brain cells to prenatal environmental insults and may be a key component in the pathogenesis of late-onset neuropsychiatric disorders.
Collapse
|
31
|
Abstract
Heat shock factor 1 (HSF1) protects neurons from death caused by the accumulation of misfolded proteins. It is believed that this protective effect is mediated by the transcriptional stimulation of genes encoding heat shock proteins (HSPs), a family of chaperones that refold or degrade misfolded proteins. Whether HSF1 is protective when neuronal death is not caused by protein misfolding has not been studied. Here, we report that HSF1 expression is necessary for the survival of rat neurons and that HSF1 mRNA and protein expression is reduced in neurons primed to die. Knock-down of HSF1 induces death of otherwise healthy neurons, whereas reestablishment of elevated levels of HSF1 protects neurons even when death is not due to accumulation of misfolded proteins. Neuroprotection by HSF1 does not require its trimerization, an event obligatory for the binding of HSF1 to heat shock elements within HSP gene promoters. Moreover, knock-down of HSP70 or blockade of HSP90 signaling does not reduce neuroprotection by HSF1. Although several neuroprotective molecules and signaling pathways, including CaMK, PKA, Casein kinase-II, and the Raf-MEK-ERK and PI-3K-Akt pathways, are not required for HSF1-mediated neuroprotection, protection is abrogated by inhibition of classical histone deacetylases (HDACs). We report that the novel mechanism of neuroprotection by HSF1 involves cooperation with SIRT1, an HDAC with well documented neuroprotective effects. Using a cell culture model of Huntington's disease, we show that HSF1 trimerization is not required for protection against mutant huntingtin-induced neurotoxicity, suggesting that HSF1 can protect neurons against both proteinopathic and nonproteinopathic death through a noncanonical pathway.
Collapse
|
32
|
Maheshwari M, Bhutani S, Das A, Mukherjee R, Sharma A, Kino Y, Nukina N, Jana NR. Dexamethasone induces heat shock response and slows down disease progression in mouse and fly models of Huntington's disease. Hum Mol Genet 2013; 23:2737-51. [PMID: 24381308 DOI: 10.1093/hmg/ddt667] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by abnormal expansion of glutamine repeats in the protein huntingtin. In HD brain, mutant huntingtin undergoes proteolytic processing, and its N-terminal fragment containing poly-glutamine repeats accumulate as insoluble aggregates leading to the defect in cellular protein quality control system and heat shock response (HSR). Here we demonstrate that the defective HSR in the brain is due to the down-regulation of heat shock factor 1 (HSF1) in both mice and fly models of HD. Interestingly, treatment of dexamethasone (a synthetic glucocorticoid) to HD mice or flies significantly increased the expression and transactivation of HSF1 and induction of HSR and these effects are mediated through the down-regulation of HSP90. Dexamethasone treatment also significantly decreased the aggregate load and transient recovery of HD-related behavioural phenotypes in both disease models. These results suggest that dexamethasone could be a potential therapeutic molecule for the treatment of HD and related poly-glutamine disorders.
Collapse
Affiliation(s)
- Megha Maheshwari
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122 050, India
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Eroglu B, Min JN, Zhang Y, Szurek E, Moskophidis D, Eroglu A, Mivechi NF. An essential role for heat shock transcription factor binding protein 1 (HSBP1) during early embryonic development. Dev Biol 2013; 386:448-60. [PMID: 24380799 DOI: 10.1016/j.ydbio.2013.12.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 12/23/2013] [Accepted: 12/24/2013] [Indexed: 10/25/2022]
Abstract
Heat shock factor binding protein 1 (HSBP1) is a 76 amino acid polypeptide that contains two arrays of hydrophobic heptad repeats and was originally identified through its interaction with the oligomerization domain of heat shock factor 1 (Hsf1), suppressing Hsf1's transcriptional activity following stress. To examine the function of HSBP1 in vivo, we generated mice with targeted disruption of the hsbp1 gene and examined zebrafish embryos treated with HSBP1-specific morpholino oligonucleotides. Our results show that hsbp1 is critical for preimplantation embryonic development. Embryonic stem (ES) cells deficient in hsbp1 survive and proliferate normally into the neural lineage in vitro; however, lack of hsbp1 in embryoid bodies (EBs) leads to disorganization of the germ layers and a reduction in the endoderm-specific markers (such as α-fetoprotein). We further show that hsbp1-deficient mouse EBs and knockdown of HSBP1 in zebrafish leads to an increase in the expression of the neural crest inducers Snail2, Tfap2α and Foxd3, suggesting a potential role for HSBP1 in the Wnt pathway. The hsbp1-deficient ES cells, EBs and zebrafish embryos with reduced HSBP1 levels exhibit elevated levels of Hsf1 activity and expression of heat shock proteins (Hsps). We conclude that HSBP1 plays an essential role during early mouse and zebrafish embryonic development.
Collapse
Affiliation(s)
- Binnur Eroglu
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, United States; Chaperone Biology, Georgia Regents University (GRU) Cancer Center, Medical College of Georgia (MCG), 1120 15th St., Augusta, GA 30912, United States
| | - Jin-Na Min
- Chaperone Biology, Georgia Regents University (GRU) Cancer Center, Medical College of Georgia (MCG), 1120 15th St., Augusta, GA 30912, United States.
| | - Yan Zhang
- Chaperone Biology, Georgia Regents University (GRU) Cancer Center, Medical College of Georgia (MCG), 1120 15th St., Augusta, GA 30912, United States.
| | - Edyta Szurek
- Institute of Molecular Medicine and Genetics, GRU, MCG, GA, United States
| | - Demetrius Moskophidis
- Chaperone Biology, Georgia Regents University (GRU) Cancer Center, Medical College of Georgia (MCG), 1120 15th St., Augusta, GA 30912, United States
| | - Ali Eroglu
- Institute of Molecular Medicine and Genetics, GRU, MCG, GA, United States.
| | - Nahid F Mivechi
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, United States; Chaperone Biology, Georgia Regents University (GRU) Cancer Center, Medical College of Georgia (MCG), 1120 15th St., Augusta, GA 30912, United States.
| |
Collapse
|
34
|
Na D, Rouf M, O'Kane CJ, Rubinsztein DC, Gsponer J. NeuroGeM, a knowledgebase of genetic modifiers in neurodegenerative diseases. BMC Med Genomics 2013; 6:52. [PMID: 24229347 PMCID: PMC3833180 DOI: 10.1186/1755-8794-6-52] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/08/2013] [Indexed: 11/26/2022] Open
Abstract
Background Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons in the human brain. Although the majority of NDs are sporadic, evidence is accumulating that they have a strong genetic component. Therefore, significant efforts have been made in recent years to not only identify disease-causing genes but also genes that modify the severity of NDs, so-called genetic modifiers. To date there exists no compendium that lists and cross-links genetic modifiers of different NDs. Description In order to address this need, we present NeuroGeM, the first comprehensive knowledgebase providing integrated information on genetic modifiers of nine different NDs in the model organisms D. melanogaster, C. elegans, and S. cerevisiae. NeuroGeM cross-links curated genetic modifier information from the different NDs and provides details on experimental conditions used for modifier identification, functional annotations, links to homologous proteins and color-coded protein-protein interaction networks to visualize modifier interactions. We demonstrate how this database can be used to generate new understanding through meta-analysis. For instance, we reveal that the Drosophila genes DnaJ-1, thread, Atx2, and mub are generic modifiers that affect multiple if not all NDs. Conclusion As the first compendium of genetic modifiers, NeuroGeM will assist experimental and computational scientists in their search for the pathophysiological mechanisms underlying NDs. http://chibi.ubc.ca/neurogem.
Collapse
Affiliation(s)
| | | | | | | | - Jörg Gsponer
- Department of Biochemistry and Molecular Biology, Centre for High-throughput Biology, University of British Columbia, 2125 East Mall, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
35
|
The ubiquitin ligase CHIP prevents SirT6 degradation through noncanonical ubiquitination. Mol Cell Biol 2013; 33:4461-72. [PMID: 24043303 DOI: 10.1128/mcb.00480-13] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The ubiquitin ligase CHIP (carboxyl terminus of Hsp70-interacting protein) regulates protein quality control, and CHIP deletion accelerates aging and reduces the life span in mice. Here, we reveal a mechanism for CHIP's influence on longevity by demonstrating that CHIP stabilizes the sirtuin family member SirT6, a lysine deacetylase/ADP ribosylase involved in DNA repair, metabolism, and longevity. In CHIP-deficient cells, SirT6 protein half-life is substantially reduced due to increased proteasome-mediated degradation, but CHIP overexpression in these cells increases SirT6 protein expression without affecting SirT6 transcription. CHIP noncanonically ubiquitinates SirT6 at K170, which stabilizes SirT6 and prevents SirT6 canonical ubiquitination by other ubiquitin ligases. In CHIP-depleted cells, SirT6 K170 mutation increases SirT6 half-life and prevents proteasome-mediated degradation. The global decrease in SirT6 expression in the absence of CHIP is associated with decreased SirT6 promoter occupancy, which increases histone acetylation and promotes downstream gene transcription in CHIP-depleted cells. Cells lacking CHIP are hypersensitive to DNA-damaging agents, but DNA repair and cell viability are rescued by enforced expression of SirT6. The discovery of this CHIP-SirT6 interaction represents a novel protein-stabilizing mechanism and defines an intersection between protein quality control and epigenetic regulation to influence pathways that regulate the biology of aging.
Collapse
|
36
|
Tabuchi Y, Kondo T. Targeting heat shock transcription factor 1 for novel hyperthermia therapy (review). Int J Mol Med 2013; 32:3-8. [PMID: 23636216 DOI: 10.3892/ijmm.2013.1367] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/14/2013] [Indexed: 11/06/2022] Open
Abstract
Hyperthermia (HT) has shown promising antitumor effects against various types of malignant tumors, and its pleiotropic effects support its combined use with radiotherapy and/or chemotherapy. However, HT is rendered less effective by the acquisition of thermoresistance in tumors, which arises through the elevation of heat shock proteins (HSPs) or other tumor responses. In mammals, the induction of HSPs is principally regulated at the transcriptional level by the activation of heat shock transcription factor 1 (HSF1). This transactivator has been shown to be abundantly expressed in a wide variety of tumors in humans. In addition, HSF1 participates in the initiation, proliferation and maintenance of tumors. Of note, HSF1 silencing has been shown to prevent the progression of tumors and to enhance their sensitivity to HT. Here, we review the physiological and pathological roles of HSF1 in cancer cells, and discuss its potential as a therapeutic target for HT therapy.
Collapse
Affiliation(s)
- Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan.
| | | |
Collapse
|
37
|
Dietary restriction supports peripheral nerve health by enhancing endogenous protein quality control mechanisms. Exp Gerontol 2012; 48:1085-90. [PMID: 23267845 DOI: 10.1016/j.exger.2012.12.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/07/2012] [Accepted: 12/17/2012] [Indexed: 01/07/2023]
Abstract
The peripheral nervous system (PNS) comprises of an extensive network of connections that convey information between the central nervous system (CNS) and peripheral organs. Long myelinated nerve fibers are particularly susceptible to age-related changes, as maintenance of the insulating glial membrane requires extensive synthesis and processing of many proteins. In rodent models, peripheral demyelination caused by genetic risk factors or by normal aging are attenuated by intermittent fasting (IF) or calorie restriction (CR) supporting a role for dietary intervention in preserving neural function. This review will summarize recent studies examining mechanisms by which life-long CR or extended IF supports peripheral nerve health.
Collapse
|
38
|
Dottorini T, Persampieri T, Palladino P, Baker DA, Spaccapelo R, Senin N, Crisanti A. Regulation of Anopheles gambiae male accessory gland genes influences postmating response in female. FASEB J 2012; 27:86-97. [PMID: 22997226 DOI: 10.1096/fj.12-219444] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In Drosophila, the accessory gland proteins (Acps) secreted from the male accessory glands (MAGs) and transferred along with sperm into the female reproductive tract have been implicated in triggering postmating behavioral changes, including refractoriness to subsequent mating and propensity to egg laying. Recently, Acps have been found also in Anopheles, suggesting similar functions. Understanding the mechanisms underlying transcriptional regulation of Acps and their functional role in modulating Anopheles postmating behavior may lead to the identification of novel vector control strategies to reduce mosquito populations. We identified heat-shock factor (HSF) binding sites within the Acp promoters of male Anopheles gambiae and discovered three distinct Hsf isoforms; one being significantly up-regulated in the MAGs after mating. Through genome-wide transcription analysis of Hsf-silenced males, we observed significant down-regulation in 50% of the Acp genes if compared to control males treated with a construct directed against an unrelated bacterial sequence. Treated males retained normal life span and reproductive behavior compared to control males. However, mated wild-type females showed a ∼46% reduction of egg deposition rate and a ∼23% reduction of hatching rate (∼58% combined reduction of progeny). Our results highlight an unsuspected role of HSF in regulating Acp transcription in A. gambiae and provide evidence that Acp down-regulation in males leads a significant reduction of progeny, thus opening new avenues toward the development of novel vector control strategies.
Collapse
Affiliation(s)
- Tania Dottorini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
West JD, Wang Y, Morano KA. Small molecule activators of the heat shock response: chemical properties, molecular targets, and therapeutic promise. Chem Res Toxicol 2012; 25:2036-53. [PMID: 22799889 DOI: 10.1021/tx300264x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
All cells have developed various mechanisms to respond and adapt to a variety of environmental challenges, including stresses that damage cellular proteins. One such response, the heat shock response (HSR), leads to the transcriptional activation of a family of molecular chaperone proteins that promote proper folding or clearance of damaged proteins within the cytosol. In addition to its role in protection against acute insults, the HSR also regulates lifespan and protects against protein misfolding that is associated with degenerative diseases of aging. As a result, identifying pharmacological regulators of the HSR has become an active area of research in recent years. Here, we review progress made in identifying small molecule activators of the HSR, what cellular targets these compounds interact with to drive response activation, and how such molecules may ultimately be employed to delay or reverse protein misfolding events that contribute to a number of diseases.
Collapse
Affiliation(s)
- James D West
- Biochemistry and Molecular Biology Program, Departments of Biology and Chemistry, The College of Wooster, Wooster, Ohio 44691, USA.
| | | | | |
Collapse
|
40
|
Xi C, Hu Y, Buckhaults P, Moskophidis D, Mivechi NF. Heat shock factor Hsf1 cooperates with ErbB2 (Her2/Neu) protein to promote mammary tumorigenesis and metastasis. J Biol Chem 2012; 287:35646-35657. [PMID: 22847003 DOI: 10.1074/jbc.m112.377481] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
ErbB2/Neu oncogene is overexpressed in 25% of invasive/metastatic breast cancers. We have found that deletion of heat shock factor Hsf1 in mice overexpressing ErbB2/Neu significantly reduces mammary tumorigenesis and metastasis. Hsf1(+/-)ErbB2/Neu(+) tumors exhibit reduced cellular proliferative and invasive properties associated with reduced activated ERK1/2 and reduced epithelial-mesenchymal transition (EMT). Hsf1(+/+)Neu(+) mammary epithelial cells exposed to TGFβ show high levels of ERK1/2 activity and EMT; this is associated with reduced expression of E-cadherin and increased expression of Slug and vimentin, a mesenchymal marker. In contrast, Hsf1(-/-)Neu(+) or Hsf1(+/+)Neu(+) cells do not exhibit activated ERK1/2 and show reduced EMT in the presence of TGFβ. The ineffective activation of the RAS/RAF/MEK/ERK1/2 signaling pathway in cells with reduced levels of HSF1 is due to the low levels of HSP90 in complex with RAF1 that are required for RAF1 stability and maturation. These results indicate a powerful inhibitory effect conferred by HSF1 downstream target genes in the inhibition of ErbB2-induced breast cancers in the absence of the Hsf1 gene.
Collapse
Affiliation(s)
- Caixia Xi
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Yanzhong Hu
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Phillip Buckhaults
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Demetrius Moskophidis
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Nahid F Mivechi
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Georgia Health Sciences University, Augusta, Georgia 30912.
| |
Collapse
|
41
|
Abstract
Although the adult human brain has a small number of neural stem cells, they are insufficient to repair the damaged brain to achieve significant functional recovery for neurodegenerative diseases and stroke. Stem cell therapy, by either enhancing endogenous neurogenesis, or transplanting stem cells, has been regarded as a promising solution. However, the harsh environment of the diseased brain posts a severe threat to the survival and correct differentiation of those new stem cells. Hormesis (or preconditioning, stress adaptation) is an adaptation mechanism by which cells or organisms are potentiated to survive an otherwise lethal condition, such as the harsh oxidative stress in the stroke brain. Stem cells treated by low levels of chemical, physical, or pharmacological stimuli have been shown to survive better in the neurodegenerative brain. Thus combining hormesis and stem cell therapy might improve the outcome for treatment of these diseases. In addition, since the cell death patterns and their underlying molecular mechanism may vary in different neurodegenerative diseases, even in different progression stages of the same disease, it is essential to design a suitable and optimum hormetic strategy that is tailored to the individual patient.
Collapse
Affiliation(s)
- Guanghu Wang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Georgia Health Sciences University
| |
Collapse
|
42
|
Skibinski GA, Boyd L. Ubiquitination is involved in secondary growth, not initial formation of polyglutamine protein aggregates in C. elegans. BMC Cell Biol 2012; 13:10. [PMID: 22494772 PMCID: PMC3368771 DOI: 10.1186/1471-2121-13-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 04/11/2012] [Indexed: 11/13/2022] Open
Abstract
Background Protein misfolding and subsequent aggregation are hallmarks of several human diseases. The cell has a variety of mechanisms for coping with misfolded protein stress, including ubiquitin-mediated protein degradation. In fact, the presence of ubiquitin at protein aggregates is a common feature of protein misfolding diseases. Ubiquitin conjugating enzymes (UBCs) are part of the cascade of enzymes responsible for the regulated attachment of ubiquitin to protein substrates. The specific UBC used during ubiquitination can determine the type of polyubiquitin chain linkage, which in turn plays an important role in determining the fate of the ubiquitinated protein. Thus, UBCs may serve an important role in the cellular response to misfolded proteins and the fate of protein aggregates. Results The Q82 strain of C. elegans harbors a transgene encoding an aggregation prone tract of 82 glutamine residues fused to green fluorescent protein (Q82::GFP) that is expressed in the body wall muscle. When measured with time-lapse microscopy in young larvae, the initial formation of individual Q82::GFP aggregates occurs in approximately 58 minutes. This process is largely unaffected by a mutation in the C. elegans E1 ubiquitin activating enzyme. RNAi of ubc-22, a nematode homolog of E2-25K, resulted in higher pre-aggregation levels of Q82::GFP and a faster initial aggregation rate relative to control. Knockdown of ubc-1 (RAD6 homolog), ubc-13, and uev-1 did not affect the kinetics of initial aggregation. However, RNAi of ubc-13 decreases the rate of secondary growth of the aggregate. This result is consistent with previous findings that aggregates in young adult worms are smaller after ubc-13 RNAi. mCherry::ubiquitin becomes localized to Q82::GFP aggregates during the fourth larval (L4) stage of life, a time point long after most aggregates have formed. FLIP and FRAP analysis indicate that mCherry::ubiquitin is considerably more mobile than Q82::GFP within aggregates. Conclusions These data indicate that initial formation of Q82::GFP aggregates in C. elegans is not directly dependent on ubiquitination, but is more likely a spontaneous process driven by biophysical properties in the cytosol such as the concentration of the aggregating species. The effect of ubiquitination appears to be most significant in later, secondary aggregate growth.
Collapse
Affiliation(s)
- Gregory A Skibinski
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville AL 35899, USA.
| | | |
Collapse
|
43
|
Jin X, Eroglu B, Cho W, Yamaguchi Y, Moskophidis D, Mivechi NF. Inactivation of heat shock factor Hsf4 induces cellular senescence and suppresses tumorigenesis in vivo. Mol Cancer Res 2012; 10:523-34. [PMID: 22355043 DOI: 10.1158/1541-7786.mcr-11-0530] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Studies suggest that Hsf4 expression correlates with its role in cell growth and differentiation. However, the role of Hsf4 in tumorigenesis in vivo remains unexplored. In this article, we provide evidence that absence of the Hsf4 gene suppresses evolution of spontaneous tumors arising in p53- or Arf-deficient mice. Furthermore, deletion of hsf4 alters the tumor spectrum by significantly inhibiting development of lymphomas that are normally observed in the majority of mice lacking p53 or Arf tumor suppressor genes. Using mouse embryo fibroblasts deficient in the hsf4 gene, we have found that these cells exhibit reduced proliferation that is associated with induction of senescence and senescence-associated β-galactosidase (SA-β-gal). Cellular senescence in hsf4-deficient cells is associated with the increased expression of the cyclin-dependent kinase inhibitors, p21 and p27 proteins. Consistent with the cellular senescence observed in vitro, specific normal tissues of hsf4(-/-) mice and tumors that arose in mice deficient in both hsf4 and p53 genes exhibit increased SA-β-gal activity and elevated levels of p27 compared with wild-type mice. These results suggest that hsf4 deletion-induced senescence is also present in vivo. Our results therefore indicate that Hsf4 is involved in modulation of cellular senescence, which can be exploited during cancer therapy.
Collapse
Affiliation(s)
- Xiongjie Jin
- Charlie Norwood VA Medical Center, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | | | | | | | | | |
Collapse
|
44
|
Bar-Lavan Y, Kosolapov L, Frumkin A, Ben-Zvi A. Regulation of cellular protein quality control networks in a multicellular organism. FEBS J 2012; 279:526-31. [PMID: 22177281 DOI: 10.1111/j.1742-4658.2011.08455.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The long-term health of all metazoan cells is linked to protein quality control, which is achieved by proteostasis, a complex network of molecular interactions that determines the health of the proteome under physiological or stress conditions. Studying the regulation of cellular proteostasis in the context of the whole organism has unraveled multiple layers of cell-nonautonomous regulation, including neuronal regulation, cell-to-cell stress signals and endocrine signaling that affect growth, development and aging. Here, we discuss emerging concepts in cell-nonautonomous regulation of protein quality control networks. The identification of organismal modulators of cellular proteostasis may present novel, yet general targets for misfolding disease intervention.
Collapse
Affiliation(s)
- Yael Bar-Lavan
- Department of Life Sciences and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | | | | |
Collapse
|
45
|
Riva L, Koeva M, Yildirim F, Pirhaji L, Dinesh D, Mazor T, Duennwald ML, Fraenkel E. Poly-glutamine expanded huntingtin dramatically alters the genome wide binding of HSF1. J Huntingtons Dis 2012; 1:33-45. [PMID: 23293686 PMCID: PMC3537492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
In Huntington's disease (HD), polyglutamine expansions in the huntingtin (Htt) protein cause subtle changes in cellular functions that, over-time, lead to neurodegeneration and death. Studies have indicated that activation of the heat shock response can reduce many of the effects of mutant Htt in disease models, suggesting that the heat shock response is impaired in the disease. To understand the basis for this impairment, we have used genome-wide chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-Seq) to examine the effects of mutant Htt on the master regulator of the heat shock response, HSF1. We find that, under normal conditions, HSF1 function is highly similar in cells carrying either wild-type or mutant Htt. However, polyQ-expanded Htt severely blunts the HSF1-mediated stress response. Surprisingly, we find that the HSF1 targets most affected upon stress are not directly associated with proteostasis, but with cytoskeletal binding, focal adhesion and GTPase activity. Our data raise the intriguing hypothesis that the accumulated damage from life-long impairment in these stress responses may contribute significantly to the etiology of Huntington's disease.
Collapse
Affiliation(s)
- Laura Riva
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Martina Koeva
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
- The Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Ferah Yildirim
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Leila Pirhaji
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Deepika Dinesh
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | - Tali Mazor
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| | | | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass Ave., Cambridge, MA, USA
| |
Collapse
|
46
|
Hu Y, Mivechi NF. Promotion of heat shock factor Hsf1 degradation via adaptor protein filamin A-interacting protein 1-like (FILIP-1L). J Biol Chem 2011; 286:31397-408. [PMID: 21784850 DOI: 10.1074/jbc.m111.255851] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Heat shock factor Hsf1 is involved in the regulation of a variety of cellular processes including heat shock response, development and differentiation, aging, and tumorigenesis. Hsf1 transcriptional activity is tightly controlled through phosphorylation, sumoylation, and acetylation, and through association with a number of regulatory proteins. However, regulation of Hsf1 protein stability or turnover remains unknown. We have identified a novel Hsf1-interacting protein, FILIP-1L, that was found to bind to Hsf1 through yeast two-hybrid screening. FILIP-1L encodes multiple isoforms spanning from 711 to 1135 amino acid residues. FILIP-1L contains four coiled-coil and two N-terminal leucine zipper domains. Ectopic expression of FILIP-1L reduces the expression of the Hsf1 protein because FILIP-1L promotes Hsf1 ubiquitination and degradation through the ubiquitin-proteasome system, leading to a reduction in Hsf1-mediated transcription. FILIP-1L, Hsf1, and the ubiquitin-binding domain of HhR23A, a receptor that transports polyubiquitinated proteins to the 19 S proteasome subunit targeting them for degradation, are found in a complex. This indicates that FILIP-1L is a potential adaptor that is involved in the Hsf1 degradation pathway. Taken together, our results indicate that FILIP-1L interacts with Hsf1, controlling its stability and thus modulating the heat shock response. These data indicate a novel function for FILIP-1L and a pathway for Hsf1 degradation through the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Yanzhong Hu
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | |
Collapse
|
47
|
Implication of heat shock factors in tumorigenesis: therapeutical potential. Cancers (Basel) 2011; 3:1158-81. [PMID: 24212658 PMCID: PMC3756408 DOI: 10.3390/cancers3011158] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 02/23/2011] [Indexed: 12/17/2022] Open
Abstract
Heat Shock Factors (HSF) form a family of transcription factors (four in mammals) which were named according to the discovery of their activation by a heat shock. HSFs trigger the expression of genes encoding Heat Shock Proteins (HSPs) that function as molecular chaperones, contributing to establish a cytoprotective state to various proteotoxic stresses and in pathological conditions. Increasing evidence indicates that this ancient transcriptional protective program acts genome-widely and performs unexpected functions in the absence of experimentally defined stress. Indeed, HSFs are able to re-shape cellular pathways controlling longevity, growth, metabolism and development. The most well studied HSF, HSF1, has been found at elevated levels in tumors with high metastatic potential and is associated with poor prognosis. This is partly explained by the above-mentioned cytoprotective (HSP-dependent) function that may enable cancer cells to adapt to the initial oncogenic stress and to support malignant transformation. Nevertheless, HSF1 operates as major multifaceted enhancers of tumorigenesis through, not only the induction of classical heat shock genes, but also of “non-classical” targets. Indeed, in cancer cells, HSF1 regulates genes involved in core cellular functions including proliferation, survival, migration, protein synthesis, signal transduction, and glucose metabolism, making HSF1 a very attractive target in cancer therapy. In this review, we describe the different physiological roles of HSFs as well as the recent discoveries in term of non-cogenic potential of these HSFs, more specifically associated to the activation of “non-classical” HSF target genes. We also present an update on the compounds with potent HSF1-modulating activity of potential interest as anti-cancer therapeutic agents.
Collapse
|
48
|
Impaired hippocampal spinogenesis and neurogenesis and altered affective behavior in mice lacking heat shock factor 1. Proc Natl Acad Sci U S A 2011; 108:1681-6. [PMID: 21205885 DOI: 10.1073/pnas.1016424108] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aberrant transcriptional regulation in the brain is thought to be one of the key components of the pathogenesis and pathophysiology of neuropsychiatric disorders. Heat shock factors (HSFs) modulate cellular homeostasis through the control of gene expression. However, the roles of HSFs in brain function have yet to be elucidated fully. In the present study, we attempted to clarify the role of HSF1-mediated gene regulation in neuronal and behavioral development using HSF1-deficient (HSF1(-/-)) mice. We found granule neurons of aberrant morphology and impaired neurogenesis in the dentate gyrus of HSF1(-/-) mice. In addition, HSF1(-/-) mice showed aberrant affective behavior, including reduced anxiety and sociability but increased depression-like behavior and aggression. Furthermore, HSF1 deficiency enhanced behavioral vulnerability to repeated exposure to restraint stress. Importantly, rescuing the HSF1 deficiency in the neonatal but not the adult hippocampus reversed the aberrant anxiety and depression-like behaviors. These results indicate a crucial role for hippocampal HSF1 in neuronal and behavioral development. Analysis of the molecular mechanisms revealed that HSF1 directly modulates the expression of polysialyltransferase genes, which then modulate polysialic acid-neural cell adhesion molecule (PSA-NCAM) levels in the hippocampus. Enzymatic removal of PSA from the neonatal hippocampus resulted in aberrant behavior during adulthood, similar to that observed in HSF1(-/-) mice. Thus, these results suggest that one role of HSF1 is to control hippocampal PSA-NCAM levels through the transcriptional regulation of polysialyltransferases, a process that might be involved in neuronal and behavioral development in mice.
Collapse
|
49
|
Abstract
Heat-shock transcription factors (Hsfs) regulate transcription of heat-shock proteins as well as other genes whose promoters contain heat-shock elements. There are at least five Hsfs in mammalian cells, Hsf1, Hsf2, Hsf3, Hsf4, and Hsfy. To understand the physiological roles of Hsf1, Hsf2, and Hsf4 in vivo, we generated knockout mouse lines for these factors. In this chapter, we describe the design of the targeting vectors, the plasmids used, and the successful generation of mice lacking the individual genes. We also briefly describe what we have learned about the physiological functions of these genes in vivo.
Collapse
Affiliation(s)
- Xiongjie Jin
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | |
Collapse
|
50
|
Abstract
The heat shock response was originally characterized as the induction of a set of major heat shock proteins encoded by heat shock genes. Because heat shock proteins act as molecular chaperones that facilitate protein folding and suppress protein aggregation, this response plays a major role in maintaining protein homeostasis. The heat shock response is regulated mainly at the level of transcription by heat shock factors (HSFs) in eukaryotes. HSF1 is a master regulator of the heat shock genes in mammalian cells, as is HSF3 in avian cells. HSFs play a significant role in suppressing protein misfolding in cells and in ameliorating the progression of Caenorhabditis elegans, Drosophila and mouse models of protein-misfolding disorders, by inducing the expression of heat shock genes. Recently, numerous HSF target genes were identified, such as the classical heat shock genes and other heat-inducible genes, called nonclassical heat shock genes in this study. Importance of the expression of the nonclassical heat shock genes was evidenced by the fact that mouse HSF3 and chicken HSF1 play a substantial role in the protection of cells from heat shock without inducing classical heat shock genes. Furthermore, HSF2 and HSF4, as well as HSF1, shown to have roles in development, were also revealed to be necessary for the expression of certain nonclassical heat shock genes. Thus, the heat shock response regulated by the HSF family should consist of the induction of classical as well as of nonclassical heat shock genes, both of which might be required to maintain protein homeostasis.
Collapse
|