1
|
Wuputra K, Hsu WH, Ku CC, Yang YH, Kuo KK, Yu FJ, Yu HS, Nagata K, Wu DC, Kuo CH, Yokoyama KK. The AHR-NRF2-JDP2 gene battery: Ligand-induced AHR transcriptional activation. Biochem Pharmacol 2025; 233:116761. [PMID: 39855429 DOI: 10.1016/j.bcp.2025.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Aryl hydrocarbon receptor (AHR) and nuclear factor-erythroid 2-related factor 2 (NRF2) can regulate a series of genes encoding the detoxifying phase I and II enzymes, via a signaling crosstalk known as the "AHR-NRF2 gene battery". The chromatin transcriptional regulator Jun dimerization protein 2 (JDP2) plays a central role in thetranscription of AHR gene in response to the phase I enzyme ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin. It forms a transcriptional complex with AHR-AHR nuclear translocator (ARNT) and NRF2-small musculoaponeurotic fibrosarcoma proteins (sMAF), which are then recruited to the respective cis-elements, such as dioxin response elements and antioxidant response elements, respectively, in the AHR promoter. Here, we present a revised description of the AHR-NRF2 gene battery as the AHR-NRF2-JDP2 gene battery for transactivating the AHR promoter by phase I enzyme ligands. The chromatin regulator JDP2 was found to be involved in the movement of AHR-NRF2 complexes from the dioxin response element to the antioxidant response element in the AHR promoter, during its activation in a spatiotemporal manner. This new epigenetic and chromatin remodeling role of AHR-NRF2-JDP2 axis is useful for identifying new therapeutic targets for various diseases, including immunological response, detoxification, development, and cancer-related diseases.
Collapse
Affiliation(s)
- Kenly Wuputra
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chia-Chen Ku
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ya-Han Yang
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Kung-Kai Kuo
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Fang-Jung Yu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.
| | - Hsin-Su Yu
- Emeritus Professor in College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Kyosuke Nagata
- Professor, Insitutte of Medicine, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Superintendant in Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan.
| | - Kazunari K Yokoyama
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
2
|
Stem AD, Michel CR, Harris PS, Rogers KL, Gibb M, Roncal-Jimenez CA, Reisdorph R, Johnson RJ, Roede JR, Fritz KS, Brown JM. Modulation of the thiol redox proteome by sugarcane ash-derived silica nanoparticles: insights into chronic kidney disease of unknown etiology. Part Fibre Toxicol 2025; 22:3. [PMID: 39910563 PMCID: PMC11800628 DOI: 10.1186/s12989-025-00619-8] [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: 10/21/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION Chronic kidney disease of unknown etiology (CKDu) is an epidemic which is increasingly prevalent among agricultural workers and nearby communities, particularly those involved in the harvest of sugarcane. While CKDu is likely multifactorial, occupational exposure to silica nanoparticles (SiNPs), a major constituent within sugarcane ash, has gained increased attention as a potential contributor. SiNPs have high potential for generation of reactive oxygen species (ROS), and their accumulation in kidney could result in oxidative stress induced kidney damage consistent with CKDu pathology. METHODS In order to characterize the impact of sugarcane ash derived (SAD) SiNPs on human kidney proximal convoluted tubule (PCT) cells and identify potential mechanisms of toxicity, HK-2 cells were exposed to treatments of either pristine, manufactured, 200 nm SiNPs or SAD SiNPs and changes to cellular energy metabolism and redox state were determined. To determine how the cellular redox environment may influence PCT cell function and toxicity, the redox proteome was examined using cysteine-targeted click chemistry proteomics. RESULTS Pristine, 200 nm SiNPs induced minimal changes to energy metabolism and proteomic profiles in vitro while treatment with SAD SiNPs resulted in mitochondrial membrane hyperpolarization, inhibited mitochondrial respiration, increased reactive oxygen species generation, and redox proteomic trends suggesting activation of aryl hydrocarbon receptor (AHR) and other signaling pathways with known roles in mitochondrial inhibition and CKD progression. CONCLUSION Results suggest that PCT cell exposure to SAD SiNPs could promote glycolytic and fibrotic shifts consistent with CKDu pathology via oxidative stress-mediated disruption of redox signaling pathways.
Collapse
Affiliation(s)
- Arthur D Stem
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Cole R Michel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Peter S Harris
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Keegan L Rogers
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Matthew Gibb
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Carlos A Roncal-Jimenez
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Kristofer S Fritz
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Jared M Brown
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Campus, Aurora, CO, USA.
| |
Collapse
|
3
|
Elbassiouny AA, Buck LT, Abatti LE, Mitchell JA, Crampton WGR, Lovejoy NR, Chang BSW. Evolution of a novel regulatory mechanism of hypoxia inducible factor in hypoxia-tolerant electric fishes. J Biol Chem 2024; 300:105727. [PMID: 38325739 PMCID: PMC10958119 DOI: 10.1016/j.jbc.2024.105727] [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: 10/23/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Hypoxia is a significant source of metabolic stress that activates many cellular pathways involved in cellular differentiation, proliferation, and cell death. Hypoxia is also a major component in many human diseases and a known driver of many cancers. Despite the challenges posed by hypoxia, there are animals that display impressive capacity to withstand lethal levels of hypoxia for prolonged periods of time and thus offer a gateway to a more comprehensive understanding of the hypoxic response in vertebrates. The weakly electric fish genus Brachyhypopomus inhabits some of the most challenging aquatic ecosystems in the world, with some species experiencing seasonal anoxia, thus providing a unique system to study the cellular and molecular mechanisms of hypoxia tolerance. In this study, we use closely related species of Brachyhypopomus that display a range of hypoxia tolerances to probe for the underlying molecular mechanisms via hypoxia inducible factors (HIFs)-transcription factors known to coordinate the cellular response to hypoxia in vertebrates. We find that HIF1⍺ from hypoxia tolerant Brachyhypopomus species displays higher transactivation in response to hypoxia than that of intolerant species, when overexpressed in live cells. Moreover, we identified two SUMO-interacting motifs near the oxygen-dependent degradation and transactivation domains of the HIF1⍺ protein that appear to boost transactivation of HIF1, regardless of the genetic background. Together with computational analyses of selection, this shows that evolution of HIF1⍺ are likely to underlie adaptations to hypoxia tolerance in Brachyhypopomus electric fishes, with changes in two SUMO-interacting motifs facilitating the mechanism of this tolerance.
Collapse
Affiliation(s)
- Ahmed A Elbassiouny
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada.
| | - Leslie T Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Luis E Abatti
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | | | - Nathan R Lovejoy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada; Department of Biological Sciences, University of Toronto Scarborough, Scarborough, Ontario, Canada
| | - Belinda S W Chang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Batie M, Fasanya T, Kenneth NS, Rocha S. Oxygen-regulated post-translation modifications as master signalling pathway in cells. EMBO Rep 2023; 24:e57849. [PMID: 37877678 DOI: 10.15252/embr.202357849] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/22/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Oxygen is essential for viability in mammalian organisms. However, cells are often exposed to changes in oxygen availability, due to either increased demand or reduced oxygen supply, herein called hypoxia. To be able to survive and/or adapt to hypoxia, cells activate a variety of signalling cascades resulting in changes to chromatin, gene expression, metabolism and viability. Cellular signalling is often mediated via post-translational modifications (PTMs), and this is no different in response to hypoxia. Many enzymes require oxygen for their activity and oxygen can directly influence several PTMS. Here, we review the direct impact of changes in oxygen availability on PTMs such as proline, asparagine, histidine and lysine hydroxylation, lysine and arginine methylation and cysteine dioxygenation, with a focus on mammalian systems. In addition, indirect hypoxia-dependent effects on phosphorylation, ubiquitination and sumoylation will also be discussed. Direct and indirect oxygen-regulated changes to PTMs are coordinated to achieve the cell's ultimate response to hypoxia. However, specific oxygen sensitivity and the functional relevance of some of the identified PTMs still require significant research.
Collapse
Affiliation(s)
- Michael Batie
- Department of Biochemistry, Cell and Systems Biology, Institute of Molecular Systems and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Temitope Fasanya
- Department of Biochemistry, Cell and Systems Biology, Institute of Molecular Systems and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Niall S Kenneth
- Department of Biochemistry, Cell and Systems Biology, Institute of Molecular Systems and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sonia Rocha
- Department of Biochemistry, Cell and Systems Biology, Institute of Molecular Systems and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
5
|
Opitz CA, Holfelder P, Prentzell MT, Trump S. The complex biology of aryl hydrocarbon receptor activation in cancer and beyond. Biochem Pharmacol 2023; 216:115798. [PMID: 37696456 PMCID: PMC10570930 DOI: 10.1016/j.bcp.2023.115798] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
The aryl hydrocarbon receptor (AHR) signaling pathway is a complex regulatory network that plays a critical role in various biological processes, including cellular metabolism, development, and immune responses. The complexity of AHR signaling arises from multiple factors, including the diverse ligands that activate the receptor, the expression level of AHR itself, and its interaction with the AHR nuclear translocator (ARNT). Additionally, the AHR crosstalks with the AHR repressor (AHRR) or other transcription factors and signaling pathways and it can also mediate non-genomic effects. Finally, posttranslational modifications of the AHR and its interaction partners, epigenetic regulation of AHR and its target genes, as well as AHR-mediated induction of enzymes that degrade AHR-activating ligands may contribute to the context-specificity of AHR activation. Understanding the complexity of AHR signaling is crucial for deciphering its physiological and pathological roles and developing therapeutic strategies targeting this pathway. Ongoing research continues to unravel the intricacies of AHR signaling, shedding light on the regulatory mechanisms controlling its diverse functions.
Collapse
Affiliation(s)
- Christiane A Opitz
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, 69120 Heidelberg, Germany.
| | - Pauline Holfelder
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Mirja Tamara Prentzell
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Saskia Trump
- Molecular Epidemiology Unit, Berlin Institute of Health at Charité and the German Cancer Consortium (DKTK), Partner Site Berlin, a partnership between DKFZ and Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
6
|
Gu Y, Fang Y, Wu X, Xu T, Hu T, Xu Y, Ma P, Wang Q, Shu Y. The emerging roles of SUMOylation in the tumor microenvironment and therapeutic implications. Exp Hematol Oncol 2023; 12:58. [PMID: 37415251 PMCID: PMC10324244 DOI: 10.1186/s40164-023-00420-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Tumor initiation, progression, and response to therapies depend to a great extent on interactions between malignant cells and the tumor microenvironment (TME), which denotes the cancerous/non-cancerous cells, cytokines, chemokines, and various other factors around tumors. Cancer cells as well as stroma cells can not only obtain adaption to the TME but also sculpt their microenvironment through a series of signaling pathways. The post-translational modification (PTM) of eukaryotic cells by small ubiquitin-related modifier (SUMO) proteins is now recognized as a key flexible pathway. Proteins involved in tumorigenesis guiding several biological processes including chromatin organization, DNA repair, transcription, protein trafficking, and signal conduction rely on SUMOylation. The purpose of this review is to explore the role that SUMOylation plays in the TME formation and reprogramming, emphasize the importance of targeting SUMOylation to intervene in the TME and discuss the potential of SUMOylation inhibitors (SUMOi) in ameliorating tumor prognosis.
Collapse
Affiliation(s)
- Yunru Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Yuan Fang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Xi Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Tingting Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Tong Hu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Yangyue Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui Province People’s Republic of China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, People’s Republic of China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Karandikar P, Gerstl JVE, Kappel AD, Won SY, Dubinski D, Garcia-Segura ME, Gessler FA, See AP, Peruzzotti-Jametti L, Bernstock JD. SUMOtherapeutics for Ischemic Stroke. Pharmaceuticals (Basel) 2023; 16:ph16050673. [PMID: 37242456 DOI: 10.3390/ph16050673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The small, ubiquitin-like modifier (SUMO) is a post-translational modifier with a profound influence on several key biological processes, including the mammalian stress response. Of particular interest are its neuroprotective effects, first recognized in the 13-lined ground squirrel (Ictidomys tridecemlineatus), in the context of hibernation torpor. Although the full scope of the SUMO pathway is yet to be elucidated, observations of its importance in managing neuronal responses to ischemia, maintaining ion gradients, and the preconditioning of neural stem cells make it a promising therapeutic target for acute cerebral ischemia. Recent advances in high-throughput screening have enabled the identification of small molecules that can upregulate SUMOylation, some of which have been validated in pertinent preclinical models of cerebral ischemia. Accordingly, the present review aims to summarize current knowledge and highlight the translational potential of the SUMOylation pathway in brain ischemia.
Collapse
Affiliation(s)
- Paramesh Karandikar
- T. H. Chan School of Medicine, University of Massachusetts, Worcester, MA 01655, USA
| | - Jakob V E Gerstl
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Ari D Kappel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA 02215, USA
| | - Sae-Yeon Won
- Department of Neurosurgery, University Medicine Rostock, 18057 Rostock, Germany
| | - Daniel Dubinski
- Department of Neurosurgery, University Medicine Rostock, 18057 Rostock, Germany
| | - Monica Emili Garcia-Segura
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
- NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Florian A Gessler
- Department of Neurosurgery, University Medicine Rostock, 18057 Rostock, Germany
| | - Alfred Pokmeng See
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA 02215, USA
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
- NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Department of Neurosurgery, University Medicine Rostock, 18057 Rostock, Germany
- Koch Institute for Integrated Cancer Research, MIT, Cambridge, MA 02142, USA
| |
Collapse
|
8
|
Tomc J, Debeljak N. Molecular Insights into the Oxygen-Sensing Pathway and Erythropoietin Expression Regulation in Erythropoiesis. Int J Mol Sci 2021; 22:ijms22137074. [PMID: 34209205 PMCID: PMC8269393 DOI: 10.3390/ijms22137074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Erythropoiesis is regulated by several factors, including the oxygen-sensing pathway as the main regulator of erythropoietin (EPO) synthesis in the kidney. The release of EPO from the kidney and its binding to the EPO receptor (EPOR) on erythrocyte progenitor cells in the bone marrow results in increased erythropoiesis. Any imbalance in these homeostatic mechanisms can lead to dysregulated erythropoiesis and hematological disorders. For example, mutations in genes encoding key players of oxygen-sensing pathway and regulation of EPO production (HIF-EPO pathway), namely VHL, EGLN, EPAS1 and EPO, are well known causative factors that contribute to the development of erythrocytosis. We aimed to investigate additional molecular mechanisms involved in the HIF-EPO pathway that correlate with erythropoiesis. To this end, we conducted an extensive literature search and used several in silico tools. We identified genes encoding transcription factors and proteins that control transcriptional activation or repression; genes encoding kinases, deacetylases, methyltransferases, conjugating enzymes, protein ligases, and proteases involved in post-translational modifications; and genes encoding nuclear transport receptors that regulate nuclear transport. All these genes may modulate the stability or activity of HIF2α and its partners in the HIF-EPO pathway, thus affecting EPO synthesis. The theoretical information we provide in this work can be a valuable tool for a better understanding of one of the most important regulatory pathways in the process of erythropoiesis. This knowledge is necessary to discover the causative factors that may contribute to the development of hematological diseases and improve current diagnostic and treatment solutions in this regard.
Collapse
Affiliation(s)
- Jana Tomc
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nataša Debeljak
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Correspondence: ; Tel.: +386-1-543-7645
| |
Collapse
|
9
|
The Role of Sumoylation in the Response to Hypoxia: An Overview. Cells 2020; 9:cells9112359. [PMID: 33114748 PMCID: PMC7693722 DOI: 10.3390/cells9112359] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Sumoylation is the covalent attachment of the small ubiquitin-related modifier (SUMO) to a vast variety of proteins in order to modulate their function. Sumoylation has emerged as an important modification with a regulatory role in the cellular response to different types of stress including osmotic, hypoxic and oxidative stress. Hypoxia can occur under physiological or pathological conditions, such as ischemia and cancer, as a result of an oxygen imbalance caused by low supply and/or increased consumption. The hypoxia inducible factors (HIFs), and the proteins that regulate their fate, are critical molecular mediators of the response to hypoxia and modulate procedures such as glucose and lipid metabolism, angiogenesis, erythropoiesis and, in the case of cancer, tumor progression and metastasis. Here, we provide an overview of the sumoylation-dependent mechanisms that are activated under hypoxia and the way they influence key players of the hypoxic response pathway. As hypoxia is a hallmark of many diseases, understanding the interrelated connections between the SUMO and the hypoxic signaling pathways can open the way for future molecular therapeutic interventions.
Collapse
|
10
|
Jin X, Hapsari ND, Lee S, Jo K. DNA binding fluorescent proteins as single-molecule probes. Analyst 2020; 145:4079-4095. [DOI: 10.1039/d0an00218f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
DNA binding fluorescent proteins are useful probes for a broad range of biological applications.
Collapse
Affiliation(s)
- Xuelin Jin
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| | - Natalia Diyah Hapsari
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
- Chemistry Education Program
| | - Seonghyun Lee
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| | - Kyubong Jo
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| |
Collapse
|
11
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. Aryl-Hydrocarbon Receptor as a Potential Target for Anticancer Therapy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819010116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
12
|
Abstract
Cells respond to hypoxia by shifting cellular processes from general housekeeping functions to activating specialized hypoxia-response pathways. Oxygen plays an important role in generating ATP to maintain a productive rate of protein synthesis in normoxia. In hypoxia, the rate of the canonical protein synthesis pathway is significantly slowed and impaired due to limited ATP availability, necessitating an alternative mechanism to mediate protein synthesis and facilitate adaptation. Hypoxia adaptation is largely mediated by hypoxia-inducible factors (HIFs). While HIFs are well known for their transcriptional functions, they also play imperative roles in translation to mediate hypoxic protein synthesis. Such adaptations to hypoxia are often hyperactive in solid tumors, contributing to the expression of cancer hallmarks, including treatment resistance. The current literature on protein synthesis in hypoxia is reviewed here, inclusive of hypoxia-specific mRNA selection to translation termination. Current HIF targeting therapies are also discussed as are the opportunities involved with targeting hypoxia specific protein synthesis pathways.
Collapse
Affiliation(s)
- Nancy T Chee
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL, 33136, USA
| | - Ines Lohse
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL, 33136, USA
| | - Shaun P Brothers
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
13
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. [Aryl-hydrocarbon receptor as a potential target for anticancer therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:397-415. [PMID: 30378556 DOI: 10.18097/pbmc20186405397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Aryl-hydrocarbon receptor (Aryl Hydrocarbon Receptor, AHR) is a ligand-dependent transcription factor, whose functions are related to xenobiotic detoxification, response to inflammation, and maintenance of tissue homeostasis. Recent investigations suggest that AHR also plays an important role in the processes of carcinogenesis. Increased expression of AHR is observed in several types of tumors and tumor cell lines. In addition, it turned out that the composition of pharmaceutical drugs used in oncotherapy includes some ligands AHR. These facts allow us to consider an aryl-hydrocarbon receptor as a potential target for anticancer therapy, especially for the treatment of severe cancers whose treatment options are very limited or do not exist at all. In this review the examples of AHR ligands' effect on tumor cell cultures and on model mice lines with AHR-dependent response are discussed.
Collapse
Affiliation(s)
- J E Vorontsova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - R O Cherezov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - B A Kuzin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - O B Simonova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
14
|
Elizondo G, Vega L. Ubiquitination/sumoylation: An alternative pathway to modify gene regulation directed by xenosensors. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2018.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Steffensen AB, Andersen MN, Mutsaers N, Mujezinovic A, Schmitt N. SUMO co-expression modifies K V 11.1 channel activity. Acta Physiol (Oxf) 2018; 222. [PMID: 28888063 DOI: 10.1111/apha.12974] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 08/31/2017] [Accepted: 09/06/2017] [Indexed: 11/27/2022]
Abstract
AIM The voltage-gated potassium channel KV 11.1 is the molecular basis for the IKr current, which plays an important role in cardiac physiology. Its malfunction is associated with both inherited and acquired cardiac arrhythmias. Native currents differ from those in experimental models, suggesting additional regulatory mechanisms. We hypothesized that the post-translational modification sumoylation fine-tunes channel activity. METHODS The functional effects of sumoylation on KV 11.1 were addressed by employing two-electrode voltage-clamp (TEVC) experiments in Xenopus laevis oocytes. Site-directed mutagenesis enabled a further analysis of the SUMO-target amino acids. We assessed protein expression levels and used confocal imaging for localization studies. RESULTS Co-expression with Ubc9 and SUMO alters the electrophysiological properties of KV 11.1 leading to a decrease in steady-state current amplitude largely due to faster inactivation and alteration of deactivation kinetics. We identified three lysines (K21, K93 and K116) in the PAS domain as the putative SUMO-targets. CONCLUSION This study indicates KV 11.1 as a sumoylation target and offers three main targets: K21, K93, and K116. Furthermore, it proposes an underlying mechanism for the observed kinetic impact of the PAS domain.
Collapse
Affiliation(s)
- A. B. Steffensen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - M. N. Andersen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - N. Mutsaers
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - A. Mujezinovic
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - N. Schmitt
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
16
|
Sakurai S, Shimizu T, Ohto U. The crystal structure of the AhRR-ARNT heterodimer reveals the structural basis of the repression of AhR-mediated transcription. J Biol Chem 2017; 292:17609-17616. [PMID: 28904176 DOI: 10.1074/jbc.m117.812974] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/12/2017] [Indexed: 12/19/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin and related compounds are extraordinarily potent environmental toxic pollutants. Most of the 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicities are mediated by aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor belonging to the basic helix-loop-helix (bHLH) Per-ARNT-Sim (PAS) family. Upon ligand binding, AhR forms a heterodimer with AhR nuclear translocator (ARNT) and induces the expression of genes involved in various biological responses. One of the genes induced by AhR encodes AhR repressor (AhRR), which also forms a heterodimer with ARNT and represses the activation of AhR-dependent transcription. The control of AhR activation is critical for managing AhR-mediated diseases, but the mechanisms by which AhRR represses AhR activation remain poorly understood, because of the lack of structural information. Here, we determined the structure of the AhRR-ARNT heterodimer by X-ray crystallography, which revealed an asymmetric intertwined domain organization presenting structural features that are both conserved and distinct among bHLH-PAS family members. The structures of AhRR-ARNT and AhR-ARNT were similar in the bHLH-PAS-A region, whereas the PAS-B of ARNT in the AhRR-ARNT complex exhibited a different domain arrangement in this family reported so far. The structure clearly disclosed that AhRR competitively represses AhR binding to ARNT and target DNA and further suggested the existence of an AhRR-ARNT-specific repression mechanism. This study provides a structural basis for understanding the mechanism by which AhRR represses AhR-mediated gene transcription.
Collapse
Affiliation(s)
- Shunya Sakurai
- From the Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan and
| | - Toshiyuki Shimizu
- From the Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan and .,the Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Umeharu Ohto
- From the Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan and
| |
Collapse
|
17
|
Abstract
Protein modification with the small ubiquitin-related modifier (SUMO) can affect protein function, enzyme activity, protein-protein interactions, protein stability, protein targeting and cellular localization. SUMO influences the function and regulation of metabolic enzymes within pathways, and in some cases targets entire metabolic pathways by affecting the activity of transcription factors or by facilitating the translocation of entire metabolic pathways to subcellular compartments. SUMO modification is also a key component of nutrient- and metabolic-sensing mechanisms that regulate cellular metabolism. In addition to its established roles in maintaining metabolic homeostasis, there is increasing evidence that SUMO is a key factor in facilitating cellular stress responses through the regulation and/or adaptation of the most fundamental metabolic processes, including energy and nucleotide metabolism. This review focuses on the role of SUMO in cellular metabolism and metabolic disease.
Collapse
|
18
|
Wang F, Cai F, Shi R, Wei JN, Wu XT. Hypoxia regulates sumoylation pathways in intervertebral disc cells: implications for hypoxic adaptations. Osteoarthritis Cartilage 2016; 24:1113-24. [PMID: 26826302 DOI: 10.1016/j.joca.2016.01.134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the hypoxic regulation of sumoylation pathways and cell viability in nucleus pulposus (NP) and annulus fibrosus (AF) cells. DESIGN Expression of small ubiquitin-like modifier (SUMO) molecules, SUMO E1 activating enzymes SAE1 and SAE2, SUMO E2 conjugating enzyme UBC9, and de-sumoylation enzyme sentrin/SUMO-specific proteases (SENP)1 was immunolocalized in rat intervertebral disc (IVD) cells. NP and AF cells were cultured in hypoxia and cell viability was evaluated by quantifying cell proliferation, cellular senescence, apoptosis, and cell cycle distribution. Hypoxic regulation of sumoylation pathways was studied by analyzing the transcription and expression of SUMO molecules and sumoylation enzymes. Loss of function study using SENP1 siRNA was performed to investigate the regulatory role of sumoylation on the function of hypoxia inducible factor 1α (HIF-1α) and the hypoxic tolerance of IVD cells. RESULTS Sumoylation pathways were expressed in IVD cells and localized predominantly in nuclei. Both NP and AF cells maintained viability under hypoxia and upregulated the expression of SENP1. In NP cells hypoxia transiently increased the expression of SUMO-1, SUMO-2/3, SAE2, and UBC9, whereas SUMO-1 was elevated while SUMO-2/3, SAE1, SAE2, and UBC9 were reduced by low oxygen tensions in AF cells. Although downregulation of SENP1 decreased the transcriptional activity of HIF-1α, the viability of disc cells showed no significant loss under hypoxia. CONCLUSIONS NP and AF cells equally tolerate oxygen deficiency, but differently regulate the sumoylation pathways under hypoxia. The distinct sumoylation dynamics may help extend our understanding of the cell-specific regulation of the molecular basis that promotes cell survival in the hypoxic IVD.
Collapse
Affiliation(s)
- F Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - F Cai
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - R Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - J-N Wei
- Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - X-T Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| |
Collapse
|
19
|
Abstract
The small ubiquitin-like modifier SUMO regulates many aspects of cellular physiology to maintain cell homeostasis, both under normal conditions and during cell stress. Components of the transcriptional apparatus and chromatin are among the most prominent SUMO substrates. The prevailing view is that SUMO serves to repress transcription. However, as we will discuss in this review, this model needs to be refined, because recent studies have revealed that SUMO can also have profound positive effects on transcription.
Collapse
Affiliation(s)
- Pierre Chymkowitch
- Department of Molecular Biology, Institute of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Aurélie Nguéa P
- Department of Molecular Biology, Institute of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Jorrit M Enserink
- Department of Molecular Biology, Institute of Microbiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
20
|
Jiang Y, Wang J, Tian H, Li G, Zhu H, Liu L, Hu R, Dai A. Increased SUMO-1 expression in response to hypoxia: Interaction with HIF-1α in hypoxic pulmonary hypertension. Int J Mol Med 2015; 36:271-81. [PMID: 25976847 DOI: 10.3892/ijmm.2015.2209] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/28/2015] [Indexed: 11/06/2022] Open
Abstract
Pulmonary hypertension (PH) develops in 30-70% of chronic obstructive pulmonary disease patients and increases morbidity and mortality. The present study aimed to investigate the regulation of small ubiquitin‑related modifier‑1 (SUMO‑1) expression in response to hypoxia. The experiments were carried out in vitro in rat pulmonary arterial smooth muscle cells (PASMCs) and in vivo using a rat hypoxic PH (HPH) model. A significant increase in SUMO‑1 mRNA and protein levels was observed following hypoxic stimulation in vivo and in vitro. SUMO‑1 is known to interact with various transcription factors, including hypoxia‑inducible factor‑1α (HIF‑1α) in vitro. Notably, the expression of HIF‑1α and its target gene, vascular endothelial growth factor, was increased by hypoxia in HPH. In addition, the present data suggest that SUMO‑1 regulated HIF‑1α in response to hypoxia (gene silencing and overexpression). Finally, the co‑immunoprecipitation assays suggest a direct and specific interaction between SUMO‑1 and HIF‑1α. In conclusion, SUMO‑1 may participate in the modulation of HIF‑1α through sumoylation in HPH. However, further studies are required to confirm this.
Collapse
Affiliation(s)
- Yongliang Jiang
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Jing Wang
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Hua Tian
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Guang Li
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Hao Zhu
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Lei Liu
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Ruicheng Hu
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| | - Aiguo Dai
- Institute of Respiratory Medicine, Hunan Province Geriatric Hospital, Changsha, Hunan 410016, P.R. China
| |
Collapse
|
21
|
Cho SH, Park SY, Lee EJ, Cho YH, Park HS, Hong SH, Kim WJ. Regulation of CYP1A1 and Inflammatory Cytokine by NCOA7 Isoform 4 in Response to Dioxin Induced Airway Inflammation. Tuberc Respir Dis (Seoul) 2015; 78:99-105. [PMID: 25861343 PMCID: PMC4388907 DOI: 10.4046/trd.2015.78.2.99] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/10/2014] [Accepted: 12/10/2014] [Indexed: 11/24/2022] Open
Abstract
Background Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, binds to a wide variety of synthetic and naturally occurring compounds. AhR is involved in the regulation of inflammatory response during acute and chronic respiratory diseases. We investigated whether nuclear receptor coactivator 7 (NCOA7) could regulate transcriptional levels of AhR target genes and inflammatory cytokines in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated human bronchial epithelial cells. This study was based on our previous study that NCOA7 was differentially expressed between normal and chronic obstructive pulmonary disease lung tissues. Methods BEAS-2B and A549 cells grown under serum-free conditions were treated with or without TCDD (0.15 nM and 6.5 nM) for 24 hours after transfection of pCMV-NCOA7 isoform 4. Expression levels of cytochrome P4501A1 (CYP1A1), IL-6, and IL-8 were measured by quantitative real-time polymerase chain reaction. Results The transcriptional activities of CYP1A1 and inflammatory cytokines were strongly induced by TCDD treatment in both BEAS-2B and A549 cell lines. The NCOA7 isoform 4 oppositely regulated the transcriptional activities of CYP1A1 and inflammatory cytokines between BEAS-2B and A549 cell lines. Conclusion Our results suggest that NCOA7 could act as a regulator in the TCDD-AhR signaling pathway with dual roles in normal and abnormal physiological conditions.
Collapse
Affiliation(s)
- Sung-Hwan Cho
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea
| | - Shin Young Park
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea
| | - Eun Jeong Lee
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Yo Han Cho
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hyun Sun Park
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Woo Jin Kim
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea. ; Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
22
|
Silveirinha V, Stephens GJ, Cimarosti H. Molecular targets underlying SUMO-mediated neuroprotection in brain ischemia. J Neurochem 2013; 127:580-91. [PMID: 23786482 DOI: 10.1111/jnc.12347] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 01/11/2023]
Abstract
SUMOylation (small ubiquitin-like modifier conjugation) is an important post-translational modification which is becoming increasingly implicated in the altered protein dynamics associated with brain ischemia. The function of SUMOylation in cells undergoing ischemic stress and the identity of small ubiquitin-like modifier (SUMO) targets remain in most cases unknown. However, the emerging consensus is that SUMOylation of certain proteins might be part of an endogenous neuroprotective response. This review brings together the current understanding of the underlying mechanisms and downstream effects of SUMOylation in brain ischemia, including processes such as autophagy, mitophagy and oxidative stress. We focus on recent advances and controversies regarding key central nervous system proteins, including those associated with the nucleus, cytoplasm and plasma membrane, such as glucose transporters (GLUT1, GLUT4), excitatory amino acid transporter 2 glutamate transporters, K+ channels (K2P1, Kv1.5, Kv2.1), GluK2 kainate receptors, mGluR8 glutamate receptors and CB1 cannabinoid receptors, which are reported to be SUMO-modified. A discussion of the roles of these molecular targets for SUMOylation could play following an ischemic event, particularly with respect to their potential neuroprotective impact in brain ischemia, is proposed.
Collapse
Affiliation(s)
- Vasco Silveirinha
- School of Pharmacy, Hopkins Building, University of Reading, Whiteknights, Reading, UK
| | | | | |
Collapse
|
23
|
Abstract
Studies on the regulation of nuclear receptors, such as the peroxisome proliferator-activated receptors (PPARs), are important to enhance our understanding of their molecular, cellular, and physiological behavior. A decade ago, it was shown that the SUMOylation pathway plays a very important role in the regulation of transcription factor activity. The SUMOylation process involves the covalent binding of SUMO protein to the target protein. However, experimental procedures to demonstrate that low-expressed proteins, such as PPARs, are SUMOylated, remain tricky, and require specific optimization for each protein.Here, we provide a simple and useful experimental method to investigate the SUMOylation of PPARs in a cellular context. The procedure for studying SUMOylation in living cells is based on the purification under denaturating conditions of total SUMOylated proteins followed by the specific detection of the PPAR proteins. For that purpose, cells are transfected with both 6xHistidine-tagged SUMO and PPAR expression vectors. Since the polyHistidine tag binds to nickel cationic ion-linked agarose matrix (Ni-NTA matrix), His-tagged SUMO proteins covalently linked to the protein substrate can be specifically precipitated and separated from the unSUMOylated proteins. The SUMO-modified PPAR proteins can subsequently be visualized by western blotting using anti-PPAR antibodies. Many questions relative to the regulation of PPAR SUMOylation can be appropriately addressed by adapting this protocol.
Collapse
Affiliation(s)
- Benoit Pourcet
- Center for Clinical Pharmacology, Division of Medicine, University College of London, London, UK
| | | | | |
Collapse
|
24
|
Yang YC, Yoshikai Y, Hsu SW, Saitoh H, Chang LK. Role of RNF4 in the ubiquitination of Rta of Epstein-Barr virus. J Biol Chem 2013; 288:12866-79. [PMID: 23504328 DOI: 10.1074/jbc.m112.413393] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epstein-Barr virus (EBV) encodes a transcription factor, Rta, which is required to activate the transcription of EBV lytic genes. This study demonstrates that treating P3HR1 cells with a proteasome inhibitor, MG132, causes the accumulation of SUMO-Rta and promotes the expression of EA-D. GST pulldown and coimmunoprecipitation studies reveal that RNF4, a RING-domain-containing ubiquitin E3 ligase, interacts with Rta. RNF4 also targets SUMO-2-conjugated Rta and promotes its ubiquitination in vitro. Additionally, SUMO interaction motifs in RNF4 are important to the ubiquitination of Rta because the RNF4 mutant with a mutation at the motifs eliminates ubiquitination. The mutation of four lysine residues on Rta that abrogated SUMO-3 conjugation to Rta also decreases the enhancement of the ubiquitination of Rta by RNF4. This finding demonstrates that RNF4 is a SUMO-targeted ubiquitin E3 ligase of Rta. Finally, knockdown of RNF4 enhances the expression of Rta and EA-D, subsequently promoting EBV lytic replication and virions production. Results of this study significantly contribute to efforts to elucidate a SUMO-targeted ubiquitin E3 ligase that regulates Rta ubiquitination to influence the lytic development of EBV.
Collapse
Affiliation(s)
- Ya-Chun Yang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | | | | | | | | |
Collapse
|
25
|
Xing X, Bi H, Chang AK, Zang MX, Wang M, Ao X, Li S, Pan H, Guo Q, Wu H. SUMOylation of AhR modulates its activity and stability through inhibiting its ubiquitination. J Cell Physiol 2012; 227:3812-9. [PMID: 22495806 DOI: 10.1002/jcp.24092] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aryl hydrocarbon receptor (AhR) is a transcription factor that belongs to the basic helix-loop-helix (bHLH) Per-Arnt-Sim homology domain (PAS) family. AhR can be activated by 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (2, 3, 7, 8-TCDD) and once activated, it promotes the abnormal expression of cytochrome P450, leading to several diseases, including cancer. In this study, we showed that AhR is subjected to post-translational modification by SUMOylation and this modification could be reversed by SENP1. Two SUMOylation sites were identified, one in the bHLH domain (K63) and the other in the TAD domain (K510) of AhR. Substitution of either K63 or K510 with arginine resulted in reduced SUMOylation for AhR. Treatment of MCF-7 cells with TCDD led to a reduced level of SUMOylated AhR in a time-dependent manner, and this occurred mainly in the nucleus. SUMOylation of AhR enhanced its stability through inhibiting its ubiquitination. Moreover, SUMOylation also repressed the transactivation activity of AhR and this could be reversed by TCDD. These results suggested that SUMOylation of AhR might play an important role in the regulation of its function, and TCDD may activate the transcriptional activity of AhR through downregulating its SUMOylation.
Collapse
Affiliation(s)
- Xinrong Xing
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kuschel A, Simon P, Tug S. Functional regulation of HIF-1α under normoxia--is there more than post-translational regulation? J Cell Physiol 2012; 227:514-24. [PMID: 21503885 DOI: 10.1002/jcp.22798] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The hypoxia-inducible factor-1 (HIF-1) is an oxygen-regulated transcriptional activator playing a pivotal role in mammalian physiology and disease pathogenesis, e.g., HIF-1 is indispensable in a broad range of developmental stages in different tumors. Its post-translational regulation via PHDs under the influence of hypoxia is widely investigated and accepted. Different non-hypoxic stimuli such as lipopolysaccharides (LPS), thrombin, and angiotensin II (Ang II), have been proven to enhance HIF-1 levels through activation of regulative mechanisms distinct from protein stabilization. Some of these stimuli specifically regulate HIF-1α at the transcriptional, post-transcriptional, or translational level, whereas others additionally influence post-translational modifications. Thus, it is difficult for the investigators to discern the impact of the different mechanisms leading to functional HIF-1 protein. Nevertheless, profound knowledge of additional regulatory networks appears to depict new therapeutic opportunities and thus is an interesting and important field for further investigations.
Collapse
Affiliation(s)
- A Kuschel
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes-Gutenberg-University Mainz, Germany
| | | | | |
Collapse
|
27
|
Huang WC, Chen ST, Chang WC, Chang KY, Chang WC, Chen BK. Involvement of aryl hydrocarbon receptor nuclear translocator in EGF-induced c-Jun/Sp1-mediated gene expression. Cell Mol Life Sci 2010; 67:3523-33. [PMID: 20508969 PMCID: PMC11115555 DOI: 10.1007/s00018-010-0392-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 04/26/2010] [Accepted: 04/27/2010] [Indexed: 11/27/2022]
Abstract
Aryl hydrocarbon receptor nuclear translocator (ARNT) binds to other basic helix-loop-helix Per/ARNT/Sim (bHLH-PAS) proteins to form functional transcriptional complexes in order to regulate specific biological pathways. Here, we report a novel mechanism that upon EGF treatment, ARNT associated with non-bHLH-PAS transcription factors, c-Jun/Sp1, and regulated gene expression, through forming a c-Jun/ARNT/Sp1 complex and binding to the Sp1 site of the gene promoter. EGF-induced promoter activity and the mRNA level of 12(S)-lipoxygenase as well as the association between c-Jun and Sp1 were reduced by ARNT knockdown. Notably, dominant negative c-Jun mutant, TAM-67, blocked ARNT-mediated 12(S)-lipoxygenase expression, demonstrating that c-Jun was responsible for the transcriptional activation. Moreover, ARNT knockdown also inhibited other EGF-induced c-Jun/Sp1 mediated gene expression, such as p21( WAF1/CIP1 ). Our results reveal a novel mechanism by which ARNT acts as a modulator to bridge the c-Jun/Sp1 interaction and plays a role in EGF-mediated gene expression under normoxic conditions.
Collapse
Affiliation(s)
- Wan-Chen Huang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701 Taiwan, ROC
| | - Shu-Ting Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701 Taiwan, ROC
| | - Wei-Chiao Chang
- Graduate Institute of Medical Genetics, Kaohsiung Medical University, 100 Tz-You First Road, Kaohsiung, 807 Taiwan, ROC
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 701 Taiwan, ROC
| | - Wen-Chang Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701 Taiwan, ROC
- Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan, 701 Taiwan, ROC
- Institute of Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701 Taiwan, ROC
| | - Ben-Kuen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701 Taiwan, ROC
- Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan, 701 Taiwan, ROC
- Institute of Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701 Taiwan, ROC
| |
Collapse
|
28
|
Pourcet B, Pineda-Torra I, Derudas B, Staels B, Glineur C. SUMOylation of human peroxisome proliferator-activated receptor alpha inhibits its trans-activity through the recruitment of the nuclear corepressor NCoR. J Biol Chem 2009; 285:5983-92. [PMID: 19955185 DOI: 10.1074/jbc.m109.078311] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is a key regulator of genes implicated in lipid homeostasis and inflammation. PPARalpha trans-activity is enhanced by recruitment of coactivators such as SRC1 and CBP/p300 and is inhibited by binding of corepressors such as NCoR and SMRT. In addition to ligand binding, PPARalpha activity is regulated by post-translational modifications such as phosphorylation and ubiquitination. In this report, we demonstrate that hPPARalpha is SUMOylated by SUMO-1 on lysine 185 in the hinge region. The E2-conjugating enzyme Ubc9 and the SUMO E3- ligase PIASy are implicated in this process. In addition, ligand treatment decreases the SUMOylation rate of hPPARalpha. Finally, our results demonstrate that SUMO-1 modification of hPPARalpha down-regulates its trans-activity through the specific recruitment of corepressor NCoR but not SMRT leading to the differential expression of a subset of PPARalpha target genes. In conclusion, hPPARalpha SUMOylation on lysine 185 down-regulates its trans-activity through the selective recruitment of NCoR.
Collapse
|
29
|
Generating specificity and diversity in the transcriptional response to hypoxia. Nat Rev Genet 2009; 10:821-32. [PMID: 19884889 DOI: 10.1038/nrg2665] [Citation(s) in RCA: 275] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The sensing of oxygen levels and maintenance of oxygen homeostasis is crucial for cells. The hypoxic-sensitive regulation of gene expression allows information about the oxygen status to be converted into appropriate cellular responses. Although there is a core transcriptional pathway, the signalling cascade can be modified to allow diversity and specificity in the transcriptional output. In this Review, we discuss recent advances in our understanding of the mechanisms and factors that contribute to the observed diversity and specificity. A deeper knowledge about how hypoxic signalling is tuned will further our understanding of the cellular hypoxic response in normal physiology and how it becomes derailed in disease.
Collapse
|
30
|
Oshima M, Mimura J, Sekine H, Okawa H, Fujii-Kuriyama Y. SUMO modification regulates the transcriptional repressor function of aryl hydrocarbon receptor repressor. J Biol Chem 2009; 284:11017-26. [PMID: 19251700 DOI: 10.1074/jbc.m808694200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) repressor (AhRR) inhibits the AhR activity. AhRR acts by competing with AhR for heterodimer formation with the AhR nuclear translocator (Arnt) and preventing the AhR.Arnt complex from binding the xenobiotic-responsive elements. Here, we report that AhRR has three evolutionarily conserved SUMOylation consensus sequences within its C-terminal repression domain and that Lys-542, Lys-583, and Lys-660 at the SUMOylation sites are modified by SUMO-1 in vivo. Arginine mutation of the three lysines results in a significant reduction of transcriptional repression activity. SUMOylation of the three lysine residues is important for the interaction between AhRR and ANKRA2, HDAC4, and HDAC5, which are important corepressors for AhRR. Arnt, a heterodimer partner for AhRR, markedly enhanced the SUMOylation of AhRR. AhRR, but not AhR, also significantly enhanced the SUMOylation of Arnt. The SUMOylation of both AhRR and Arnt is important for the efficient transcriptional repression activity of the AhRR/Arnt heterodimer.
Collapse
Affiliation(s)
- Motohiko Oshima
- Center for Tsukuba Advanced Research Alliance and Institute of Basic Medical Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8577, Japan
| | | | | | | | | |
Collapse
|
31
|
Taylor RT, Wang F, Hsu EL, Hankinson O. Roles of coactivator proteins in dioxin induction of CYP1A1 and CYP1B1 in human breast cancer cells. Toxicol Sci 2008; 107:1-8. [PMID: 18842620 DOI: 10.1093/toxsci/kfn217] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cytochrome P450 (CYP) 1A1 and CYP1B1 are inducible by 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) in the human breast cancer cell line, MCF-7. Since CYP1A1 was inducible to a much greater degree than CYP1B1, we hypothesized that there may be differences in coactivator recruitment to the promoter and/or enhancer regions of these genes. Dioxin treatment leads to recruitment of the aryl hydrocarbon receptor to the enhancer regions but not to the proximal promoter regions of both the CYP1A1 and CYP1B1 genes. On the other hand, dioxin treatment facilitated recruitment of RNA polymerase II to the promoters but not the enhancer regions. Dioxin treatment also elicited recruitment of the transcriptional coactivators, steroid receptor coactivator 1 (SRC-1) and steroid receptor coactivator 2 (SRC-2) and p300, which possess intrinsic histone acetyltranferase activities, to both genes, whereas Brahma (BRM)/Switch 2-related gene 1 (BRG-1), a subunit of nucleosomal remodeling factors, was recruited more robustly to CYP1A1 relative to CYP1B1. Small inhibitory RNA-mediated knockdown of p300 and SRC-2 adversely affected dioxin induction of both genes, whereas knockdown of BRM/BRG-1 reduced CYP1A1 induction but had little, if any, effect on CYP1B1 induction. These results suggest that nucleosomal remodeling is less significant for dioxin-mediated induction of CYP1B1 than that of CYP1A1 and may be related to the more modest inducibility of the former. Interestingly, simultaneous knockdown of SRC-2 and BRM/BRG-1 had no greater effect on CYP1A1 induction than knockdown of each coactivator individually, while simultaneous knockdown of p300 and BRM/BRG-1 had a much greater effect than knockdown of each individual gene, suggesting that the recruitment of SRC-2 to CYP1A1 depends upon BRM/BRG-1, while the recruitments of p300 and BRM/BRG-1 are independent of each other. These observations provide novel insights into the functional roles of the endogenous coactivators in dioxin induction of the human CYP1A1 and CYP1B1 genes in their natural chromosomal configurations.
Collapse
Affiliation(s)
- Robert T Taylor
- Molecular Toxicology Program, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
32
|
Yang CK, Kim JH, Ann DK, Stallcup MR. Differential regulation of the two transcriptional activation domains of the coiled-coil coactivator CoCoA by sumoylation. BMC Mol Biol 2008; 9:12. [PMID: 18218142 PMCID: PMC2263068 DOI: 10.1186/1471-2199-9-12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Accepted: 01/25/2008] [Indexed: 12/02/2022] Open
Abstract
Background The coiled-coil coactivator (CoCoA) enhances transcriptional activity of nuclear receptors, the xenobiotic aryl hydrocarbon receptor, and the lymphocyte enhancer factors (LEF) in the Wnt/β-catenin signaling pathway. CoCoA is comprised of a large central coiled coil domain flanked by N-terminal and C-terminal activation domains (AD). The N-terminal AD of CoCoA is required for coactivator function with LEF and β-catenin, while the C-terminal AD of CoCoA is required for coactivator function with nuclear receptors. We explored the role of sumoylation in regulating the activities of the two ADs and the coactivator function of CoCoA. Results The N-terminus of CoCoA is covalently modified by SUMO1 at Lys-29; both PIAS1 and ARIP3 function as E3 ligases. Fusion of SUMO1 to the N-terminus (mimicking sumoylation) reduced coactivator function of CoCoA with LEF1 and the activity of the N-terminal AD. The N- and C-termini of CoCoA can bind to each other, and C-terminal transactivation activity is attenuated in the presence of the N-terminus, indicating that the N-C interaction regulates the activity of the C-terminal AD. Fusion of SUMO1 to the N-terminal fragment of CoCoA reduced the N-C interaction and inhibition of C-terminal AD activity by the N-terminal fragment. Conclusion Sumoylation of CoCoA differentially regulates the coactivator activity of CoCoA with nuclear receptors versus LEF1, by attenuating the N-terminal AD activity and enhancing the activity of the C-terminal AD.
Collapse
Affiliation(s)
- Catherine K Yang
- Department of Biochemistry and Molecular Biology and the Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089-9176, USA.
| | | | | | | |
Collapse
|
33
|
Beischlag TV, Morales JL, Hollingshead BD, Perdew GH. The aryl hydrocarbon receptor complex and the control of gene expression. Crit Rev Eukaryot Gene Expr 2008; 18:207-50. [PMID: 18540824 PMCID: PMC2583464 DOI: 10.1615/critreveukargeneexpr.v18.i3.20] [Citation(s) in RCA: 566] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that controls the expression of a diverse set of genes. The toxicity of the potent AhR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin is almost exclusively mediated through this receptor. However, the key alterations in gene expression that mediate toxicity are poorly understood. It has been established through characterization of AhR-null mice that the AhR has a required physiological function, yet how endogenous mediators regulate this orphan receptor remains to be established. A picture as to how the AhR/ARNT heterodimer actually mediates gene transcription is starting to emerge. The AhR/ARNT complex can alter transcription both by binding to its cognate response element and through tethering to other transcription factors. In addition, many of the coregulatory proteins necessary for AhR-mediated transcription have been identified. Cross talk between the estrogen receptor and the AhR at the promoter of target genes appears to be an important mode of regulation. Inflammatory signaling pathways and the AhR also appear to be another important site of cross talk at the level of transcription. A major focus of this review is to highlight experimental efforts to characterize nonclassical mechanisms of AhR-mediated modulation of gene transcription.
Collapse
Affiliation(s)
- Timothy V. Beischlag
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - J. Luis Morales
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Brett D. Hollingshead
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gary H. Perdew
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
34
|
Hypoxia-inducible factors: Crosstalk between their protein stability and protein degradation. Cancer Lett 2007; 257:145-56. [DOI: 10.1016/j.canlet.2007.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2007] [Revised: 08/02/2007] [Accepted: 08/06/2007] [Indexed: 11/21/2022]
|
35
|
Berta MA, Mazure N, Hattab M, Pouysségur J, Brahimi-Horn MC. SUMOylation of hypoxia-inducible factor-1alpha reduces its transcriptional activity. Biochem Biophys Res Commun 2007; 360:646-52. [PMID: 17610843 DOI: 10.1016/j.bbrc.2007.06.103] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 06/19/2007] [Indexed: 12/25/2022]
Abstract
The hypoxic response of mammalian cells is controlled through a transcriptional pathway that is mediated by the hypoxia-inducible factor (HIF). Here, we show that HIF-1alpha undergoes post-translational modification by the three isoforms of the small ubiquitin-related modifier (SUMO-1, -2 and -3) in vitro in proximity to and within the oxygen-dependent degradation domain (ODDD). SUMO conjugation is promoted in vitro by the E3 SUMO ligase RanBP2/Nup538 and SUMO modification in vivo does not change HIF-1alpha turnover rate. Using cotransfection of siRNA targeted to endogenous HIF-1alpha together with HIF-1alpha siRNA-resistant expression vectors carrying mutations for SUMO modification we demonstrate increased hypoxia-response element-dependent transcriptional activity for SUMO-deficient HIF-1alpha. These results indicate that when HIF-1alpha is conjugated to SUMO its transcriptional activity is decreased and that this is not mediated by a change in the protein's half-life.
Collapse
Affiliation(s)
- Mélanie A Berta
- Institute of Signalling, Developmental Biology and Cancer Research, University of Nice, CNRS UMR 6543, Centre A. Lacassagne, 33 Avenue Valombrose, 06189 Nice, France
| | | | | | | | | |
Collapse
|
36
|
Utsubo-Kuniyoshi R, Terui Y, Mishima Y, Rokudai A, Mishima Y, Sugimura N, Kojima K, Sonoda Y, Kasahara T, Hatake K. MEK-ERK is involved in SUMO-1 foci formation on apoptosis. Cancer Sci 2007; 98:569-76. [PMID: 17284251 PMCID: PMC11158486 DOI: 10.1111/j.1349-7006.2007.00422.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Small ubiquitin-related modifier (SUMO) modification appears to regulate the activity, intracellular localization, and stability of the targeted proteins. To explore the relationship among sumoylation, antitumor reagent, and apoptosis, we treated green fluorescence protein (GFP)-SUMO-1-overexpressed K562 cells (K562/GFP-SUMO-1) with mitoxantrone (MIT) as an antitumor reagent. By the treatment with MIT, GFP-SUMO-1 formed foci in nuclei. While by the treatment with a tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA), GFP-SUMO-1 located homogeneously in nuclei. When K562/GFP-SUMO-1 cells were treated with TPA plus MIT, GFP-SUMO-1 foci became larger and apoptosis was induced more than with MIT alone. The apoptosis induced by TPA plus MIT was prevented by blockage of GFP-SUMO-1 foci by small interfering RNA (siRNA) against SUMO-1. The formation of GFP-SUMO-1 foci was reduced by a MEK inhibitor U0126 or a nuclear export inhibitor leptomycin B, and endogenous SUMO-1 foci were reduced in K562 cells expressing the dominant-negative MEK1 mutant. These results suggest that the formation of SUMO-1 foci is regulated by the MEK-ERK pathway and may induce apoptosis.
Collapse
Affiliation(s)
- Ryoko Utsubo-Kuniyoshi
- Division of Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-10-6 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Period2 (Per2) is an essential component of the mammalian clock mechanism and robust circadian expression of Per2 is essential for the maintenance of circadian rhythms. Although recent studies have shown that the circadian E2 enhancer (a non-canonical E-box) accounts for most of the circadian transcriptional drive of mPer2, little is known about the other cis-elements of mPer2 oscillatory transcription. Here, we examined the contribution of E4BP4 to Per2 mRNA oscillation in the cell-autonomous clock. Knockdown experiments of E4BP4 in both Northern blots and real-time luciferase assays suggested that endogenous E4BP4 negatively regulates Per2 mRNA oscillation. Sequence analysis revealed two putative E4BP4-binding sites (termed A-site and B-site) on mammalian Per2 promoter regions. Luciferase assays with mutant constructs showed that a novel E4BP4-binding site (B-site) is responsible for E4BP4-mediated transcriptional repression of Per2. Furthermore, chromatin immunoprecipitation assays in vivo showed that the peak of E4BP4 binding to the B-site on the Per2 promoter almost matched the trough of Per2 mRNA expression. Importantly, real-time luciferase assays showed that the B-site in addition to the E2 enhancer is required for robust circadian expression of Per2 in the cell-autonomous clock. These findings indicated that E4BP4 is required for the negative regulation of mammalian circadian clocks.
Collapse
Affiliation(s)
- Tomoya Ohno
- Clock Cell Biology, Institute for Biological Resources and Functions, National Institute of Advanced Industrial Science and Technology (AIST)Tsukuba 305-8566, Japan
- Graduate School of Life and Environmental Sciences, University of TsukubaTsukuba 305-8576, Japan
| | - Yoshiaki Onishi
- Clock Cell Biology, Institute for Biological Resources and Functions, National Institute of Advanced Industrial Science and Technology (AIST)Tsukuba 305-8566, Japan
| | - Norio Ishida
- Clock Cell Biology, Institute for Biological Resources and Functions, National Institute of Advanced Industrial Science and Technology (AIST)Tsukuba 305-8566, Japan
- Graduate School of Life and Environmental Sciences, University of TsukubaTsukuba 305-8576, Japan
- To whom correspondence should be addressed at Clock Cell Biology, National Institute of Advanced Industrial Science and Technology, Central 6-5, 1-1-1 Higashi, Tsukuba 305-8566, Japan. Tel: +81 298 61 6053; Fax: +81 298 61 9499;
| |
Collapse
|
38
|
Tsuruzoe S, Ishihara K, Uchimura Y, Watanabe S, Sekita Y, Aoto T, Saitoh H, Yuasa Y, Niwa H, Kawasuji M, Baba H, Nakao M. Inhibition of DNA binding of Sox2 by the SUMO conjugation. Biochem Biophys Res Commun 2006; 351:920-6. [PMID: 17097055 DOI: 10.1016/j.bbrc.2006.10.130] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Accepted: 10/25/2006] [Indexed: 01/03/2023]
Abstract
Sox2 is a member of the high mobility group (HMG) domain DNA-binding proteins for transcriptional control and chromatin architecture. The HMG domain of Sox2 binds the DNA to facilitate transactivation by the cooperative transcription factors such as Oct3/4. We report that mouse Sox2 is modified by SUMO at lysine 247. Substitution of the target lysine to arginine lost the sumoylation but little affected transcriptional potential or nuclear localization of Sox2. By contrast with the unmodified form, Sox2 fused to SUMO-1 did not augment transcription via the Fgf4 enhancer in the presence of Oct3/4. Further, SUMO-1-conjugated Sox2 at the lysine 247 or at the carboxyl terminus reduced the binding to the Fgf4 enhancer. These indicate that Sox2 sumoylation negatively regulates its transcriptional role through impairing the DNA binding.
Collapse
Affiliation(s)
- Shu Tsuruzoe
- Department of Regeneration Medicine, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cheng J, Bawa T, Lee P, Gong L, Yeh ETH. Role of desumoylation in the development of prostate cancer. Neoplasia 2006; 8:667-76. [PMID: 16925949 PMCID: PMC1601940 DOI: 10.1593/neo.06445] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
SUMO is a novel ubiquitin-like protein that can covalently modify a large number of nuclear proteins. SUMO modification has emerged as an important regulatory mechanism for protein function and localization. Sumoylation is a dynamic process that is mediated by activating (E1), conjugating (E2), and ligating (E3) enzymes and is readily reversed by a family of SUMO-specific proteases (SENPs). Since SUMO was discovered 10 years ago, the biologic contribution of this posttranslational modification has remained unclear. In this review, we report that SENP1, a member of the SENP family, is overexpressed in human prostate cancer specimens. The induction of SENP1 is observed with the chronic exposure of prostate cancer cells to androgen and/or interleukin (IL) 6. SENP1 upregulation modulates the transcriptional activity of androgen receptors (ARs) and c-Jun, as well as cyclin D1 expression. Initial in vivo data from transgenic mice indicate that overexpression of SENP1 in the prostate leads to the development of prostatic intraepithelial neoplasia at an early age. Collectively, these studies indicate that overexpression of SENP1 is associated with prostate cancer development.
Collapse
Affiliation(s)
- Jinke Cheng
- Department of Cardiology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
40
|
O'Donnell JL, Joyce MR, Shannon AM, Harmey J, Geraghty J, Bouchier-Hayes D. Oncological implications of hypoxia inducible factor-1alpha (HIF-1alpha) expression. Cancer Treat Rev 2006; 32:407-16. [PMID: 16889900 DOI: 10.1016/j.ctrv.2006.05.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 05/01/2006] [Accepted: 05/03/2006] [Indexed: 01/23/2023]
Abstract
Solid tumours contain regions of hypoxia, which may be a prognostic indicator and determinant of malignant progression, metastatic development and chemoradio-resistance. The degree of intra-tumoural hypoxia has been shown to be positively correlated with the expression of the transcription factor hypoxia-inducible factor 1. HIF-1 is composed of 2 sub-units, namely HIF-1alpha and HIF-1beta. The production of hypoxia inducible factor 1-alpha has been identified as a key element in allowing cells to adapt and survive in a hostile hypoxic environment via a variety of pathways. HIF-1alpha is stabilised by hypoxia at the protein level, and also by the oncogenes HER2neu, v-src and ras. There are over 60 target genes for HIF-1, many of which are activated in cancers in comparison to equivalent normal tissues. Chemotherapeutic modulation of HIF-1 pathways has shown promise for patients with chemo-radio resistant or recurrent tumours in Phase II clinical trials. We herein review the existing literature on hypoxia inducible factor-1alpha, particularly its role in carcinogenesis and clinical implications of its over-expression.
Collapse
Affiliation(s)
- Jill L O'Donnell
- RCSI Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
41
|
Saitoh N, Uchimura Y, Tachibana T, Sugahara S, Saitoh H, Nakao M. In situ SUMOylation analysis reveals a modulatory role of RanBP2 in the nuclear rim and PML bodies. Exp Cell Res 2006; 312:1418-30. [PMID: 16688858 DOI: 10.1016/j.yexcr.2006.01.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
SUMO modification plays a critical role in a number of cellular functions including nucleocytoplasmic transport, gene expression, cell cycle and formation of subnuclear structures such as promyelocytic leukemia (PML) bodies. In order to identify the sites where SUMOylation takes place in the cell, we developed an in situ SUMOylation assay using a semi-intact cell system and subsequently combined it with siRNA-based knockdown of nucleoporin RanBP2, also known as Nup358, which is one of the known SUMO E3 proteins. With the in situ SUMOylation assay, we found that both nuclear rim and PML bodies, besides mitotic apparatuses, are major targets for active SUMOylation. The ability to analyze possible SUMO conjugation sites would be a valuable tool to investigate where SUMO E3-like activities and/or SUMO substrates exist in the cell. Specific knockdown of RanBP2 completely abolished SUMOylation along the nuclear rim and dislocated RanGAP1 from the nuclear pore complexes. Interestingly, the loss of RanBP2 markedly reduced the number of PML bodies, in contrast to other, normal-appearing nuclear compartments including the nuclear lamina, nucleolus and chromatin, suggesting a novel link between RanBP2 and PML bodies. SUMOylation facilitated by RanBP2 at the nuclear rim may be a key step for the formation of a particular subnuclear organization. Our data imply that SUMO E3 proteins like RanBP2 facilitate spatio-temporal SUMOylation for certain nuclear structure and function.
Collapse
Affiliation(s)
- Noriko Saitoh
- Department of Regeneration Medicine, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumanmoto 860-0811, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Yamaguchi T, Sharma P, Athanasiou M, Kumar A, Yamada S, Kuehn MR. Mutation of SENP1/SuPr-2 reveals an essential role for desumoylation in mouse development. Mol Cell Biol 2005; 25:5171-82. [PMID: 15923632 PMCID: PMC1140580 DOI: 10.1128/mcb.25.12.5171-5182.2005] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The covalent modification of proteins by the small ubiquitin-like protein SUMO has been implicated in the regulation of numerous biological processes, including nucleocytoplasmic transport, genomic stability, and gene transcription. Sumoylation occurs by a multienzyme process similar to ubiquitination and, in Saccharomyces cerevisiae, is reversed by desumoylating enzymes encoded by the Ulp1 and Smt4/Ulp2 genes. The physiological importance of desumoylation has been revealed by mutations in either gene, which lead to nonoverlapping defects in cell cycle transition and meiosis. Several mammalian Ulp homologues have been identified, but, to date, nothing is known of the phenotypic effects of their loss of function. Here, we describe a random retroviral insertional mutation of one homolog, mouse SENP1/SuPr-2. The mutation causes increased steady-state levels of the sumoylated forms of a number of proteins and results in placental abnormalities incompatible with embryonic development. Our findings provide the first insight into the critical importance of regulating sumoylation in mammals.
Collapse
Affiliation(s)
- Taihei Yamaguchi
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, NCI-Frederick, Bldg. 560, Rm. 12-90, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
43
|
Oh S, Im H, Oh E, Lee J, Khim JY, Mun J, Kim Y, Lee E, Kim J, Sul D. Effects of benzo(a)pyrene on protein expression in Jurkat T-cells. Proteomics 2005; 4:3514-26. [PMID: 15529408 DOI: 10.1002/pmic.200400981] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental pollutants of air, water and soil, and are produced by the incomplete combustion of organic materials. The International Agency for Research on Cancer has characterized PAHs as carcinogens. In this study, we investigated the effects of benzo(a)pyrene (B(a)P), which is the most carcinogenic member of the PAHs, on Jurkat cell protein by proteomic analysis. Jurkat cells were treated with various concentrations of B(a)P (0, 2.5, 5, 10, 20 or 40 microM) for 24 or 48 h and 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and lactate dehydrogenase assays were carried out to determine cytotoxicity and a Comet assay was used to determinate genotoxicity. The cytotoxicity assays showed that 2.5 microM of B(a)P was the maximal concentration that did not cause any toxicity, but nevertheless, at this level B(a)P produced significant DNA damage in Jurkat cells at 48 h. Proteomic analysis using three different pI ranges and large two-dimensional gel electrophoresis showed 3427 protein spots. A total of 46 (13 up- and 33 down-regulated) proteins were identified as biomarkers of B(a)P and showed dose-dependent expressions in Jurkat T-cell line exposed to B(a)P. Of these, 27 protein spots were identified by matrix-assisted laser desorption/ionization-time of flight mass spectrometry. Two functionally differentiated protein groups were found. The protein group involving apoptosis and tumor suppression were found to be up-regulated, and B(a)P down-regulated enzyme was involved in energy metabolism, DNA synthesis and in cell structure and motility.
Collapse
Affiliation(s)
- Sangnam Oh
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
SUMO (small ubiquitin-related modifier) family proteins are not only structurally but also mechanistically related to ubiquitin in that they are posttranslationally attached to other proteins. As ubiquitin, SUMO is covalently linked to its substrates via amide (isopeptide) bonds formed between its C-terminal glycine residue and the epsilon-amino group of internal lysine residues. The enzymes involved in the reversible conjugation of SUMO are similar to those mediating the ubiquitin conjugation. Since its discovery in 1996, SUMO has received a high degree of attention because of its intriguing and essential functions, and because its substrates include a variety of biomedically important proteins such as tumor suppressor p53, c-jun, PML and huntingtin. SUMO modification appears to play important roles in diverse processes such as chromosome segregation and cell division, DNA replication and repair, nuclear protein import, protein targeting to and formation of certain subnuclear structures, and the regulation of a variety of processes including the inflammatory response in mammals and the regulation of flowering time in plants.
Collapse
Affiliation(s)
- R Jürgen Dohmen
- Institute for Genetics, University of Cologne, Zülpicher Str. 47, D-50674 Cologne, Germany.
| |
Collapse
|
45
|
Brahimi-Horn MC, Pouysségur J. The hypoxia-inducible factor and tumor progression along the angiogenic pathway. ACTA ACUST UNITED AC 2005; 242:157-213. [PMID: 15598469 DOI: 10.1016/s0074-7696(04)42004-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The hypoxia-inducible factor (HIF) is a transcription factor that plays a key role in the response of cells to oxygen levels. HIF is a heterodimer of alpha- and beta-subunits where the alpha-subunit is translated constitutively but has a very short half-life under normal oxygen concentrations. Negative regulation of the half-life and activity of the alpha-subunit is dependent on its posttranslational hydroxylation by hydroxylases that are dependent on oxygen for activity. Thus under low oxygen (hypoxic) conditions the hydroxylases are inactive and the alpha-subunit is stable and able to interact with the beta-subunit to bind and induce transcription of target genes. Hypoxic conditions are encountered in development and in disease states such as cancer. Tumors that have outstripped their blood supply become hypoxic and express high levels of HIF. HIF in turn targets genes that induce survival, glycolysis, and angiogenesis, a form of neovascularization, which ensures the tumor with a continued supply of oxygen and nutrients for further growth.
Collapse
Affiliation(s)
- M Christiane Brahimi-Horn
- Institute of Signaling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre A. Lacassagne, 06189 Nice, France
| | | |
Collapse
|
46
|
Brahimi-Horn C, Pouysségur J. When hypoxia signalling meets the ubiquitin-proteasomal pathway, new targets for cancer therapy. Crit Rev Oncol Hematol 2005; 53:115-23. [PMID: 15661562 DOI: 10.1016/j.critrevonc.2004.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2004] [Indexed: 11/17/2022] Open
Abstract
The ubiquitin-proteasomal pathway of degradation of proteins is activated or repressed in response to a number of environmental stresses and thereby plays an essential role in cell function and survival. Hypoxic stress, resulting from a decrease in the concentration of oxygen in tissues, is encountered in both physiological and pathological situations, in particular in cancer. The transcriptional complex hypoxia-inducible factor (HIF) is the key player in the signalling pathway that controls the hypoxic response of mammalian cells. Under hypoxic conditions it transactivates an impressive number of genes involved in a multitude of cellular functions. Tight regulation of this response in part involves the ubiquitin-proteasomal system where oxygen-dependent prolyl-4-hydroxylation of the alpha subunit of HIF triggers a cascade of events that leads to its degradation by the 26S proteasome. Inhibition of the proteasome in conjunction with topoisomerase inhibition has shown some promise in the treatment of experimental cancer. Such treatment may impact on the hypoxic adaptation of tumour cells.
Collapse
Affiliation(s)
- Christiane Brahimi-Horn
- Institute of Signaling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre A. Lacassagne, 33 Avenue Valombrose, 06189 Nice, France.
| | | |
Collapse
|
47
|
Role of coactivators in transcriptional activation by the aryl hydrocarbon receptor. Arch Biochem Biophys 2005; 433:379-86. [PMID: 15581594 DOI: 10.1016/j.abb.2004.09.031] [Citation(s) in RCA: 229] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Revised: 09/24/2004] [Indexed: 11/29/2022]
Abstract
The aryl hydrocarbon receptor (AHR) mediates the carcinogenic and other toxic effects of a variety of environmental pollutants, including 2,37,8-tetrachlorodibenzo-p-dioxin (TCDD), and some polycyclic aromatic hydrocarbons (PAHs). In most if not all cases, these deleterious effects depend upon modulation of gene transcription effected by the ligand-bound AHR. The responsive genes required for toxicity of TCDD have yet to be defined. However, induction of Cyp1a1 is known to represent a significant event in the toxicity of PAHs. Furthermore, the Cyp1a1 gene provides a model system for studying the mechanism of gene transcription by AHR. This review discusses the roles of transcriptional coactivator proteins in induction of Cyp1a1 by AHR ligands. Coactivators physically associate with the gene upon induction, and provide a bridge between AHR molecules, located at 5'enhancer elements, and general transcription factors, located at the promoter of the gene. Studies on the endogenous Cyp1a1 gene in its natural chromosomal setting are emphasized. The recent development of several new experimental techniques including the chromatin immunoprecipitation (ChIP) assay, RNA interference, and real-time PCR has provided a major boost to such studies. Future directions for research are also discussed. Since variations in coactivator expression or activity may result in inter-individual differences in response to AHR ligands, and may also underlie tissue-specific differences in sensitivity to such ligands during development, and in adulthood, the role of coactivators in transcriptional activation by AHR constitutes a very important area of research.
Collapse
|
48
|
Brahimi-Horn C, Mazure N, Pouysségur J. Signalling via the hypoxia-inducible factor-1α requires multiple posttranslational modifications. Cell Signal 2005; 17:1-9. [PMID: 15451019 DOI: 10.1016/j.cellsig.2004.04.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Accepted: 04/24/2004] [Indexed: 11/15/2022]
Abstract
Cellular hypoxia, a local decrease in the oxygen concentration below normal (21%) atmospheric concentrations, occurs in both physiological and pathological situations. The transcriptional complex Hypoxia-Inducible Factor-1 (HIF-1) is the key player in the signalling pathway that controls the hypoxic response of mammalian cells. Tight regulation of this response involves posttranslational modification of the alpha subunit of HIF-1. Hydroxylation, ubiquitination, acetylation, S-nitrosation and phosphorylation have been shown to determine its half-life and/or transcriptional activity. The precise spatio-temporal occurrence of these multiple modifications is still not fully understood but is dependent on the microenvironment and determines the driving force of variable cellular responses.
Collapse
Affiliation(s)
- Christiane Brahimi-Horn
- Institute of Signaling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre A. Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France.
| | | | | |
Collapse
|
49
|
Abstract
The small ubiquitin-like modifier (SUMO) is covalently attached to lysine residues in target proteins and in doing so changes the properties of the modified protein. Here we examine the role of SUMO modification in transcriptional regulation. SUMO addition to components of the transcriptional apparatus does not have a common consequence as it can both activate and repress transcription. In most cases, however, SUMO modification of transcription factors leads to repression and various models to explain this, ranging from retention in nuclear bodies to recruitment of histone deacetylases are discussed.
Collapse
Affiliation(s)
- David W H Girdwood
- Centre for Biomolecular Sciences, School of Biology, University of St. Andrews, North Haugh, St Andrews KY16 9ST, UK
| | | | | |
Collapse
|
50
|
Floyd ZE, Stephens JM. Control of peroxisome proliferator-activated receptor gamma2 stability and activity by SUMOylation. ACTA ACUST UNITED AC 2004; 12:921-8. [PMID: 15229330 DOI: 10.1038/oby.2004.112] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine whether small ubiquitin-related modifier (SUMO)ylation of lysine 107 plays a role in regulating the activity of peroxisome proliferator-activated receptor gamma (PPARgamma). RESEARCH METHODS AND PROCEDURES Transient expression of wild-type and K107R-PPARgamma2 in the NIH 3T3 fibroblast cell line was carried out in conjunction with half-life studies, luciferase activity assays, and indirect immunofluorescence localization studies. Additional in vitro analysis was carried out using recombinant SUMOylation pathway proteins along with in vitro transcribed and translated wild-type or K107R-PPARgamma2 to examine the SUMO-1 modification state of wild-type and SUMO-deficient K107R-PPARgamma2. RESULTS While examining PPARgamma2 for potential ubiquitylation sites, we identified a strong consensus site for SUMO modification that contains lysine 107. In vitro, SUMOylation studies showed that lysine 107 of PPARgamma2 is a major SUMOylation site and that at least one other SUMOylation site is present in PPARgamma. In addition, our results demonstrated that SUMO-1 affects PPARgamma stability and transcriptional activity but not the nuclear localization of PPARgamma. DISCUSSION These results indicated that SUMOylation plays a role in regulating PPARgamma, both indirectly and directly by modification of lysine 107. Because PPARgamma is regulated in numerous animal models of obesity, understanding the covalent modifications of PPARgamma may enhance our understanding of the metabolic syndrome.
Collapse
Affiliation(s)
- Z Elizabeth Floyd
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| | | |
Collapse
|