1
|
Rampioni Vinciguerra GL, Capece M, Scafetta G, Rentsch S, Vecchione A, Lovat F, Croce CM. Role of Fra-2 in cancer. Cell Death Differ 2024; 31:136-149. [PMID: 38104183 PMCID: PMC10850073 DOI: 10.1038/s41418-023-01248-4] [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: 08/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
Fos-related antigen-2 (Fra-2) is the most recently discovered member of the Fos family and, by dimerizing with Jun proteins, forms the activator protein 1 (AP-1) transcription factor. By inducing or repressing the transcription of several target genes, Fra-2 is critically involved in the modulation of cell response to a variety of extracellular stimuli, stressors and intracellular changes. In physiological conditions, Fra-2 has been found to be ubiquitously expressed in human cells, regulating differentiation and homeostasis of bone, muscle, nervous, lymphoid and other tissues. While other AP-1 members, like Jun and Fos, are well characterized, studies of Fra-2 functions in cancer are still at an early stage. Due to the lack of a trans-activating domain, which is present in other Fos proteins, it has been suggested that Fra-2 might inhibit cell transformation, eventually exerting an anti-tumor effect. In human malignancies, however, Fra-2 activity is enhanced (or induced) by dysregulation of microRNAs, oncogenes and extracellular signaling, suggesting a multifaceted role. Therefore, Fra-2 can promote or prevent transformation, proliferation, migration, epithelial-mesenchymal transition, drug resistance and metastasis formation in a tumor- and context-dependent manner. Intriguingly, recent data reports that Fra-2 is also expressed in cancer associated cells, contributing to the intricate crosstalk between neoplastic and non-neoplastic cells, that leads to the evolution and remodeling of the tumor microenvironment. In this review we summarize three decades of research on Fra-2, focusing on its oncogenic and anti-oncogenic effects in tumor progression and dissemination.
Collapse
Affiliation(s)
- Gian Luca Rampioni Vinciguerra
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Marina Capece
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Giorgia Scafetta
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Sydney Rentsch
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Francesca Lovat
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Chen Y, Toth R, Chocarro S, Weichenhan D, Hey J, Lutsik P, Sawall S, Stathopoulos GT, Plass C, Sotillo R. Club cells employ regeneration mechanisms during lung tumorigenesis. Nat Commun 2022; 13:4557. [PMID: 35931677 PMCID: PMC9356049 DOI: 10.1038/s41467-022-32052-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 07/12/2022] [Indexed: 11/09/2022] Open
Abstract
The high plasticity of lung epithelial cells, has for many years, confounded the correct identification of the cell-of-origin of lung adenocarcinoma (LUAD), one of the deadliest malignancies worldwide. Here, we employ lineage-tracing mouse models to investigate the cell of origin of Eml4-Alk LUAD, and show that Club and Alveolar type 2 (AT2) cells give rise to tumours. We focus on Club cell originated tumours and find that Club cells experience an epigenetic switch by which they lose their lineage fidelity and gain an AT2-like phenotype after oncogenic transformation. Single-cell transcriptomic analyses identified two trajectories of Club cell evolution which are similar to the ones used during lung regeneration, suggesting that lung epithelial cells leverage on their plasticity and intrinsic regeneration mechanisms to give rise to a tumour. Together, this study highlights the role of Club cells in LUAD initiation, identifies the mechanism of Club cell lineage infidelity, confirms the presence of these features in human tumours, and unveils key mechanisms conferring LUAD heterogeneity.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Reka Toth
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Bioinformatics Platform, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Sara Chocarro
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Joschka Hey
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Pavlo Lutsik
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-Ray Imaging and CT, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU), Max-Lebsche-Platz 31, 81377, Munich, Bavaria, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Rocio Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,German Center for Lung Research (DZL), Heidelberg, Germany. .,Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany. .,German Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany.
| |
Collapse
|
3
|
Wu Z, Nicoll M, Ingham RJ. AP-1 family transcription factors: a diverse family of proteins that regulate varied cellular activities in classical hodgkin lymphoma and ALK+ ALCL. Exp Hematol Oncol 2021; 10:4. [PMID: 33413671 PMCID: PMC7792353 DOI: 10.1186/s40164-020-00197-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/07/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) and anaplastic lymphoma kinase-positive, anaplastic large cell lymphoma (ALK+ ALCL) are B and T cell lymphomas respectively, which express the tumour necrosis factor receptor superfamily member, CD30. Another feature shared by cHL and ALK+ ALCL is the aberrant expression of multiple members of the activator protein-1 (AP-1) family of transcription factors which includes proteins of the Jun, Fos, ATF, and Maf subfamilies. In this review, we highlight the varied roles these proteins play in the pathobiology of these lymphomas including promoting proliferation, suppressing apoptosis, and evading the host immune response. In addition, we discuss factors contributing to the elevated expression of these transcription factors in cHL and ALK+ ALCL. Finally, we examine therapeutic strategies for these lymphomas that exploit AP-1 transcriptional targets or the signalling pathways they regulate.
Collapse
Affiliation(s)
- Zuoqiao Wu
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.17063.330000 0001 2157 2938Present Address: Department of Medicine, University of Toronto, Toronto, Canada
| | - Mary Nicoll
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.14709.3b0000 0004 1936 8649Present Address: Department of Biology, McGill University, Montreal, Canada
| | - Robert J. Ingham
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| |
Collapse
|
4
|
Papavassiliou AG, Musti AM. The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion. Cells 2020; 9:cells9112470. [PMID: 33202877 PMCID: PMC7697663 DOI: 10.3390/cells9112470] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
c-Jun is a major component of the dimeric transcription factor activator protein-1 (AP-1), a paradigm for transcriptional response to extracellular signaling, whose components are basic-Leucine Zipper (bZIP) transcription factors of the Jun, Fos, activating transcription factor (ATF), ATF-like (BATF) and Jun dimerization protein 2 (JDP2) gene families. Extracellular signals regulate c-Jun/AP-1 activity at multiple levels, including transcriptional and posttranscriptional regulation of c-Jun expression and transactivity, in turn, establishing the magnitude and the duration of c-Jun/AP-1 activation. Another important level of c-Jun/AP-1 regulation is due to the capability of Jun family members to bind DNA as a heterodimer with every other member of the AP-1 family, and to interact with other classes of transcription factors, thereby acquiring the potential to integrate diverse extrinsic and intrinsic signals into combinatorial regulation of gene expression. Here, we review how these features of c-Jun/AP-1 regulation underlie the multifaceted output of c-Jun biological activity, eliciting quite distinct cellular responses, such as neoplastic transformation, differentiation and apoptosis, in different cell types. In particular, we focus on the current understanding of the role of c-Jun/AP-1 in the response of CD8 T cells to acute infection and cancer. We highlight the transcriptional and epigenetic regulatory mechanisms through which c-Jun/AP-1 participates in the productive immune response of CD8 T cells, and how its downregulation may contribute to the dysfunctional state of tumor infiltrating CD8 T cells. Additionally, we discuss recent insights pointing at c-Jun as a suitable target for immunotherapy-based combination approaches to reinvigorate anti-tumor immune functions.
Collapse
Affiliation(s)
- Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Anna Maria Musti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Correspondence: ; Tel.: +39-3337543732
| |
Collapse
|
5
|
Nijhawan P, Behl T. The Role of Endostatin in Rheumatoid Arthritis. Curr Rheumatol Rev 2020; 17:68-75. [PMID: 32348230 DOI: 10.2174/1573397115666191127141801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/19/2020] [Accepted: 04/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Endostatin by its therapeutic value against rheumatoid arthritis has recently gained significant interest in biomedical science. A recent study revealed that various approaches have been made to prevent rheumatoid arthritis by either controlling or inhibiting the progression of angiogenesis. OBJECTIVE The main objective of the current manuscript is to enumerate the intrinsic role of endostatin in rheumatoid arthritis. METHODS A thorough and detailed review of literature from the papers published from the year 1997-2019 was studied for the preparation of the current article. RESULTS Endostatin is one such agent of the subfamily of ECM called as multiplexins obtained from proteolytic cleavage of XVIII and its carboxylic terminal fragments and is known for its antiangiogenic and antiproliferative property. The exact mechanism of endostatin is still unclear, but it acts by downregulating or inhibiting the responses of various factors, including Id1, Id3, matrix metalloproteinase, and Nuclear factor Kappa B that are liable for angiogenesis. The mutual effects on adipogenesis and angiogenesis, endostatin inhibits dietary-induced obesity and its related metabolic disorders, such as insulin resistance, glucose intolerance, and hepatic steatosis. CONCLUSION The present review demonstrates the intrinsic usage of endostatin as a novel molecule in rheumatoid arthritis. It focuses on the status of the therapeutic potential of endostatin in inhibiting the activity of angiogenesis is also very well explored.
Collapse
Affiliation(s)
- Priya Nijhawan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
6
|
Hampton MB, Vick KA, Skoko JJ, Neumann CA. Peroxiredoxin Involvement in the Initiation and Progression of Human Cancer. Antioxid Redox Signal 2018; 28:591-608. [PMID: 29237274 PMCID: PMC9836708 DOI: 10.1089/ars.2017.7422] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
SIGNIFICANCE It has been proposed that cancer cells are heavily dependent on their antioxidant defenses for survival and growth. Peroxiredoxins are a family of abundant thiol-dependent peroxidases that break down hydrogen peroxide, and they have a central role in the maintenance and response of cells to alterations in redox homeostasis. As such, they are potential targets for disrupting tumor growth. Recent Advances: Genetic disruption of peroxiredoxin expression in mice leads to an increased incidence of neoplastic disease, consistent with a role for peroxiredoxins in protecting genomic integrity. In contrast, many human tumors display increased levels of peroxiredoxin expression, suggesting that strengthened antioxidant defenses provide a survival advantage for tumor progression. Peroxiredoxin inhibitors are being developed and explored as therapeutic agents in different cancer models. CRITICAL ISSUES It is important to complement peroxiredoxin knockout and expression studies with an improved understanding of the biological function of the peroxiredoxins. Although current results can be interpreted within the context that peroxiredoxins scavenge hydroperoxides, some peroxiredoxin family members appear to have more complex roles in regulating the response of cells to oxidative stress through protein interactions with constituents of other signaling pathways. FUTURE DIRECTIONS Further mechanistic information is required for understanding the role of oxidative stress in cancer, the function of peroxiredoxins in normal versus cancer cells, and for the design and testing of specific peroxiredoxin inhibitors that display selectivity to malignant cells. Antioxid. Redox Signal. 28, 591-608.
Collapse
Affiliation(s)
- Mark B Hampton
- 1 Department of Pathology, Centre for Free Radical Research, University of Otago , Christchurch, Christchurch, New Zealand
| | - Kate A Vick
- 1 Department of Pathology, Centre for Free Radical Research, University of Otago , Christchurch, Christchurch, New Zealand
| | - John J Skoko
- 2 Womens Cancer Research Center, University of Pittsburgh Cancer Center , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Carola A Neumann
- 2 Womens Cancer Research Center, University of Pittsburgh Cancer Center , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
7
|
Sha Z, Zhu X, Li N, Li Y, Li D. Proto-oncogenic miR-744 is upregulated by transcription factor c-Jun via a promoter activation mechanism. Oncotarget 2018; 7:64977-64986. [PMID: 27533465 PMCID: PMC5323131 DOI: 10.18632/oncotarget.11285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/08/2016] [Indexed: 11/25/2022] Open
Abstract
Upregulation of miR-744 is associated with poor prognosis in many types of cancer patients, but it is still unclear how miR-744 becomes elevated in these tumors. In this study, we found that ectopic c-Jun elevated miR-744 expression, whereas c-Jun attenuation reduced miR-744 expression. Chromatin immunoprecipitation assay confirmed the direct binding of c-Jun to the promoter of miR-744. The binding site of −343 to −349 bp within the most potential promoter like sequence of miR-744 was further validated by luciferase reporter gene assays. C-Jun-induced miR-744 upregulation could significantly promote migration and invasion of nasopharyngeal carcinoma cells and non-small cell lung cancer (NSCLC) cells, hence ectopic c-Jun was sufficient to rescue the migratory and invasive ability of these cells when miR-744 was knockdown. Additionally, a positive correlation between the expression levels of miR-744 and c-Jun was revealed in NSCLC samples with high (top 10%) level of miR-744 expression from the TCGA dataset. Taken together, our results demonstrated for the first time the regulatory mechanism of miR-744 transcription by c-Jun, providing a potential mechanism underlying the upregulation of miR-744 in cancers.
Collapse
Affiliation(s)
- Zhou Sha
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoxia Zhu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Na Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiyi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dianhe Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
8
|
Liu C, Qiu H, Yu M, Wang Z, Yuan Y, Jiang Z, Shao X, Hua D, Liu M, Wu S. c-Jun-mediated β-1,3-N-acetylglucosaminyltransferase 8 expression: A novel mechanism regulating the invasion and metastasis of colorectal carcinoma cells. Oncol Lett 2017; 14:3722-3728. [PMID: 28927138 DOI: 10.3892/ol.2017.6624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 02/03/2017] [Indexed: 11/05/2022] Open
Abstract
β-1,3-N-Acetylglucosaminyltransferase 8 (β3GnT8) is a key enzyme that catalyzes the formation of polylactosamine glycan structures by transferring GlcNAc to tetra-antennary β1-6-branched N-glycans, and it has been reported to participate in tumor invasion and metastasis by regulating the expression of matrix metalloproteinases (MMPs), cluster of differentiation 147 (CD147) and polylactosamine. By contrast, the role of transcription factor c-Jun in cell cycle progression has been well established. c-Jun has an important role in tumor cell invasion and metastasis. However, the precise molecular mechanisms by which c-Jun regulates these processes in colorectal carcinoma cells are not fully elucidated. In the present study, c-Jun had a significant effect on the invasive and migratory abilities of SW480 and LoVo cells. Additionally, overexpression of c-Jun was able to increase the expression of β3GnT8, MMPs, CD147 and polylactosamine. Similarly, knockdown of c-Jun was able to decrease the expression of β3GnT8, MMPs, CD147 and polylactosamine. These results suggest that c-Jun is able to regulate colorectal carcinoma cell invasion and metastasis via β3GnT8. A chromatin immunoprecipitation assay indicated that c-Jun is able to bind directly to the promoter regions of β3GnT8 in SW480 and LoVo cells. This leads to transcriptional activation of β3GnT8, which in turn regulates the expression of tumor invasion and metastasis-associated genes. The results of the present study demonstrate a novel mechanism underlying colorectal carcinoma cell invasion and metastasis, where β3GnT8 is transcriptionally activated via c-Jun binding to its promoter.
Collapse
Affiliation(s)
- Chunliang Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Hao Qiu
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Meiyun Yu
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Zerong Wang
- The Fifth People's Hospital of Suzhou, Suzhou, Jiangsu 215007, P.R. China
| | - Yaqin Yuan
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Zhi Jiang
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xuejun Shao
- The Affiliated Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Dong Hua
- The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Min Liu
- Suzhou Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu 215009, P.R. China
| | - Shiliang Wu
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
9
|
Gào X, Schöttker B. Reduction-oxidation pathways involved in cancer development: a systematic review of literature reviews. Oncotarget 2017; 8:51888-51906. [PMID: 28881698 PMCID: PMC5584299 DOI: 10.18632/oncotarget.17128] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/03/2017] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress results from an imbalance of the reactive oxygen species/reactive nitrogen species (ROS/RNS) production and the oxidants defense system. Extensive research during the last decades has revealed that oxidative stress can mediate cancer initiation and development by leading not only to molecular damage but also to a disruption of reduction-oxidation (redox) signaling. In order to provide a global overview of the redox signaling pathways, which play a role in cancer formation, we conducted a systematic literature search in PubMed and ISI Web of Science and identified 185 relevant reviews published in the last 10 years. The 20 most frequently described pathways were selected to be presented in this systematic review and could be categorized into 3 groups: Intracellular ROS/RNS generating organelles and enzymes, signal transduction cascades kinases/phosphatases and transcription factors. Intracellular ROS/RNS generation organelles are mitochondria, endoplasmic reticulum and peroxisomes. Enzymes, including NOX, COX, LOX and NOS, are the most prominent enzymes generating ROS/RNS. ROS/RNS act as redox messengers of transmembrane receptors and trigger the activation or inhibition of signal transduction kinases/phosphatases, such as the family members of protein tyrosine kinases and protein tyrosine phosphatases. Furthermore, these reactions activate downstream signaling pathways including protein kinase of the MAPK cascade, PI3K and PKC. The kinases and phosphatases regulate the phosphorylation status of transcription factors including APE1/Ref-1, HIF-1α, AP-1, Nrf2, NF-κB, p53, FOXO, STAT, and β-catenin. Finally, we briefly discuss cancer prevention and treatment opportunities, which address redox pathways and further research needs.
Collapse
Affiliation(s)
- Xīn Gào
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,Network Aging Research, University of Heidelberg, Heidelberg, Germany.,Institute of Health Care and Social Sciences, FOM University, Essen, Germany
| |
Collapse
|
10
|
Lee HL, Park MH, Son DJ, Song HS, Kim JH, Ko SC, Song MJ, Lee WH, Yoon JH, Ham YW, Han SB, Hong JT. Anti-cancer effect of snake venom toxin through down regulation of AP-1 mediated PRDX6 expression. Oncotarget 2016; 6:22139-51. [PMID: 26061816 PMCID: PMC4673152 DOI: 10.18632/oncotarget.4192] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 05/20/2015] [Indexed: 11/25/2022] Open
Abstract
Snake venom toxin (SVT) from Vipera lebetina turanica contains a mixture of different enzymes and proteins. Peroxiredoxin 6 (PRDX6) is known to be a stimulator of lung cancer cell growth. PRDX6 is a member of peroxidases, and has calcium-independent phospholipase A2 (iPLA2) activities. PRDX6 has an AP-1 binding site in its promoter region of the gene. Since AP-1 is implicated in tumor growth and PRDX6 expression, in the present study, we investigated whether SVT inhibits PRDX6, thereby preventing human lung cancer cell growth (A549 and NCI-H460) through inactivation of AP-1. A docking model study and pull down assay showed that SVT completely fits on the basic leucine zipper (bZIP) region of c-Fos of AP-1. SVT (0–10 μg/ml) inhibited lung cancer cell growth in a concentration dependent manner through induction of apoptotic cell death accompanied by induction of cleaved caspase-3, -8, -9, Bax, p21 and p53, but decreased cIAP and Bcl2 expression via inactivation of AP-1. In an xenograft in vivo model, SVT (0.5 mg/kg and 1 mg/kg) also inhibited tumor growth accompanied with the reduction of PRDX6 expression, but increased expression of proapoptotic proteins. These data indicate that SVT inhibits tumor growth via inhibition of PRDX6 activity through interaction with its transcription factor AP-1.
Collapse
Affiliation(s)
- Hye Lim Lee
- College of Pharmacy, Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy, Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy, Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Ho Sueb Song
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, Bokjeong-dong, Sujeong-gu, Seongnam, Gyeonggii, Republic of Korea
| | - Jung Hyun Kim
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, Bokjeong-dong, Sujeong-gu, Seongnam, Gyeonggii, Republic of Korea
| | - Seong Cheol Ko
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, Bokjeong-dong, Sujeong-gu, Seongnam, Gyeonggii, Republic of Korea
| | - Min Jong Song
- Department of Obstetrics and Gynecology, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Jung-gu, Daejeon, Republic of Korea
| | - Won Hyoung Lee
- Department of Nuclear Medicine Chungbuk National University Hospital, Seowon, Cheongju, Chungbuk, Republic of Korea
| | - Joo Hee Yoon
- Department of Obstetrics and Gynecology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Paldal-gu, Suwon, Gyeonggi-do, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, USA
| | - Sang Bae Han
- College of Pharmacy, Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
11
|
Lv Z, Fan J, Zhang X, Huang Q, Han J, Wu F, Hu G, Guo M, Jin Y. Integrative genomic analysis of interleukin-36RN and its prognostic value in cancer. Mol Med Rep 2015; 13:1404-12. [PMID: 26676204 DOI: 10.3892/mmr.2015.4667] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/28/2015] [Indexed: 11/05/2022] Open
Abstract
Interleukin (IL)-36RN, previously known as IL1-F5 and IL-1δ, shares a 360-kb region of chromosome 2q13 with members of IL-1 systems. IL-36RN encodes an anti-inflammatory cytokine, IL-36 receptor antagonist (IL-36Ra). In spite of IL-36Ra showing the highest homology to IL-1 receptor (IL-1R) antagonist, it differs from the latter in aspects including its binding to IL-lRrp2 but not to IL-1R1. IL-36RN is mainly expressed in epithelial cells and has important roles in inflammatory diseases. In the present study, IL-36RN was identified in the genomes of 27 species, including human, chimpanzee, mouse, horse and dolphin. Human IL-36RN was mainly expressed in the eye, head and neck, fetal heart, lung, testis, cervix and placenta; furthermore, it was highly expressed in bladder and parathyroid tumors. Furthermore, a total of 30 single nucleotide polymorphisms causing missense mutations were determined, which are considered to be the causes of various diseases, such as generalized pustular psoriasis. In addition, the link between IL-36RN and the prognosis of certain cancer types was revealed through meta-analysis. Tumor-associated transcriptional factors c-Fos, activator protein-1, c-Jun and nuclear factor κB were found to bind to the upstream region in the IL-36RN gene. This may indicate that IL-36RN is involved in tumorigenesis and tumor progression through the regulation of tumor-associated transcriptional factors. The present study identified IL-36RN in various species and investigated the associations between IL-36RN and cancer prognosis, which would determine whether IL-36RN drove the evolution of the various species with regard to tumorigenesis.
Collapse
Affiliation(s)
- Zhilei Lv
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jinshuo Fan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiuxiu Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qi Huang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jieli Han
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guorong Hu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
12
|
Bi X, Xia X, Fan D, Mu T, Zhang Q, Iozzo RV, Yang W. Oncogenic activin C interacts with decorin in colorectal cancer in vivo and in vitro. Mol Carcinog 2015; 55:1786-1795. [DOI: 10.1002/mc.22427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 10/09/2015] [Accepted: 10/18/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Xiuli Bi
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Xichun Xia
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Dongdong Fan
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Teng Mu
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Qiuhua Zhang
- Department of Pharmacology; Liaoning Traditional Chinese Medicine University; Liaoning 110036 China
| | - Renato V. Iozzo
- Department of Pathology; Anatomy and Cell Biology; Thomas Jefferson University; Philadelphia Pennsylvania 19107
| | - Wancai Yang
- Department of Pathology and Institute of Precision Medicine; Jining Medical University; Jining Shandong 272067 China
- Department of Pathology; University of Illinois at Chicago; Chicago Illinois 60612
| |
Collapse
|
13
|
Behl T, Kotwani A. Possible role of endostatin in the antiangiogenic therapy of diabetic retinopathy. Life Sci 2015; 135:131-7. [DOI: 10.1016/j.lfs.2015.06.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 05/15/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023]
|
14
|
Dikshit B, Irshad K, Madan E, Aggarwal N, Sarkar C, Chandra PS, Gupta DK, Chattopadhyay P, Sinha S, Chosdol K. FAT1 acts as an upstream regulator of oncogenic and inflammatory pathways, via PDCD4, in glioma cells. Oncogene 2012; 32:3798-808. [DOI: 10.1038/onc.2012.393] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Extensive evolutionary changes in regulatory element activity during human origins are associated with altered gene expression and positive selection. PLoS Genet 2012; 8:e1002789. [PMID: 22761590 PMCID: PMC3386175 DOI: 10.1371/journal.pgen.1002789] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 05/07/2012] [Indexed: 11/19/2022] Open
Abstract
Understanding the molecular basis for phenotypic differences between humans and other primates remains an outstanding challenge. Mutations in non-coding regulatory DNA that alter gene expression have been hypothesized as a key driver of these phenotypic differences. This has been supported by differential gene expression analyses in general, but not by the identification of specific regulatory elements responsible for changes in transcription and phenotype. To identify the genetic source of regulatory differences, we mapped DNaseI hypersensitive (DHS) sites, which mark all types of active gene regulatory elements, genome-wide in the same cell type isolated from human, chimpanzee, and macaque. Most DHS sites were conserved among all three species, as expected based on their central role in regulating transcription. However, we found evidence that several hundred DHS sites were gained or lost on the lineages leading to modern human and chimpanzee. Species-specific DHS site gains are enriched near differentially expressed genes, are positively correlated with increased transcription, show evidence of branch-specific positive selection, and overlap with active chromatin marks. Species-specific sequence differences in transcription factor motifs found within these DHS sites are linked with species-specific changes in chromatin accessibility. Together, these indicate that the regulatory elements identified here are genetic contributors to transcriptional and phenotypic differences among primate species. The human genome shares a remarkable amount of genomic sequence with our closest living primate relatives. Researchers have long sought to understand what regions of the genome are responsible for unique species-specific traits. Previous studies have shown that many genes are differentially expressed between species, but the regulatory elements contributing to these differences are largely unknown. Here we report a genome-wide comparison of active gene regulatory elements in human, chimpanzee, and macaque, and we identify hundreds of regulatory elements that have been gained or lost in the human or chimpanzee genomes since their evolutionary divergence. These elements contain evidence of natural selection and correlate with species-specific changes in gene expression. Polymorphic DNA bases in transcription factor motifs that we found in these regulatory elements may be responsible for the varied biological functions across species. This study directly links phenotypic and transcriptional differences between species with changes in chromatin structure.
Collapse
|
16
|
Elbaz HA, Stueckle TA, Tse W, Rojanasakul Y, Dinu CZ. Digitoxin and its analogs as novel cancer therapeutics. Exp Hematol Oncol 2012; 1:4. [PMID: 23210930 PMCID: PMC3506989 DOI: 10.1186/2162-3619-1-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/05/2012] [Indexed: 01/18/2023] Open
Abstract
A growing body of evidence indicates that digitoxin cardiac glycoside is a promising anticancer agent when used at therapeutic concentrations. Digitoxin has a prolonged half-life and a well-established clinical profile. New scientific avenues have shown that manipulating the chemical structure of the saccharide moiety of digitoxin leads to synthetic analogs with increased cytotoxic activity. However, the anticancer mechanism of digitoxin or synthetic analogs is still subject to study while concerns about digitoxin's cardiotoxicity preclude its clinical application in cancer therapeutics. This review focuses on digitoxin and its analogs, and their cytotoxicity against cancer cells. Moreover, a new perspective on the pharmacological aspects of digitoxin and its analogs is provided to emphasize new research directions for developing potent chemotherapeutic drugs.
Collapse
Affiliation(s)
- Hosam A Elbaz
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | | | | | | | | |
Collapse
|
17
|
Li Y, Liu J, Yu S, Proksch P, Gu J, Lin W. TNF-α inhibitory diterpenoids from the Chinese mangrove plant Excoecaria agallocha L. PHYTOCHEMISTRY 2010; 71:2124-2131. [PMID: 20822783 DOI: 10.1016/j.phytochem.2010.08.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 06/06/2010] [Accepted: 08/09/2010] [Indexed: 05/29/2023]
Abstract
Chemical examination of the stems and twigs of the mangrove plant Excoecaria agallocha L. resulted in the isolation of six ent-kaurane diterpenoids named agallochaols K-P (1-6), an atisane-type diterpenoid agallochaol Q (7), along with eight known diterpenoids (8-15). Their structures were elucidated on the basis of extensive spectroscopic analysis and by comparison of their NMR spectroscopic data with those reported in literature, in association with the biogenetic relationship with the X-ray structure of 9. Compounds 1, 5-7, 9-10, and 13 showed anti-inflammatory potency to suppress expression of NF-κB and AP-1 targeted genes including TNF-α and IL-6 induced by lipopolysaccharide (LPS) in mouse macrophages Raw 264.7 cells. In addition, compounds 1, 5-7, 9-10, and 13 block NF-κB activation, while compounds 1 and 7 block AP-1 activation dramatically, indicating these compounds possess an anti-inflammatory potential in vitro.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/isolation & purification
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Diterpenes/chemistry
- Diterpenes/isolation & purification
- Diterpenes/pharmacology
- Diterpenes, Kaurane/chemistry
- Diterpenes, Kaurane/isolation & purification
- Diterpenes, Kaurane/pharmacology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Euphorbiaceae/chemistry
- Interleukin-6/antagonists & inhibitors
- Lipopolysaccharides/pharmacology
- Macrophages/drug effects
- Mice
- NF-kappa B/antagonists & inhibitors
- Nuclear Magnetic Resonance, Biomolecular
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Yongxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China
| | | | | | | | | | | |
Collapse
|
18
|
Enterovirus 71 induces COX-2 expression via MAPKs, NF-kappaB, and AP-1 in SK-N-SH cells: Role of PGE(2) in viral replication. Cell Signal 2009; 22:234-46. [PMID: 19800403 DOI: 10.1016/j.cellsig.2009.09.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 09/10/2009] [Accepted: 09/14/2009] [Indexed: 12/21/2022]
Abstract
The enterovirus 71 (EV71) causes severe neurological diseases that were mediated through cyclooxygenase-2 (COX-2) expression in brain. However, the mechanisms underlying EV71-initiated intracellular signaling pathways leading to COX-2 expression remain unknown in neurons. Here we report that exposure of SK-N-SH cells to EV71 increased COX-2 expression and PGE(2) generation in a time- and virus titer-dependent manner, revealed by Western blot, real-time PCR, and PGE(2) analyses. These EV71-induced responses were mediated through activation of p42/p44 MAPK, p38 MAPK, JNK, NF-kappaB, and AP-1, revealed by using selective pharmacological inhibitors or transfection with respective siRNAs. Consistently, EV71-stimulated translocation of NF-kappaB into the nucleus and degradation of IkappaBalpha in the cytosol was blocked by pretreatment with the selective inhibitors of MEK1/2 (U0126) and NF-kappaB (Bay11-7085), respectively, suggesting that MEK1/2-p42/p44 MAPK cascade linking to NF-kappaB was involved in COX-2 expression. In addition, EV71-induced AP-1 subunits (c-jun and c-fos mRNA) expression was also attenuated by pretreatment with a selective JNK inhibitor SP600125, suggesting that JNK cascade linking to AP-1 was involved in COX-2 expression induced by EV71. These findings suggested that up-regulation of COX-2 associated with the release of PGE(2) from EV71-infected SK-N-SH cells which was mediated through activation of p38 MAPK, JNK, p42/p44 MAPK, NF-kappaB, and AP-1 pathways.
Collapse
|
19
|
Hussain S, Bharti AC, Salam I, Bhat MA, Mir MM, Hedau S, Siddiqi MA, Basir SF, Das BC. Transcription factor AP-1 in esophageal squamous cell carcinoma: alterations in activity and expression during human Papillomavirus infection. BMC Cancer 2009; 9:329. [PMID: 19758438 PMCID: PMC2758900 DOI: 10.1186/1471-2407-9-329] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 09/16/2009] [Indexed: 01/01/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a leading cause of cancer-related deaths in Jammu and Kashmir (J&K) region of India. A substantial proportion of esophageal carcinoma is associated with infection of high-risk HPV type 16 and HPV18, the oncogenic expression of which is controlled by host cell transcription factor Activator Protein-1 (AP-1). We, therefore, have investigated the role of DNA binding and expression pattern of AP-1 in esophageal cancer with or without HPV infection. Methods Seventy five histopathologically-confirmed esophageal cancer and an equal number of corresponding adjacent normal tissue biopsies from Kashmir were analyzed for HPV infection, DNA binding activity and expression of AP-1 family of proteins by PCR, gel shift assay and immunoblotting respectively. Results A high DNA binding activity and elevated expression of AP-1 proteins were observed in esophageal cancer, which differed between HPV positive (19%) and HPV negative (81%) carcinomas. While JunB, c-Fos and Fra-1 were the major contributors to AP-1 binding activity in HPV negative cases, Fra-1 was completely absent in HPV16 positive cancers. Comparison of AP-1 family proteins demonstrated high expression of JunD and c-Fos in HPV positive tumors, but interestingly, Fra-1 expression was extremely low or nil in these tumor tissues. Conclusion Differential AP-1 binding activity and expression of its specific proteins between HPV - positive and HPV - negative cases indicate that AP-1 may play an important role during HPV-induced esophageal carcinogenesis.
Collapse
Affiliation(s)
- Showket Hussain
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Inhibition of mammalian thioredoxin reductase by black tea and its constituents: implications for anticancer actions. Biochimie 2008; 91:434-44. [PMID: 19059456 DOI: 10.1016/j.biochi.2008.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Accepted: 11/18/2008] [Indexed: 11/20/2022]
Abstract
Black tea is recently reported to have anti-carcinogenic effects through pro-oxidant property, but the underlying mechanisms remain unclear. Mammalian cytosolic thioredoxin reductase (TrxR1) is well -known for its anti-oxidation activity. In this study, we found that black tea extract (BTE) and theaflavins (TFs), the major black tea polyphenols, inhibited the purified TrxR1 with IC(50) 44 microg/ml and 21+/-1 microg/ml, respectively. Kinetics of TFs exhibited a mixed type of competitive and non-competitive inhibition, with K(is) 4+/-1 microg/ml and K(ii) 26+/-5 microg/ml against coenzyme NADPH, and with K(is) 12+/-3 microg/ml and K(ii) 27+/-5 microg/ml against substrate DTNB. In addition, TFs inhibited TrxR1 in a time-dependent manner. In an equilibrium step, a reversible TrxR1-TFs complex (E*I) forms, which is followed by a slow irreversible first-order inactivation step. Rate constant of the inactivation was 0.7 min(-1), and dissociation constant of E*I was 51.9 microg/ml. Treatment of NADPH-reduced TrxR1 with TFs decreased 5-(Iodoacetamido) fluorescein incorporation, a fluorescent thiol-reactive reagent, suggesting that Sec/Cys residue(s) in the active site may be involved in the binding of TFs. The inhibitory capacity of TFs depends on their structure. Among the TFs tested, gallated forms had strong inhibitory effects. The interactions between TFs and TrxR1 were investigated by molecular docking, which revealed important features of the binding mechanism of theaflavins. An inhibitory effect of BTE on viability of HeLa cells was observed with IC(50) 29 microg/ml. At 33 microg/ml of BTE, TrxR1 activity in HeLa cells was decreased by 73% at 22 h after BTE treatment. TFs inhibited cell viability with IC(50) 10+/-4 microg/ml for HeLa cells and with IC(50) 20+/-5 microg/ml for EAhy926 cells. The cell susceptibility to TFs was inversely correlated to cellular levels of TrxR1. The inhibitory actions of TFs on TrxR1 may be an important mechanism of their anti-cancer properties.
Collapse
|
21
|
Garaude J, Farrás R, Bossis G, Charni S, Piechaczyk M, Hipskind RA, Villalba M. SUMOylation regulates the transcriptional activity of JunB in T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2008; 180:5983-90. [PMID: 18424718 DOI: 10.4049/jimmunol.180.9.5983] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The AP-1 family member JunB is a critical regulator of T cell function. JunB is a transcriptional activator of various cytokine genes, such as IL-2, IL-4, and IL-10; however, the post-translational modifications that regulate JunB activity in T cells are poorly characterized. We show here that JunB is conjugated with small ubiquitin-like modifier (SUMO) on lysine 237 in resting and activated primary T cells and T cell lines. Sumoylated JunB associated with the chromatin-containing insoluble fraction of cells, whereas nonsumoylated JunB was also in the soluble fraction. Blocking JunB sumoylation by mutation or use of a dominant-negative form of the SUMO-E2 Ubc-9 diminished its ability to transactivate IL-2 and IL-4 reporter genes. In contrast, nonsumoylable JunB mutants showed unimpaired activity with reporter genes controlled by either synthetic 12-O-tetradecanoylphorbol-13-acetate response elements or NF-AT/AP-1 and CD28RE sites derived from the IL-2 promoter. Ectopic expression of JunB in activated human primary CD4(+) T cells induced activation of the endogenous IL-2 promoter, whereas the nonsumoylable JunB mutant did not. Thus, our work demonstrates that sumoylation of JunB regulates its ability to induce cytokine gene transcription and likely plays a critical role in T cell activation.
Collapse
Affiliation(s)
- Johan Garaude
- Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5535, 1919 route de Mende, Montpellier cedex 5, France.
| | | | | | | | | | | | | |
Collapse
|
22
|
Kwon MH, Lee GE, Kwon SJ, Choi E, Na MJ, Cho HM, Kim YJ, Sul HJ, Cho YJ, Son JW. Identification of DNA Methylation Markers for NSCLC Using Hpall-Mspl Methylation Microarray. Tuberc Respir Dis (Seoul) 2008. [DOI: 10.4046/trd.2008.65.6.495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Mi Hye Kwon
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| | - Go Eun Lee
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| | - Sun Jung Kwon
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| | - Eugene Choi
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| | - Moon Jun Na
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| | - Hyun Min Cho
- Department of Chest Surgery, Konyang University School of Medicine, Daejeon, Korea
| | - Young Jin Kim
- Department of Chest Surgery, Konyang University School of Medicine, Daejeon, Korea
| | - Hye Jung Sul
- Department of Pathology, Konyang University School of Medicine, Daejeon, Korea
| | - Young Jun Cho
- Department of Radiology, Konyang University School of Medicine, Daejeon, Korea
| | - Ji Woong Son
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Korea
| |
Collapse
|
23
|
Abstract
The role of oxidative stress has been well appreciated in the development of sepsis-induced acute lung injury (ALI). Oxidative stress in sepsis-induced ALI is believed to be initiated by products of activated lung macrophages and infiltrated neutrophils, promptly propagating to lung epithelial and endothelial cells. This leads to tissue damage and organ dysfunction. On stimulation, neutrophils (PMNs) enable their migration machinery. The lung undergoes changes favoring adhesion and transmigration of PMNs, resulting in PMN accumulation in lung, which is a characteristic of sepsis-induced ALI. Oxidative stress turns on the redox-sensitive transcription factors (NF-kappaB, AP-1), resulting in a large output of proinflammatory cytokines and chemokines, which further aggravate inflammation and oxidative stress. During the process, transcription factor nuclear factor-erythroid 2-p45-related factor 2 (Nrf2) and heme oxygenase (HO) appear to play the counterbalancing roles to limit the propagation of oxidative stress and inflammatory responses in lung. Many antioxidants have been tested to treat sepsis-induced ALI in animal models and in patients with sepsis. However, the results are inconclusive. In this article, we focus on the current understanding of the pathogenesis of sepsis-induced ALI and novel antioxidant strategies for therapeutic purposes.
Collapse
Affiliation(s)
- Ren-Feng Guo
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA.
| | | |
Collapse
|
24
|
Masa SR, Lando R, Sarid R. Transcriptional regulation of the open reading frame 35 encoded by Kaposi's sarcoma-associated herpesvirus. Virology 2007; 371:14-31. [PMID: 17963810 DOI: 10.1016/j.virol.2007.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 07/16/2007] [Accepted: 08/17/2007] [Indexed: 11/26/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a member of the Gammaherpesvirinae and is causally associated with Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. The KSHV genome encodes over 85 genes; the function of some is entirely unknown. We have characterized the transcriptional regulation of a conserved and uncharacterized Gammaherpesvirinae open reading frame, orf35, which lies in a cluster of several overlapping genes, orf34 to orf38. We identified the transcription start site and analyzed upstream sequences. We found that expression of the KSHV lytic replication and transcription activator (RTA) strongly increased the orf35 promoter activity through a 46-nucleotide region which includes a conserved AP-1 binding site. Electrophoretic mobility shift assay demonstrated direct binding of cJUN and cFOS to the predicted AP-1 binding site. Finally, using a mutated promoter lacking the AP-1 site and dominant-negative cFOS, we established that the RTA-mediated orf35 transactivation is AP-1-dependent.
Collapse
Affiliation(s)
- Shiri-Rivka Masa
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, 52900, Israel
| | | | | |
Collapse
|