1
|
Klawitter M, El-Ayoubi A, Buch J, Rüttinger J, Ehrenfeld M, Lichtenegger E, Krüger MA, Mantwill K, Koll FJ, Kowarik MC, Holm PS, Naumann U. The Oncolytic Adenovirus XVir-N-31, in Combination with the Blockade of the PD-1/PD-L1 Axis, Conveys Abscopal Effects in a Humanized Glioblastoma Mouse Model. Int J Mol Sci 2022; 23:ijms23179965. [PMID: 36077380 PMCID: PMC9456411 DOI: 10.3390/ijms23179965] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is an obligatory lethal brain tumor with a median survival, even with the best standard of care therapy, of less than 20 months. In light of this fact, the evaluation of new GBM treatment approaches such as oncolytic virotherapy (OVT) is urgently needed. Based on our preliminary preclinical data, the YB-1 dependent oncolytic adenovirus (OAV) XVir-N-31 represents a promising therapeutic agent to treat, in particular, therapy resistant GBM. Preclinical studies have shown that XVir-N-31 prolonged the survival of GBM bearing mice. Now using an immunohumanized mouse model, we examined the immunostimulatory effects of XVir-N-31 in comparison to the wildtype adenovirus (Ad-WT). Additionally, we combined OVT with the inhibition of immune checkpoint proteins by using XVir-N-31 in combination with nivolumab, or by using a derivate of XVir-N-31 that expresses a PD-L1 neutralizing antibody. Although in vitro cell killing was higher for Ad-WT, XVir-N-31 induced a much stronger immunogenic cell death that was further elevated by blocking PD-1 or PD-L1. In vivo, an intratumoral injection of XVir-N-31 increased tumor infiltrating lymphocytes (TILs) and NK cells significantly more than Ad-WT not only in the virus-injected tumors, but also in the untreated tumors growing in the contralateral hemisphere. This suggests that for an effective treatment of GBM, immune activating properties by OAVs seem to be of greater importance than their oncolytic capacity. Furthermore, the addition of immune checkpoint inhibition (ICI) to OVT further induced lymphocyte infiltration. Consequently, a significant reduction in contralateral non-virus-injected tumors was only visible if OVT was combined with ICI. This strongly indicates that for an effective eradication of GBM cells that cannot be directly targeted by an intratumoral OV injection, additional ICI therapy is required.
Collapse
Affiliation(s)
- Moritz Klawitter
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
| | - Ali El-Ayoubi
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
| | - Jasmin Buch
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
| | - Jakob Rüttinger
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
| | - Maximilian Ehrenfeld
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Eva Lichtenegger
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Marcel A. Krüger
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, D-72076 Tübingen, Germany
| | - Klaus Mantwill
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Florestan J. Koll
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Markus C. Kowarik
- B Cell Immunology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
| | - Per Sonne Holm
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
- Department of Oral and Maxillofacial Surgery, Medical University Innsbruck, A-6020 Innsbruck, Austria
- XVir Therapeutics GmbH, D-80331 Munich, Germany
| | - Ulrike Naumann
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany
- Correspondence:
| |
Collapse
|
2
|
Gao Y, Zhao J, Huang Z, Zhao H, Guo Z, Ma S, Tang X, Song W, Chen X. In Situ Reprogramming of Tumors for Activating the OX40/OX40 Ligand Checkpoint Pathway and Boosting Antitumor Immunity. ACS Biomater Sci Eng 2022. [PMID: 35653749 DOI: 10.1021/acsbiomaterials.1c01637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OX40 (CD134, TNFRSF4) is a member of the tumor necrosis factor receptor superfamily that can be activated by its cognate ligand OX40L (CD252, TNFSF4) and functions as a pair of T cell costimulatory molecules. The interaction between OX40 and OX40L (OX40/OX40L) plays a critical role in regulating antitumor immunity, including promoting effector T cells expansion and survival, blocking natural regulatory T cells (Treg) activity, and antagonizing inducible Treg generation. However, current OX40 agonists including anti-OX40 monoclonal antibodies (aOX40) have serious side effects after systemic administration, which limits their clinical success and application. Herein, we propose a strategy to reprogram tumor cells into OX40L-expressing "artificial" antigen-presenting cells (APCs) by OX40L plasmid-loaded nanoparticles for boosting antitumor immunity in situ. A novel gene transfection carrier was prepared by a modular hierarchical assembly method, which could efficiently transfect various tumor cells and express OX40L proteins on their surface. These surface-decorated OX40L proteins were proved to stimulate T cell proliferation in vitro while stimulating strong antitumor immune responses in vivo. Importantly, this in situ reprogramming strategy did not induce any toxicity as observed in aOX40 treatment, thus providing a novel method for immune checkpoint stimulator application.
Collapse
Affiliation(s)
- Yuxi Gao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,University of Science and Technology of China, Hefei 230026, China
| | - Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,University of Science and Technology of China, Hefei 230026, China
| | - Hanqin Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,University of Science and Technology of China, Hefei 230026, China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,University of Science and Technology of China, Hefei 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| |
Collapse
|
3
|
Ding P, Ma Z, Liu D, Pan M, Li H, Feng Y, Zhang Y, Shao C, Jiang M, Lu D, Han J, Wang J, Yan X. Lysine Acetylation/Deacetylation Modification of Immune-Related Molecules in Cancer Immunotherapy. Front Immunol 2022; 13:865975. [PMID: 35585975 PMCID: PMC9108232 DOI: 10.3389/fimmu.2022.865975] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
As major post-translational modifications (PTMs), acetylation and deacetylation are significant factors in signal transmission and cellular metabolism, and are modulated by a dynamic process via two pivotal categories of enzymes, histone acetyltransferases (HATs) and histone deacetylases (HDACs). In previous studies, dysregulation of lysine acetylation and deacetylation has been reported to be associated with the genesis and development of malignancy. Scientists have recently explored acetylation/deacetylation patterns and prospective cancer therapy techniques, and the FDA has approved four HDAC inhibitors (HDACi) to be used in clinical treatment. In the present review, the most recent developments in the area of lysine acetylation/deacetylation alteration in cancer immunotherapy were investigated. Firstly, a brief explanation of the acetylation/deacetylation process and relevant indispensable enzymes that participate therein is provided. Subsequently, a multitude of specific immune-related molecules involved in the lysine acetylation/deacetylation process are listed in the context of cancer, in addition to several therapeutic strategies associated with lysine acetylation/deacetylation modification in cancer immunotherapy. Finally, a number of prospective research fields related to cancer immunotherapy concepts are offered with detailed analysis. Overall, the present review may provide a reference for researchers in the relevant field of study, with the aim of being instructive and meaningful to further research as well as the selection of potential targets and effective measures for future cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Dong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Huizi Li
- Department of Outpatient, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Menglong Jiang
- Department of Thoracic Surgery, 1st Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| | - Jinliang Wang
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| |
Collapse
|
4
|
Araki T, Tone Y, Yamamoto M, Kameda H, Ben-Shlomo A, Yamada S, Takeshita A, Yamamoto M, Kawakami Y, Tone M, Melmed S. Two Distinctive POMC Promoters Modify Gene Expression in Cushing Disease. J Clin Endocrinol Metab 2021; 106:e3346-e3363. [PMID: 34061962 PMCID: PMC8372657 DOI: 10.1210/clinem/dgab387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Mechanisms underlying pituitary corticotroph adenoma adrenocorticotropin (ACTH) production are poorly understood, yet circulating ACTH levels closely correlate with adenoma phenotype and clinical outcomes. OBJECTIVE We characterized the 5' ends of proopiomelanocortin (POMC) gene transcripts, which encode the precursor polypeptide for ACTH, in order to investigate additional regulatory mechanisms of POMC gene transcription and ACTH production. METHODS We examined 11 normal human pituitary tissues, 32 ACTH-secreting tumors, as well as 6 silent corticotroph adenomas (SCAs) that immunostain for but do not secrete ACTH. RESULTS We identified a novel regulatory region located near the intron 2/exon 3 junction in the human POMC gene, which functions as a second promoter and an enhancer. In vitro experiments demonstrated that CREB binds the second promoter and regulates its transcriptional activity. The second promoter is highly methylated in SCAs, partially demethylated in normal pituitary tissue, and highly demethylated in pituitary and ectopic ACTH-secreting tumors. In contrast, the first promoter is demethylated in all POMC-expressing cells and is highly demethylated only in pituitary ACTH-secreting tumors harboring the ubiquitin-specific protease 8 (USP8) mutation. Demethylation patterns of the second promoter correlate with clinical phenotypes of Cushing disease. CONCLUSION We identified a second POMC promoter regulated by methylation status in ACTH-secreting pituitary tumors. Our findings open new avenues for elucidating subcellular regulation of the hypothalamic-pituitary-adrenal axis and suggest the second POMC promoter may be a target for therapeutic intervention to suppress excess ACTH production.
Collapse
Affiliation(s)
- Takako Araki
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yukiko Tone
- Pacific Heart, Lung, & Blood Institute, Los Angeles, California, USA
| | - Masaaki Yamamoto
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hiraku Kameda
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Anat Ben-Shlomo
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shozo Yamada
- Department of Endocrinology and Metabolism, Toranomon Hospital, Tokyo, Japan
| | - Akira Takeshita
- Department of Endocrinology and Metabolism, Toranomon Hospital, Tokyo, Japan
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Masahide Tone
- Pacific Heart, Lung, & Blood Institute, Los Angeles, California, USA
| | - Shlomo Melmed
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Correspondence: Shlomo Melmed, MD, Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Plaza North, Room 2015, Los Angeles, CA 90048, USA.
| |
Collapse
|
5
|
Sun G, Jin H, Zhang C, Meng H, Zhao X, Wei D, Ou X, Wang Q, Li S, Wang T, Sun X, Shi W, Tian D, Liu K, Xu H, Tian Y, Li X, Guo W, Jia J, Zhang Z, Zhang D. OX40 Regulates Both Innate and Adaptive Immunity and Promotes Nonalcoholic Steatohepatitis. Cell Rep 2019; 25:3786-3799.e4. [PMID: 30590049 DOI: 10.1016/j.celrep.2018.12.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/26/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Both innate and adaptive immune cells are involved in the pathogenesis of nonalcoholic steatohepatitis (NASH), but the crosstalk between innate and adaptive immunity is largely unknown. Here we show that compared with WT mice, OX40-/- mice exhibit decreased liver fat accumulation, lobular inflammation, and focal necrosis after feeding with diets that induce NASH. Mechanistically, OX40 deficiency suppresses Th1 and Th17 differentiation, and OX40 deficiency in T cells inhibits monocyte migration, antigen presentation, and M1 polarization. Soluble OX40 stimulation alone upregulates antigen presentation, chemokine receptor expression, and proinflammatory cytokine secretion by liver monocytes. Furthermore, plasma soluble OX40 levels are positively associated with NASH in humans, suggesting clinical relevance of the findings. In conclusion, we show a mechanism for T cell regulation of innate immune cells. OX40 is a key regulator of both intrahepatic innate and adaptive immunity, generates two-way signals, and promotes both proinflammatory monocyte and macrophage and T cell function, resulting in NASH development.
Collapse
Affiliation(s)
- Guangyong Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Jin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Chunpan Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Meng
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dan Wei
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xiaojuan Ou
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Qianyi Wang
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Shuxiang Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Tianqi Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xiaojing Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Yue Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xinmin Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wei Guo
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Jidong Jia
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, 100050, China
| | - Zhongtao Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
6
|
Haymaker C, Yang Y, Wang J, Zou Q, Sahoo A, Alekseev A, Singh D, Ritthipichai K, Hailemichael Y, Hoang ON, Qin H, Schluns KS, Wang T, Overwijk WW, Sun SC, Bernatchez C, Kwak LW, Neelapu SS, Nurieva R. Absence of Grail promotes CD8 + T cell anti-tumour activity. Nat Commun 2017; 8:239. [PMID: 28798332 PMCID: PMC5552797 DOI: 10.1038/s41467-017-00252-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2017] [Indexed: 12/15/2022] Open
Abstract
T-cell tolerance is a major obstacle to successful cancer immunotherapy; thus, developing strategies to break immune tolerance is a high priority. Here we show that expression of the E3 ubiquitin ligase Grail is upregulated in CD8+ T cells that have infiltrated into transplanted lymphoma tumours, and Grail deficiency confers long-term tumour control. Importantly, therapeutic transfer of Grail-deficient CD8+ T cells is sufficient to repress established tumours. Mechanistically, loss of Grail enhances anti-tumour reactivity and functionality of CD8+ T cells. In addition, Grail-deficient CD8+ T cells have increased IL-21 receptor (IL-21R) expression and hyperresponsiveness to IL-21 signalling as Grail promotes IL-21R ubiquitination and degradation. Moreover, CD8+ T cells isolated from lymphoma patients express higher levels of Grail and lower levels of IL-21R, compared with CD8+ T cells from normal donors. Our data demonstrate that Grail is a crucial factor controlling CD8+ T-cell function and is a potential target to improve cytotoxic T-cell activity.Grail is an E3 ubiquitin ligase that inhibits T-cell receptor signalling in CD4+ T cells. Here the authors show Grail also limits IL-21 receptor expression and function in CD8+ T cells, is overactive in these cells in patients with lymphoma, and promotes tumour development in a lymphoma transplant mouse model.
Collapse
Affiliation(s)
- Cara Haymaker
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yi Yang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Department of Radiation Oncology, The Second Hospital of Jilin University, No. 218 Ziqiang St., Changchun City, Jilin Province, 130041, China
| | - Junmei Wang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Qiang Zou
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Anupama Sahoo
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Andrei Alekseev
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Divyendu Singh
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Krit Ritthipichai
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yared Hailemichael
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Oanh N Hoang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Hong Qin
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Toni Stephenson Lymphoma Center, City of Hope, 1500 East Duarte Rd., Duarte, CA, 91010, USA
| | - Kimberly S Schluns
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, No. 218 Ziqiang St., Changchun City, Jilin Province, 130041, China
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Shao-Cong Sun
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Larry W Kwak
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Toni Stephenson Lymphoma Center, City of Hope, 1500 East Duarte Rd., Duarte, CA, 91010, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Roza Nurieva
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Abstract
The tumour necrosis factor receptor OX40 (CD134) is activated by its cognate ligand OX40L (CD134L, CD252) and functions as a T cell co-stimulatory molecule. OX40-OX40L interactions have been proposed as a potential therapeutic target for treating autoimmunity. OX40 is expressed on activated T cells, and in the mouse at rest on regulatory T cells (Treg). OX40L is found on antigen-presenting cells, activated T cells and others including lymphoid tissue inducer cells, some endothelia and mast cells. Expression of both molecules is increased after antigen presentation occurs and also in response to multiple other pro-inflammatory factors including CD28 ligation, CD40L ligation and interferon-gamma signaling. Their interactions promote T cell survival, promote an effector T cell phenotype, promote T cell memory, tend to reduce regulatory function, increase effector cytokine production and enhance cell mobility. In some circumstances, OX40 agonism may be associated with increased tolerance, although timing with respect to antigenic stimulus is important. Further, recent work has suggested that OX40L blockade may be more effective than OX40 blockade in reducing autoimmunity. This article reviews the expression of OX40 and OX40L in health, the effects of their interactions and insights from their under- or over-expression. We then review OX40 and OX40L expression in human autoimmune disease, identified associations of variations in their genes (TNFRSF4 and TNFSF4, respectively) with autoimmunity, and data from animal models of human diseases. A rationale for blocking OX40-OX40L interaction in human autoimmunity is then presented along with commentary on the one trial of OX40L blockade in human disease conducted to date. Finally, we discuss potential problems with clinical use of OX40-OX40L directed pharmacotherapy.
Collapse
Affiliation(s)
- Gwilym J Webb
- MRC Centre for Immune Regulation, Institute of Biomedical Research, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK. .,National Institute for Health Research Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK.
| | - Gideon M Hirschfield
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Peter J L Lane
- MRC Centre for Immune Regulation, Institute of Biomedical Research, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| |
Collapse
|
8
|
Dillingham BC, Knoblach SM, Many GM, Harmon BT, Mullen AM, Heier CR, Bello L, McCall JM, Hoffman EP, Connor EM, Nagaraju K, Reeves EKM, Damsker JM. VBP15, a novel anti-inflammatory, is effective at reducing the severity of murine experimental autoimmune encephalomyelitis. Cell Mol Neurobiol 2015; 35:377-387. [PMID: 25392236 PMCID: PMC11486228 DOI: 10.1007/s10571-014-0133-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 10/27/2014] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis is a chronic disease of the central nervous system characterized by an autoimmune inflammatory reaction that leads to axonal demyelination and tissue damage. Glucocorticoids, such as prednisolone, are effective in the treatment of multiple sclerosis in large part due to their ability to inhibit pro-inflammatory pathways (e.g., NFκB). However, despite their effectiveness, long-term treatment is limited by adverse side effects. VBP15 is a recently described compound synthesized based on the lazeroid steroidal backbone that shows activity in acute and chronic inflammatory conditions, yet displays a much-reduced side effect profile compared to traditional glucocorticoids. The purpose of this study was to determine the effectiveness of VBP15 in inhibiting inflammation and disease progression in experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of multiple sclerosis. Our data show that VBP15 is effective at reducing both disease onset and severity. In parallel studies, we observed that VBP15 was able to inhibit the production of NFκB-regulated pro-inflammatory transcripts in human macrophages. Furthermore, treatment with prednisolone-but not VBP15-increased expression of genes associated with bone loss and muscle atrophy, suggesting lack of side effects of VBP15. These findings suggest that VBP15 may represent a potentially safer alternative to traditional glucocorticoids in the treatment of multiple sclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Blythe C Dillingham
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - Susan M Knoblach
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
- Department of Integrative Systems Biology, Children's National Medical Center and George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Gina M Many
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - Brennan T Harmon
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - Amanda M Mullen
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - Christopher R Heier
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - Luca Bello
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
| | - John M McCall
- PharMac LLC, Boca Grande, FL, 33921, USA
- ReveraGen BioPharma, Silver Spring, MD, 20910, USA
| | - Eric P Hoffman
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
- Department of Integrative Systems Biology, Children's National Medical Center and George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
- ReveraGen BioPharma, Silver Spring, MD, 20910, USA
| | | | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20010, USA
- Department of Integrative Systems Biology, Children's National Medical Center and George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
- ReveraGen BioPharma, Silver Spring, MD, 20910, USA
| | | | | |
Collapse
|
9
|
Tone Y, Kidani Y, Ogawa C, Yamamoto K, Tsuda M, Peter C, Waldmann H, Tone M. Gene expression in the Gitr locus is regulated by NF-κB and Foxp3 through an enhancer. THE JOURNAL OF IMMUNOLOGY 2014; 192:3915-24. [PMID: 24634496 DOI: 10.4049/jimmunol.1302174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucocorticoid-induced TNFR (Gitr) and Ox40, two members of the TNFR superfamily, play important roles in regulating activities of effector and regulatory T cells (Treg). Their gene expression is induced by T cell activation and further upregulated in Foxp3+ Treg. Although the role of Foxp3 as a transcriptional repressor in Treg is well established, the mechanisms underlying Foxp3-mediated transcriptional upregulation remain poorly understood. This transcription factor seems to upregulate expression not only of Gitr and Ox40, but also other genes, including Ctla4, Il35, Cd25, all critical to Treg function. To investigate how Foxp3 achieves such upregulation, we analyzed its activity on Gitr and Ox40 genes located within a 15.1-kb region. We identified an enhancer located downstream of the Gitr gene, and both Gitr and Ox40 promoter activities were shown to be upregulated by the NF-κB-mediated enhancer activity. We also show, using the Gitr promoter, that the enhancer activity was further upregulated in conjunction with Foxp3. Foxp3 appears to stabilize NF-κB p50 binding by anchoring it to the enhancer, thereby enabling local accumulation of transcriptional complexes containing other members of the NF-κB and IκB families. These findings may explain how Foxp3 can activate expression of certain genes while suppressing others.
Collapse
Affiliation(s)
- Yukiko Tone
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Lian S, Potula HHSK, Pillai MR, Van Stry M, Koyanagi M, Chung L, Watanabe M, Bix M. Transcriptional activation of Mina by Sp1/3 factors. PLoS One 2013; 8:e80638. [PMID: 24324617 PMCID: PMC3851307 DOI: 10.1371/journal.pone.0080638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/15/2013] [Indexed: 12/23/2022] Open
Abstract
Mina is an epigenetic gene regulatory protein known to function in multiple physiological and pathological contexts, including pulmonary inflammation, cell proliferation, cancer and immunity. We showed previously that the level of Mina gene expression is subject to natural genetic variation linked to 21 SNPs occurring in the Mina 5′ region [1]. In order to explore the mechanisms regulating Mina gene expression, we set out to molecularly characterize the Mina promoter in the region encompassing these SNPs. We used three kinds of assays – reporter, gel shift and chromatin immunoprecipitation – to analyze a 2 kb genomic fragment spanning the upstream and intron 1 regions flanking exon 1. Here we discovered a pair of Mina promoters (P1 and P2) and a P1-specific enhancer element (E1). Pharmacologic inhibition and siRNA knockdown experiments suggested that Sp1/3 transcription factors trigger Mina expression through additive activity targeted to a cluster of four Sp1/3 binding sites forming the P1 promoter. These results set the stage for comprehensive analysis of Mina gene regulation from the context of tissue specificity, the impact of inherited genetic variation and the nature of upstream signaling pathways.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cell Line, Tumor
- Chromatin Immunoprecipitation
- Electrophoretic Mobility Shift Assay
- Enhancer Elements, Genetic
- Epigenesis, Genetic
- Genes, Reporter
- Luciferases/genetics
- Luciferases/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymorphism, Single Nucleotide
- Promoter Regions, Genetic
- Protein Binding
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Sp1 Transcription Factor/antagonists & inhibitors
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Sp3 Transcription Factor/antagonists & inhibitors
- Sp3 Transcription Factor/genetics
- Sp3 Transcription Factor/metabolism
- Transcriptional Activation
Collapse
Affiliation(s)
- Shangli Lian
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hari Hara S. K. Potula
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Meenu R. Pillai
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Melanie Van Stry
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Madoka Koyanagi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Linda Chung
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Makiko Watanabe
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Mark Bix
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
11
|
Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat Immunol 2013; 14:489-99. [PMID: 23563690 PMCID: PMC3652626 DOI: 10.1038/ni.2570] [Citation(s) in RCA: 408] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/12/2013] [Indexed: 02/07/2023]
Abstract
Newly activated CD8+ T cells reprogram their metabolism to meet the extraordinary biosynthetic demands of clonal expansion; however, the signals mediating metabolic reprogramming remain poorly defined. Herein, we demonstrate an essential role for sterol regulatory element binding proteins (SREBPs) in the acquisition of effector cell metabolism. Without SREBP signaling, CD8+ T cells are unable to blast, resulting in markedly attenuated clonal expansion during viral infection. Mechanistic studies indicate that SREBPs are essential to meet the heightened lipid requirements of membrane synthesis during blastogenesis. SREBPs are dispensable for homeostatic proliferation, indicating a context-specific requirement for SREBPs in effector responses. These studies provide insights into the molecular signals underlying metabolic reprogramming of CD8+ T cells during the transition from quiescence to activation.
Collapse
|
12
|
Redmond WL, Triplett T, Floyd K, Weinberg AD. Dual anti-OX40/IL-2 therapy augments tumor immunotherapy via IL-2R-mediated regulation of OX40 expression. PLoS One 2012; 7:e34467. [PMID: 22496812 PMCID: PMC3319580 DOI: 10.1371/journal.pone.0034467] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/02/2012] [Indexed: 12/19/2022] Open
Abstract
The provision of T cell co-stimulation via members of the TNFR super-family, including OX40 (CD134) and 4-1BB (CD137), provides critical signals that promote T cell survival and differentiation. Recent studies have demonstrated that ligation of OX40 can augment T cell-mediated anti-tumor immunity in pre-clinical models and more importantly, OX40 agonists are under clinical development for cancer immunotherapy. OX40 is of particular interest as a therapeutic target as it is not expressed on naïve T cells but rather, is transiently up-regulated following TCR stimulation. Although TCR engagement is necessary for inducing OX40 expression, the downstream signals that regulate OX40 itself remain unclear. In this study, we demonstrate that OX40 expression is regulated through a TCR and common gamma chain cytokine-dependent signaling cascade that requires JAK3-mediated activation of the downstream transcription factors STAT3 and STAT5. Furthermore, combined treatment with an agonist anti-OX40 mAb and IL-2 augmented tumor immunotherapy against multiple tumor types. Dual therapy was also able to restore the function of anergic tumor-reactive CD8 T cells in mice with long-term well-established (>5 wks) tumors, leading to increased survival of the tumor-bearing hosts. Together, these data reveal the ability of TCR/common gamma chain cytokine signaling to regulate OX40 expression and demonstrate a novel means of augmenting cancer immunotherapy by providing dual anti-OX40/common gamma chain cytokine-directed therapy.
Collapse
MESH Headings
- Animals
- Blotting, Western
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation
- Cytokines/metabolism
- Drug Therapy, Combination
- Female
- Flow Cytometry
- Immunotherapy
- Interleukin-2/therapeutic use
- Interleukin-2 Receptor alpha Subunit/metabolism
- Janus Kinase 3/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, OX40/physiology
- STAT3 Transcription Factor/physiology
- STAT5 Transcription Factor/physiology
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/metabolism
- Sarcoma, Experimental/therapy
- Signal Transduction
- T-Lymphocytes, Cytotoxic
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon, United States of America.
| | | | | | | |
Collapse
|
13
|
Abstract
Yin Yang 1 (YY1) is a transcription factor with diverse and complex biological functions. YY1 either activates or represses gene transcription, depending on the stimuli received by the cells and its association with other cellular factors. Since its discovery, a biological role for YY1 in tumor development and progression has been suggested because of its regulatory activities toward multiple cancer-related proteins and signaling pathways and its overexpression in most cancers. In this review, we primarily focus on YY1 studies in cancer research, including the regulation of YY1 as a transcription factor, its activities independent of its DNA binding ability, the functions of its associated proteins, and mechanisms regulating YY1 expression and activities. We also discuss the correlation of YY1 expression with clinical outcomes of cancer patients and its target potential in cancer therapy. Although there is not a complete consensus about the role of YY1 in cancers based on its activities of regulating oncogene and tumor suppressor expression, most of the currently available evidence supports a proliferative or oncogenic role of YY1 in tumorigenesis.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | |
Collapse
|
14
|
T-cell activation triggers death receptor-6 expression in a NF-κB and NF-AT dependent manner. Mol Immunol 2011; 48:1439-47. [PMID: 21501873 DOI: 10.1016/j.molimm.2011.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 03/21/2011] [Accepted: 03/28/2011] [Indexed: 11/23/2022]
Abstract
Death receptor-6 (DR6) apparently participates in the regulation of T-cell activation and/or activity as its genetic disruption results in enhanced CD4+ T-cell expansion, the production of Th2 cytokines, and interestingly also the compromised migration of CD4+ T cells to sites of inflammation. However, the mechanism of regulation of DR6 expression in cells of the immune system is not fully understood. In this communication we show that DR6 is not expressed in resting T cells from human peripheral blood or murine lymph nodes but that its expression is significantly upregulated in CD3 crosslinking- or PMA/ionomycin-activated T lymphocytes. DR6 expression is transiently increased in both activated human CD4+ and CD8+ T cells and it is apparently dependent on the activation of NF-κB and NF-AT signaling pathways. In contrast to primary peripheral blood T cells, the widely used model lymphoblastic leukemia T-cell line Jurkat is DR6-positive and unexpectedly, TCR-mediated stimulation of Jurkat cells strongly downregulates DR6 expression via suppression of its transcription.
Collapse
|
15
|
Riveros C, Mellor D, Gandhi KS, McKay FC, Cox MB, Berretta R, Vaezpour SY, Inostroza-Ponta M, Broadley SA, Heard RN, Vucic S, Stewart GJ, Williams DW, Scott RJ, Lechner-Scott J, Booth DR, Moscato P, for the ANZgene Multiple Sclerosis Genetics Consortium. A transcription factor map as revealed by a genome-wide gene expression analysis of whole-blood mRNA transcriptome in multiple sclerosis. PLoS One 2010; 5:e14176. [PMID: 21152067 PMCID: PMC2995726 DOI: 10.1371/journal.pone.0014176] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 10/20/2010] [Indexed: 12/03/2022] Open
Abstract
Background Several lines of evidence suggest that transcription factors are involved in the pathogenesis of Multiple Sclerosis (MS) but complete mapping of the whole network has been elusive. One of the reasons is that there are several clinical subtypes of MS and transcription factors that may be involved in one subtype may not be in others. We investigate the possibility that this network could be mapped using microarray technologies and contemporary bioinformatics methods on a dataset derived from whole blood in 99 untreated MS patients (36 Relapse Remitting MS, 43 Primary Progressive MS, and 20 Secondary Progressive MS) and 45 age-matched healthy controls. Methodology/Principal Findings We have used two different analytical methodologies: a non-standard differential expression analysis and a differential co-expression analysis, which have converged on a significant number of regulatory motifs that are statistically overrepresented in genes that are either differentially expressed (or differentially co-expressed) in cases and controls (e.g., V$KROX_Q6, p-value <3.31E-6; V$CREBP1_Q2, p-value <9.93E-6, V$YY1_02, p-value <1.65E-5). Conclusions/Significance Our analysis uncovered a network of transcription factors that potentially dysregulate several genes in MS or one or more of its disease subtypes. The most significant transcription factor motifs were for the Early Growth Response EGR/KROX family, ATF2, YY1 (Yin and Yang 1), E2F-1/DP-1 and E2F-4/DP-2 heterodimers, SOX5, and CREB and ATF families. These transcription factors are involved in early T-lymphocyte specification and commitment as well as in oligodendrocyte dedifferentiation and development, both pathways that have significant biological plausibility in MS causation.
Collapse
Affiliation(s)
- Carlos Riveros
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - Drew Mellor
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- School of Computer Science and Software Engineering, The University of Western Australia, Crawley, Australia
| | - Kaushal S. Gandhi
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | - Fiona C. McKay
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | - Mathew B. Cox
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- Hunter Medical Research Institute, Newcastle, Australia
| | - Regina Berretta
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - S. Yahya Vaezpour
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- Department of Computer Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Mario Inostroza-Ponta
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- Departamento de Ingeniería Informática, Universidad de Santiago de Chile, Santiago, Chile
| | - Simon A. Broadley
- School of Medicine, Griffith University, Brisbane, Australia
- Department of Neurology, Gold Coast Hospital, Southport, Australia
| | - Robert N. Heard
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | - Stephen Vucic
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | - Graeme J. Stewart
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | | | - Rodney J. Scott
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - Jeanette Lechner-Scott
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - David R. Booth
- Westmead Millennium Institute, University of Sydney, Westmead, Australia
| | - Pablo Moscato
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- Australian Research Council Centre of Excellence in Bioinformatics, St Lucia, Australia
- * E-mail:
| | | |
Collapse
|
16
|
Britanova LV, Kuprash DV. New putative control elements in the promoter of the gene for the CXCL13 chemokine, a target of the alternative NF-κB pathway. Mol Biol 2009. [DOI: 10.1134/s0026893309040128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Teng H, Ballim RD, Mowla S, Prince S. Phosphorylation of histone H3 by protein kinase C signaling plays a critical role in the regulation of the developmentally important TBX2 gene. J Biol Chem 2009; 284:26368-76. [PMID: 19633291 DOI: 10.1074/jbc.m109.021360] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mechanism(s) regulating the expression of the TBX2 gene, a key regulator of development, is poorly understood and thus limits an understanding of its function(s). Here we demonstrate that 12-O-tetradecanoylphorbol-13-acetate (TPA) induces TBX2 expression in normal human fibroblasts in a protein kinase C (PKC)-dependent and MAPK-independent manner. Our data further reveal that TPA activates transcription of TBX2 through activating MSK1, which leads to an increase in phosphorylated histone H3 and the recruitment of Sp1 to the TBX2 gene. In addition, TPA was shown to activate MSK1 in a PKC-dependent and MAPK-independent manner. This study is the first to provide evidence that phosphorylation of histone H3 leads to the transcriptional activation of the TBX2 gene and to link MSK1 to PKC.
Collapse
Affiliation(s)
- Huajian Teng
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925 Cape Town, South Africa
| | | | | | | |
Collapse
|
18
|
Lee J, Hayashi M, Lo JF, Fearns C, Chu WM, Luo Y, Xiang R, Chuang TH. Nuclear factor kappaB (NF-kappaB) activation primes cells to a pro-inflammatory polarized response to a Toll-like receptor 7 (TLR7) agonist. Biochem J 2009; 421:301-10. [PMID: 19426145 PMCID: PMC2746977 DOI: 10.1042/bj20090013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
TLR7 (Toll-like receptor 7) mediates anti-viral immunity by recognizing ssRNA (single-stranded RNA) viruses. Small-molecular-mass TLR7 agonists have been approved, or are being evaluated, for treatment of cancers or infectious diseases. Although TLR7 is predominantly expressed in a restricted set of immune cell types, including pDCs (plasmacytoid dendritic cells), it is also expressed in non-native expressing cells (e.g. hepatocytes) under certain circumstances. To elucidate the molecular basis of TLR7 induction by pro-inflammatory stimulation and the subsequent cellular responses in these non-native TLR7-expressing cell types, we first cloned and characterized the 5'-promoter region of TLR7. The proximal region of this promoter drives the transcription of the TLR7 gene. Pro-inflammatory stimuli activated TLR 7 transcription via a NF-kappaB (nuclear factor kappaB)-binding motif in this region, and this activation could be blocked by mutation of the NF-kappaB binding site or addition of NF-kappaB inhibitors. Further studies showed that pretreatment of the Hep3B hepatocytes with TNF-alpha (tumour necrosis factor-alpha) or IL-1 (interleukin-1) rendered them responsive to TLR7 activation by a TLR7 agonist. However, distinct from TLR7 activation in pDCs, which respond to stimulation with Th1 polarized cytokine production, TLR7 induction by pro-inflammatory signals in hepatocytes reconstitutes the NF-kappaB-dependent cascade but not the IRF7 (interferon regulatory factor 7)-dependent cascade, resulting in a pro-inflammatory polarized response rather than a Th1 polarized response. These results indicate that inflammatory stimulation is capable of priming cells to respond to TLR7 agonist with an immune response that differs from that in native TLR7-expressing cells.
Collapse
Affiliation(s)
- Jongdae Lee
- Department of Medicine, University of California San Diego, La Jolla, CA 92093-0663
| | - Masaaki Hayashi
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| | - Jeng-Fan Lo
- Institute of Oral Biology, National Yang-Ming University, No. 155, Section 2, Li-Nung Street, Taipei 11217, Taiwan, ROC
| | - Colleen Fearns
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| | - Wen-Ming Chu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Yunping Luo
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| | - Rong Xiang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
- School of Medicine, University of Nankai, Tianjin 300071, PR China
| | - Tsung-Hsien Chuang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
19
|
Tone Y, Furuuchi K, Kojima Y, Tykocinski ML, Greene MI, Tone M. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat Immunol 2007; 9:194-202. [PMID: 18157133 DOI: 10.1038/ni1549] [Citation(s) in RCA: 634] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 11/08/2007] [Indexed: 12/17/2022]
Abstract
The transcription factor Foxp3 is involved in the differentiation, function and survival of CD4+CD25+ regulatory T (T(reg)) cells. Details of the mechanism underlying the induction of Foxp3 expression remain unknown, because studies of the transcriptional regulation of the Foxp3 gene are limited by the small number of T(reg) cells in mononuclear cell populations. Here we have generated a model system for analyzing Foxp3 induction and, by using this system with primary T cells, we have identified an enhancer element in this gene. The transcription factors Smad3 and NFAT are required for activity of this Foxp3 enhancer, and both factors are essential for histone acetylation in the enhancer region and induction of Foxp3. These biochemical properties that define Foxp3 expression explain many of the effects of transforming growth factor-beta on the function of Foxp3+ T(reg) cells.
Collapse
Affiliation(s)
- Yukiko Tone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|