1
|
The function of myeloid-derived suppressor cells in COVID-19 lymphopenia. Int Immunopharmacol 2022; 112:109277. [PMID: 36206651 PMCID: PMC9513342 DOI: 10.1016/j.intimp.2022.109277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/02/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has caused a global pandemic and presents a significant danger to public health. Lymphopenia is considered to be the defining characteristic of severe COVID-19, especially in elderly people. Lymphopenia has been suggested as a pivotal factor in disease severity. To minimize mortality in COVID-19 patients, it is essential to have a deeper understanding of the processes behind lymphocytopenia. Recently, myeloid-derived suppressor cells (MDSCs) have been confirmed as a key mediator of lymphopenia. MDSCs are characterized by their powerful capacity to suppress T cells and eventually contribute to the course of illness. Targeting these cells may improve the disease prognosis. In this article, we analyze the available research on MDSCs in lymphopenia and discuss their immunopathologic changes and prospective therapeutic targets in patients with COVID-19 lymphocytopenia.
Collapse
|
2
|
Park SJ, Nam DE, Seong HC, Hahn YS. New Discovery of Myeloid-Derived Suppressor Cell's Tale on Viral Infection and COVID-19. Front Immunol 2022; 13:842535. [PMID: 35185933 PMCID: PMC8850309 DOI: 10.3389/fimmu.2022.842535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 01/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are generated under biological stress such as cancer, inflammatory tissue damage, and viral infection. In recent years, with occurrence of global infectious diseases, new discovery on MDSCs functions has been significantly expanded during viral infection and COVID-19. For a successful viral infection, pathogens viruses develop immune evasion strategies to avoid immune recognition. Numerous viruses induce the differentiation and expansion of MDSCs in order to suppress host immune responses including natural killer cells, antigen presenting cells, and T-cells. Moreover, MDSCs play an important role in regulation of immunopathogenesis by balancing viral infection and tissue damage. In this review article, we describe the overview of immunomodulation and genetic regulation of MDSCs during viral infection in the animal model and human studies. In addition, we include up-to-date review of role of MDSCs in SARS-CoV-2 infection and COVID-19. Finally, we discuss potential therapeutics targeting MDSCs.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Da-eun Nam
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Hae Chang Seong
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
3
|
DNA Methylation as a Therapeutic Target for Bladder Cancer. Cells 2020; 9:cells9081850. [PMID: 32784599 PMCID: PMC7463638 DOI: 10.3390/cells9081850] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer (BC) is the tenth most frequent cancer worldwide and is associated with high mortality when diagnosed in its most aggressive form, which is not reverted by the current treatment options. Thus, the development of new therapeutic strategies, either alternative or complementary to the current ones, is of major importance. The disruption of normal epigenetic mechanisms, namely, DNA methylation, is a known early event in cancer development. Consequently, DNA methyltransferase (DNMT) inhibitors constitute a promising therapeutic target for the treatment of BC. Although these inhibitors, mainly nucleoside analogues such as 5-azacytidine (5-aza) and decitabine (DAC), cause re-expression of tumor suppressor genes, inhibition of tumor cell growth, and increased apoptosis in BC experimental models and clinical trials, they also show important drawbacks that prevent their use as a valuable option for the treatment of BC. However, their combination with chemotherapy and/or immune-checkpoint inhibitors could aid in their implementation in the clinical practice. Here, we provide a comprehensive review of the studies exploring the effects of DNA methylation inhibition using DNMTs inhibitors in BC, from in vitro and in vivo studies to clinical trials.
Collapse
|
4
|
Zou Y, Hu X, Schewitz-Bowers LP, Stimpson M, Miao L, Ge X, Yang L, Li Y, Bible PW, Wen X, Li JJ, Liu Y, Lee RWJ, Wei L. The DNA Methylation Inhibitor Zebularine Controls CD4 + T Cell Mediated Intraocular Inflammation. Front Immunol 2019; 10:1950. [PMID: 31475011 PMCID: PMC6706956 DOI: 10.3389/fimmu.2019.01950] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/01/2019] [Indexed: 01/13/2023] Open
Abstract
CD4+ T cell mediated uveitis is conventionally treated with systemic immunosuppressive agents, including corticosteroids and biologics targeting key inflammatory cytokines. However, their long-term utility is limited due to various side effects. Here, we investigated whether DNA methylation inhibitor zebularine can target CD4+ T cells and control intraocular inflammation. Our results showed that zebularine restrained the expression of inflammatory cytokines IFN-γ and IL-17 in both human and murine CD4+ T cells in vitro. Importantly, it also significantly alleviated intraocular inflammation and retinal tissue damage in the murine experimental autoimmune uveitis (EAU) model in vivo, suggesting that the DNA methylation inhibitor zebularine is a candidate new therapeutic agent for uveitis.
Collapse
Affiliation(s)
- Yanli Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiao Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lauren P Schewitz-Bowers
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom.,National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Madeleine Stimpson
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom.,National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Li Miao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaofei Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Liu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Paul W Bible
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jing Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Richard W J Lee
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom.,National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Andrade AF, Borges KS, Suazo VK, Geron L, Corrêa CAP, Castro-Gamero AM, de Vasconcelos EJR, de Oliveira RS, Neder L, Yunes JA, Dos Santos Aguiar S, Scrideli CA, Tone LG. The DNA methyltransferase inhibitor zebularine exerts antitumor effects and reveals BATF2 as a poor prognostic marker for childhood medulloblastoma. Invest New Drugs 2016; 35:26-36. [PMID: 27785591 DOI: 10.1007/s10637-016-0401-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/20/2016] [Indexed: 12/11/2022]
Abstract
Medulloblastoma (MB) is the most common solid tumor among pediatric patients and corresponds to 20 % of all pediatric intracranial tumors in this age group. Its treatment currently involves significant side effects. Epigenetic changes such as DNA methylation may contribute to its development and progression. DNA methyltransferase (DNMT) inhibitors have shown promising anticancer effects. The agent Zebularine acts as an inhibitor of DNA methylation and shows low toxicity and high efficacy, being a promising adjuvant agent for anti-cancer chemotherapy. Several studies have reported its effects on different types of tumors; however, there are no studies reporting its effects on MB. We analyzed its potential anticancer effects in four pediatric MB cell lines. The treatment inhibited proliferation and clonogenicity, increased the apoptosis rate and the number of cells in the S phase (p < 0.05), as well as the expression of p53, p21, and Bax, and decreased cyclin A, Survivin and Bcl-2 proteins. In addition, the combination of zebularine with the chemotherapeutic agents vincristine and cisplatin resulted in synergism and antagonism, respectively. Zebularine also modulated the activation of the SHH pathway, reducing SMO and GLI1 levels and one of its targets, PTCH1, without changing SUFU levels. A microarray analysis revealed different pathways modulated by the drug, including the Toll-Like Receptor pathway and high levels of the BATF2 gene. The low expression of this gene was associated with a worse prognosis in MB. Taken together, these data suggest that Zebularine may be a potential drug for further in vivo studies of MB treatment.
Collapse
Affiliation(s)
- Augusto Faria Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil.
| | - Kleiton Silva Borges
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Veridiana Kiill Suazo
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Lenisa Geron
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil
| | | | | | | | | | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, USP, São Paulo, Brazil
| | | | | | - Carlos Alberto Scrideli
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Luiz Gonzaga Tone
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil.,Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
6
|
Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol 2015; 11:363-75. [DOI: 10.1586/1744666x.2015.1002397] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
7
|
Goh C, Narayanan S, Hahn YS. Myeloid-derived suppressor cells: the dark knight or the joker in viral infections? Immunol Rev 2014; 255:210-21. [PMID: 23947357 DOI: 10.1111/imr.12084] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are immature cells of myeloid origin, frequently found in tumor microenvironments and in the blood of cancer patients. In recent years, MDSCs have also been found in non-cancer settings, including a number of viral infections. The evasion of host immunity employed by viruses to establish viral persistence strikingly parallels mechanisms of tumor escape, prompting investigations into the generation and function of MDSCs in chronic viral infections. Importantly, analogous to the tumor microenvironment, MDSCs effectively suppress antiviral host immunity by limiting the function of several immune cells including T cells, natural killer cells, and antigen-presenting cells. In this article, we review studies on the mechanisms of MDSC generation, accumulation, and survival in an effort to understand their emergent importance in viral infections. We include a growing list of viral infections in which MDSCs have been reported. Finally, we discuss how MDSCs might play a role in establishing chronic viral infections and identify potential therapeutics that target MDSCs.
Collapse
Affiliation(s)
- Celeste Goh
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
8
|
Gomez A, Ingelman-Sundberg M. Pharmacoepigenetic aspects of gene polymorphism on drug therapies: effects of DNA methylation on drug response. Expert Rev Clin Pharmacol 2014; 2:55-65. [DOI: 10.1586/17512433.2.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Nittby H, Ericsson P, Förnvik K, Strömblad S, Jansson L, Xue Z, Skagerberg G, Widegren B, Sjögren HO, Salford LG. Zebularine induces long-term survival of pancreatic islet allotransplants in streptozotocin treated diabetic rats. PLoS One 2013; 8:e71981. [PMID: 23991016 PMCID: PMC3753325 DOI: 10.1371/journal.pone.0071981] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 07/05/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Coping with the immune rejection of allotransplants or autologous cells in patients with an active sensitization towards their autoantigens and autoimmunity presently necessitates life-long immune suppressive therapy acting on the immune system as a whole, which makes the patients vulnerable to infections and increases their risk of developing cancer. New technologies to induce antigen selective long-lasting immunosuppression or immune tolerance are therefore much needed. METHODOLOGY/PRINCIPAL FINDINGS The DNA demethylating agent Zebularine, previously demonstrated to induce expression of the genes for the immunosuppressive enzymes indolamine-2,3-deoxygenase-1 (IDO1) and kynureninase of the kynurenine pathway, is tested for capacity to suppress rejection of allotransplants. Allogeneic pancreatic islets from Lewis rats were transplanted under the kidney capsule of Fischer rats previously made diabetic by a streptozotocin injection (40 mg/kg). One group was treated with Zebularine (225 mg/kg) daily for 14 days from day 6 or 8 after transplantation, and a control group received no further treatment. Survival of the transplants was monitored by blood sugar measurements. Rats, normoglycemic for 90 days after allografting, were subjected to transplant removal by nephrectomy to confirm whether normoglycemia was indeed due to a surviving insulin producing transplant, or alternatively was a result of recovery of pancreatic insulin production in some toxin-treated rats. Of 9 Zebularine treated rats, 4 were still normoglycemic after 90 days and became hyperglycemic after nephrectomy. The mean length of normoglycemia in the Zebularine group was 67±8 days as compared to 14±3 days in 9 controls. Seven rats (2 controls and 5 Zebularine treated) were normoglycemic at 90 days due to pancreatic recovery as demonstrated by failure of nephrectomy to induce hyperglycemia. CONCLUSIONS/SIGNIFICANCE Zebularine treatment in vivo induces a long-lasting suppression of the immune destruction of allogeneic pancreatic islets resulting in protection of allograft function for more than 10 weeks after end of treatment.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/surgery
- Gene Expression Regulation, Enzymologic/drug effects
- Graft Survival/drug effects
- Graft Survival/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/methods
- Male
- Rats
- Rats, Inbred F344
- Rats, Inbred Lew
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/drug effects
- Spleen/metabolism
- Time Factors
- Transplantation, Homologous
- Treatment Outcome
Collapse
Affiliation(s)
- Henrietta Nittby
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
- * E-mail:
| | - Peter Ericsson
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Karolina Förnvik
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Susanne Strömblad
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Linda Jansson
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Zhongtian Xue
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Gunnar Skagerberg
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Bengt Widegren
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Hans-Olov Sjögren
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Leif G. Salford
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
10
|
Naeem N, Haneef K, Kabir N, Iqbal H, Jamall S, Salim A. DNA methylation inhibitors, 5-azacytidine and zebularine potentiate the transdifferentiation of rat bone marrow mesenchymal stem cells into cardiomyocytes. Cardiovasc Ther 2013; 31:201-209. [PMID: 22954287 DOI: 10.1111/j.1755-5922.2012.00320.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have immense self-renewal capability. They can be differentiated into many cell types and therefore hold great potential in the field of regenerative medicine. MSCs can be converted into beating cardiomyocytes by treating them with DNA-demethylating agents. Some of these compounds are nucleoside analogs that are widely used for studying the role of DNA methylation in biological processes as well as for the clinical treatment of leukemia and other carcinomas. AIMS To achieve a better therapeutic option for cardiovascular regeneration, this study was carried out using MSCs treated with two synthetic compounds, zebularine and 5-azacytidine. It can be expected that treated MSCs prior to transplantation may increase the likelihood of successful regeneration of damaged myocardium. METHODS The optimized concentrations of these compounds were added separately into the culture medium and the treated cells were analyzed for the expression of cardiac-specific genes by RT-PCR and cardiac-specific proteins by immunocytochemistry and flow cytometry. Treated MSCs were cocultured with cardiomyocytes to see the fusion capability of these cells. RESULTS mRNA and protein expressions of GATA4, Nkx2.5, and cardiac troponin T were observed in the treated MSCs. Coculture studies of MSCs and cardiomyocytes have shown improved fusion with zebularine-treated MSCs as compared to untreated and 5-azacytidine-treated MSCs. CONCLUSION The study is expected to put forth another valuable aspect of certain compounds, that is, induction of transdifferentiation of MSCs into cardiomyocytes. This would serve as a tool for modified cellular therapy and may increase the probability of better myocardial regeneration.
Collapse
Affiliation(s)
- Nadia Naeem
- Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | | | | | | | | | | |
Collapse
|
11
|
Xiong X, Lan D, Li J, Zhong J, Zi X, Ma L, Wang Y. Zebularine and scriptaid significantly improve epigenetic reprogramming of yak fibroblasts and cloning efficiency. Cell Reprogram 2013; 15:293-300. [PMID: 23790013 DOI: 10.1089/cell.2012.0092] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Abnormal epigenetic reprogramming of the donor nucleus after somatic cell nuclear transfer (SCNT) is thought to be the main cause of low cloning efficiency. Following SCNT, the donor nucleus often fails to express early embryonic genes and establish a normal embryonic pattern of chromatin modification. Therefore, in this study, we have attempted to improve epigenetic reprogramming of the donor nucleus and cloned embryos with Zebularine and Scriptaid. Yak fibroblasts were treated with 20 μM Zebularine alone or 20 μM Zebularine plus 0.5 μM Scriptaid for 24 h, whereas yak cloned embryos were treated exclusively with 0.5 μM Scriptaid for 12 h. There was no effect on cellular viability and proliferation after drug treatment. The treatment of fibroblasts with Zebularine or Zebularine plus Scriptaid increased histone acetylation of histone 3 lysine 9 (H3K9), but decreased the level of DNA methylation of Oct-4 and Sox-2 promoter regions. When donor cells were used after Zebularine plus Scriptaid treatment to reconstruct cloned embryos and then treated with Scriptaid, the developmental competence and cryosurvival of embryos were improved significantly. In addition, the relative expression of Oct-4 and Sox-2 were increased significantly. The expression levels of Dnmt-1 and Hdac-1 were significantly decreased when fibroblasts and cloned embryos were treated with Zebularine or Scriptaid. This work provides functional evidence that treatment with Zebularine and Scriptaid modifies the epigenetic status of yak fibroblasts, subsequently enhancing in vitro developmental potential and the quality of yak cloned embryos.
Collapse
Affiliation(s)
- Xianrong Xiong
- College of Life Science and Technology, Southwest University for Nationalities, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Wainwright DA, Dey M, Chang A, Lesniak MS. Targeting Tregs in Malignant Brain Cancer: Overcoming IDO. Front Immunol 2013; 4:116. [PMID: 23720663 PMCID: PMC3654236 DOI: 10.3389/fimmu.2013.00116] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/30/2013] [Indexed: 01/01/2023] Open
Abstract
One of the hallmark features of glioblastoma multiforme (GBM), the most common adult primary brain tumor with a very dismal prognosis, is the accumulation of CD4+CD25+Foxp3+ regulatory T cells (Tregs). Regulatory T cells (Tregs) segregate into two primary categories: thymus-derived natural Tregs (nTregs) that develop from the interaction between immature T cells and thymic epithelial stromal cells, and inducible Tregs (iTregs) that arise from the conversion of CD4+FoxP3− T cells into FoxP3 expressing cells. Normally, these Treg subsets complement one another’s actions by maintaining tolerance of self-antigens, thereby suppressing autoimmunity, while also enabling effective immune responses toward non-self-antigens, thus promoting infectious protection. However, Tregs have also been shown to be associated with the promotion of pathological outcomes, including cancer. In the setting of GBM, nTregs appear to be primary players that contribute to immunotherapeutic failure, ultimately leading to tumor progression. Several attempts have been made to therapeutically target these cells with variable levels of success. The blood brain barrier-crossing chemotherapeutics, temozolomide, and cyclophosphamide (CTX), vaccination against the Treg transcriptional regulator, FoxP3, as well as mAbs against Treg-associated cell surface molecules CD25, CTLA-4, and GITR are all different therapeutic approaches under investigation. Contributing to the poor success of past approaches is the expression of indoleamine 2,3-dioxygenase 1 (IDO), a tryptophan catabolizing enzyme overexpressed in GBM, and critically involved in regulating tumor-infiltrating Treg levels. Herein, we review the current literature on Tregs in brain cancer, providing a detailed phenotype, causative mechanisms involved in their pathogenesis, and strategies that have been used to target this population, therapeutically.
Collapse
|
13
|
Xue ZT, Sjögren HO, Salford LG, Widegren B. An epigenetic mechanism for high, synergistic expression of indoleamine 2,3-dioxygenase 1 (IDO1) by combined treatment with zebularine and IFN-γ: potential therapeutic use in autoimmune diseases. Mol Immunol 2012; 51:101-11. [PMID: 22424783 DOI: 10.1016/j.molimm.2012.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/14/2012] [Indexed: 12/21/2022]
Abstract
IDO1 can be induced by interferon gamma (IFN-γ) in multiple cell types. We have earlier described that the DNA methyltransferase inhibitor zebularine also induces IDO1 in several rat cell clones. We now describe a synergistic induction of IDO1 expression by IFN-γ and zebularine. To elucidate the mechanism of the IDO1 induction we have studied the methylation status in the promoter region of the IDO1 gene from both human monocytic THP-1 cells and H1D2 rat colon cancer cells. Interestingly, the IDO1 promoter is hypermethylated and IFN-γ is shown to induce a significant demethylation. The synergism in effect of zebularine and IFN-γ on IDO1 expression is paralleled by a similar synergistic effect on expression of two other IFN-γ-responsive genes, the transcription factors STAT1 and IRF1 with binding sites in the IDO1 promoter region. The demonstrated synergistic activation of IDO1 expression has implications in relation to therapeutic induction of immunosuppression in autoimmunity and chronic inflammation.
Collapse
Affiliation(s)
- Zhong-Tian Xue
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
14
|
Jia W, Jackson-Cook C, Graf MR. Tumor-infiltrating, myeloid-derived suppressor cells inhibit T cell activity by nitric oxide production in an intracranial rat glioma + vaccination model. J Neuroimmunol 2010; 223:20-30. [PMID: 20452681 PMCID: PMC2883008 DOI: 10.1016/j.jneuroim.2010.03.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 02/19/2010] [Accepted: 03/15/2010] [Indexed: 12/31/2022]
Abstract
In rats bearing an intracranial T9 glioma, immunization with tumor antigens induces myeloid suppressor cells, which express neutrophil (His48) and monocyte (CD11bc) markers, to infiltrate the tumors. The His48(+)/CD11bc(+) cells were not derived from CNS microglia but were hematogenous; suppressed multiple T cell effector functions; and are myeloid-derived suppressor cells (MDSC). The glioma-infiltrating MDSC expressed arginase I, iNOS, indoleamine 2,3-dioxygenase and TGF-beta; however, inhibitor/blocking studies demonstrated that NO production was the primary mechanism of suppression which induced T cell apoptosis. These findings suggest that neuro-immunomodulation by MDSC in rat gliomas maybe mediated by a pathway requiring NO production.
Collapse
Affiliation(s)
- Wentao Jia
- Department of Neurosurgery - Harold F. Young Neurosurgical Center and the Massey Cancer Center, Virginia Commonwealth, P.O. Box 980631, Richmond, VA, 23298-0631, USA
| | - Colleen Jackson-Cook
- Department of Pathology, Virginia Commonwealth University Medical Center, P.O. Box 980662, Richmond, VA, 23298-0662, USA
| | - Martin R. Graf
- Department of Neurosurgery - Harold F. Young Neurosurgical Center and the Massey Cancer Center, Virginia Commonwealth, P.O. Box 980631, Richmond, VA, 23298-0631, USA
| |
Collapse
|
15
|
T cell regulatory plasmacytoid dendritic cells expressing indoleamine 2,3 dioxygenase. Handb Exp Pharmacol 2009:165-96. [PMID: 19031026 DOI: 10.1007/978-3-540-71029-5_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mature dendritic cells (DCs) are potent stimulators of T cells that recognize antigens presented by the DCs. In this chapter we describe mature DCs that suppress T cell responses to antigens they present due to expression of the intracellular enzyme indoleamine 2,3 dioxygenase (IDO). IDO-competent DCs are a subset of plasmacytoid DCs that can be induced to express IDO under certain inflammatory conditions in humans and mice. Though rare, IDO-expressing DCs acquire potent T cell suppressor activity that may predominate over the T cell stimulatory functions of all other antigen-presenting cells in physiologic environments due in part, to cooperation with regulatory T cells. Thus, IDO-expressing DCs are critical regulators of adaptive immunity that contribute to a wide range of inflammatory disease processes. As such, manipulating IDO expression in DCs using IDO inhibitors or IDO inducers offers considerable opportunities to improve immunotherapies in a range of clinically-significant disease syndromes.
Collapse
|