1
|
Jeremiah N, Ferran H, Antoniadou K, De Azevedo K, Nikolic J, Maurin M, Benaroch P, Manel N. RELA tunes innate-like interferon I/III responses in human T cells. J Exp Med 2023; 220:e20220666. [PMID: 36820829 PMCID: PMC9998965 DOI: 10.1084/jem.20220666] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 11/11/2022] [Accepted: 01/10/2023] [Indexed: 02/24/2023] Open
Abstract
In innate immune cells, intracellular sensors such as cGAS-STING stimulate type I/III interferon (IFN) expression, which promotes antiviral defense and immune activation. However, how IFN-I/III expression is controlled in adaptive cells is poorly understood. Here, we identify a transcriptional rheostat orchestrated by RELA that confers human T cells with innate-like abilities to produce IFN-I/III. Despite intact cGAS-STING signaling, IFN-I/III responses are stunted in CD4+ T cells compared with dendritic cells or macrophages. We find that lysine residues in RELA tune the IFN-I/III response at baseline and in response to STING stimulation in CD4+ T cells. This response requires positive feedback driven by cGAS and IRF7 expression. By combining RELA with IRF3 and DNA demethylation, IFN-I/III production in CD4+ T cells reaches levels observed in dendritic cells. IFN-I/III production provides self-protection of CD4+ T cells against HIV infection and enhances the elimination of tumor cells by CAR T cells. Therefore, innate-like functions can be tuned and leveraged in human T cells.
Collapse
Affiliation(s)
- Nadia Jeremiah
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Hermine Ferran
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Konstantina Antoniadou
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Kevin De Azevedo
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Jovan Nikolic
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Mathieu Maurin
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Philippe Benaroch
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Nicolas Manel
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| |
Collapse
|
2
|
Srinivas N, Song L, Lei KC, Gravemeyer J, Furtmann F, Gambichler T, Becker JC, Sriram A. The HDAC inhibitor domatinostat induces type I interferon α in Merkel cell carcinoma by HES1 repression. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04733-y. [PMID: 37071208 PMCID: PMC10374800 DOI: 10.1007/s00432-023-04733-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Class I selective histone deacetylase inhibitors (HDACi) have been previously demonstrated to not only increase major histocompatibility complex class I surface expression in Merkel cell carcinoma (MCC) cells by restoring the antigen processing and presentation machinery, but also exert anti-tumoral effect by inducing apoptosis. Both phenomena could be due to induction of type I interferons (IFN), as has been described for HDACi. However, the mechanism of IFN induction under HDACi is not fully understood because the expression of IFNs is regulated by both activating and inhibitory signaling pathways. Our own preliminary observations suggest that this may be caused by suppression of HES1. METHODS The effect of the class I selective HDACi domatinostat and IFNα on cell viability and the apoptosis of MCPyV-positive (WaGa, MKL-1) and -negative (UM-MCC 34) MCC cell lines, as well as, primary fibroblasts were assessed by colorimetric methods or measuring mitochondrial membrane potential and intracellular caspase-3/7, respectively. Next, the impact of domatinostat on IFNA and HES1 mRNA expression was measured by RT-qPCR; intracellular IFNα production was detected by flow cytometry. To confirm that the expression of IFNα induced by HDACi was due to the suppression of HES1, it was silenced by RNA interference and then mRNA expression of IFNA and IFN-stimulated genes was assessed. RESULTS Our studies show that the previously reported reduction in viability of MCC cell lines after inhibition of HDAC by domatinostat is accompanied by an increase in IFNα expression, both of mRNA and at the protein level. We confirmed that treatment of MCC cells with external IFNα inhibited their proliferation and induced apoptosis. Re-analysis of existing single-cell RNA sequencing data indicated that induction of IFNα by domatinostat occurs through repression of HES1, a transcriptional inhibitor of IFNA; this was confirmed by RT-qPCR. Finally, siRNA-mediated silencing of HES1 in the MCC cell line WaGa not only increased mRNA expression of IFNA and IFN-stimulated genes but also decreased cell viability. CONCLUSION Our results demonstrate that the direct anti-tumor effect of HDACi domatinostat on MCC cells is at least in part mediated via decreased HES1 expression allowing the induction of IFNα, which in turn causes apoptosis.
Collapse
Affiliation(s)
- Nalini Srinivas
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Lina Song
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kuan Cheok Lei
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Gravemeyer
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frauke Furtmann
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thilo Gambichler
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Jürgen C Becker
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, University Hospital Essen, Essen, Germany.
| | - Ashwin Sriram
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
3
|
Moran B, Davern M, Reynolds JV, Donlon NE, Lysaght J. The impact of histone deacetylase inhibitors on immune cells and implications for cancer therapy. Cancer Lett 2023; 559:216121. [PMID: 36893893 DOI: 10.1016/j.canlet.2023.216121] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/09/2023]
Abstract
Many cancers possess the ability to suppress the immune response to malignant cells, thus facilitating tumour growth and invasion, and this has fuelled research to reverse these mechanisms and re-activate the immune system with consequent important therapeutic benefit. One such approach is to use histone deacetylase inhibitors (HDACi), a novel class of targeted therapies, which manipulate the immune response to cancer through epigenetic modification. Four HDACi have recently been approved for clinical use in malignancies including multiple myeloma and T-cell lymphoma. Most research in this context has focussed on HDACi and tumour cells, however, little is known about their impact on the cells of the immune system. Additionally, HDACi have been shown to impact the mechanisms by which other anti-cancer therapies exert their effects by, for example, increasing accessibility to exposed DNA through chromatin relaxation, impairing DNA damage repair pathways and increasing immune checkpoint receptor expression. This review details the effects of HDACi on immune cells, highlights the variability in these effects depending on experimental design, and provides an overview of clinical trials investigating the combination of HDACi with chemotherapy, radiotherapy, immunotherapy and multimodal regimens.
Collapse
Affiliation(s)
- Brendan Moran
- Cancer Immunology and Immunotherapy Group, Trinity St. James's Cancer Institute, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland; Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Maria Davern
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | - Noel E Donlon
- Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Trinity St. James's Cancer Institute, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland.
| |
Collapse
|
4
|
Nazri JM, Oikonomopoulou K, de Araujo ED, Kraskouskaya D, Gunning PT, Chandran V. Histone deacetylase inhibitors as a potential new treatment for psoriatic disease and other inflammatory conditions. Crit Rev Clin Lab Sci 2023; 60:300-320. [PMID: 36846924 DOI: 10.1080/10408363.2023.2177251] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Collectively known as psoriatic disease, psoriasis and psoriatic arthritis (PsA) are immune-mediated inflammatory diseases in which patients present with cutaneous and musculoskeletal inflammation. Affecting roughly 2-3% of the world's total population, there remains unmet therapeutic needs in both psoriasis and PsA despite the availability of current immunomodulatory treatments. As a result, patients with psoriatic disease often experience reduced quality of life. Recently, a class of small molecules, commonly investigated as anti-cancer agents, called histone deacetylase (HDAC) inhibitors, have been proposed as a new promising anti-inflammatory treatment for immune- and inflammatory-related diseases. In inflammatory diseases, current evidence is derived from studies on diseases like rheumatoid arthritis (RA) and systematic lupus erythematosus (SLE), and while there are some reports studying psoriasis, data on PsA patients are not yet available. In this review, we provide a brief overview of psoriatic disease, psoriasis, and PsA, as well as HDACs, and discuss the rationale behind the potential use of HDAC inhibitors in the management of persistent inflammation to suggest its possible use in psoriatic disease.
Collapse
Affiliation(s)
- Jehan Mohammad Nazri
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | - Elvin D de Araujo
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Canada
| | - Dziyana Kraskouskaya
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Canada
| | - Patrick T Gunning
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Canada.,Department of Chemistry, University of Toronto, Toronto, Canada
| | - Vinod Chandran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Schroeder Arthritis Institute, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Medicine, Memorial University, St. John's, Canada
| |
Collapse
|
5
|
Bouyahya A, El Omari N, Bakha M, Aanniz T, El Menyiy N, El Hachlafi N, El Baaboua A, El-Shazly M, Alshahrani MM, Al Awadh AA, Lee LH, Benali T, Mubarak MS. Pharmacological Properties of Trichostatin A, Focusing on the Anticancer Potential: A Comprehensive Review. Pharmaceuticals (Basel) 2022; 15:ph15101235. [PMID: 36297347 PMCID: PMC9612318 DOI: 10.3390/ph15101235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/12/2022] [Accepted: 09/23/2022] [Indexed: 11/05/2022] Open
Abstract
Trichostatin A (TSA), a natural derivative of dienohydroxamic acid derived from a fungal metabolite, exhibits various biological activities. It exerts antidiabetic activity and reverses high glucose levels caused by the downregulation of brain-derived neurotrophic factor (BDNF) expression in Schwann cells, anti-inflammatory activity by suppressing the expression of various cytokines, and significant antioxidant activity by suppressing oxidative stress through multiple mechanisms. Most importantly, TSA exhibits potent inhibitory activity against different types of cancer through different pathways. The anticancer activity of TSA appeared in many in vitro and in vivo investigations that involved various cell lines and animal models. Indeed, TSA exhibits anticancer properties alone or in combination with other drugs used in chemotherapy. It induces sensitivity of some human cancers toward chemotherapeutical drugs. TSA also exhibits its action on epigenetic modulators involved in cell transformation, and therefore it is considered an epidrug candidate for cancer therapy. Accordingly, this work presents a comprehensive review of the most recent developments in utilizing this natural compound for the prevention, management, and treatment of various diseases, including cancer, along with the multiple mechanisms of action. In addition, this review summarizes the most recent and relevant literature that deals with the use of TSA as a therapeutic agent against various diseases, emphasizing its anticancer potential and the anticancer molecular mechanisms. Moreover, TSA has not been involved in toxicological effects on normal cells. Furthermore, this work highlights the potential utilization of TSA as a complementary or alternative medicine for preventing and treating cancer, alone or in combination with other anticancer drugs.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Mohamed Bakha
- Unit of Plant Biotechnology and Sustainable Development of Natural Resources “B2DRN”, Polydisciplinary Faculty of Beni Mellal, Sultan Moulay Slimane University, Mghila, P.O. Box 592, Beni Mellal 23000, Morocco
| | - Tarik Aanniz
- Medical Biotechnology Laboratory, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Rabat B.P. 6203, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, Imouzzer Road Fez, Fez 30050, Morocco
| | - Aicha El Baaboua
- Biotechnology and Applied Microbiology Team, Department of Biology, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan 93000, Morocco
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo 11566, Egypt
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ahmed Abdullah Al Awadh
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| | - Taoufiq Benali
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
| | - Mohammad S. Mubarak
- Department of Chemistry, The University of Jordan, Amma 11942, Jordan
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| |
Collapse
|
6
|
Inhibition of Class I Histone Deacetylase Activity Blocks the Induction of TNFAIP3 Both Directly and Indirectly via the Suppression of Endogenous TNF-α. Int J Mol Sci 2022; 23:ijms23179752. [PMID: 36077149 PMCID: PMC9456523 DOI: 10.3390/ijms23179752] [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: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Histone deacetylase inhibitors (HDIs) are promising drugs for the treatment of inflammatory diseases. However, their therapeutical exploitation is slowed down by severe adverse manifestations that can hardly be foreseen, mainly due to incomplete knowledge of how HDIs impact the delicate balance of inflammatory mediators. In this work, we characterized the effects of the HDI trichostatin A (TSA) on the expression of TNFAIP3, which is a crucial inhibitor of the classical NF-kB pathway and an LPS-induced negative feedback regulator. The accumulation of TNFAIP3 mRNA after LPS stimulation showed biphasic behavior, with one wave within the first hour of stimulation and a second wave several hours later, which were both reduced by TSA. By using inhibition and knockdown approaches, we identified two temporally and mechanistically distinct modes of action. The first wave of TNAIP3 accumulation was directly blunted by the histone deacetylase (HDAC) blockade. By contrast, the second wave was decreased mainly because of the lack of endogenous TNF-α induction, which, in turn, depended on the intact HDAC activity. In both cases, class I HDACs appeared to play a nonredundant role, with HDAC3 required, but not sufficient, for TNF-α and TNFAIP3 induction. In addition to TNFAIP3, TNF-α is known to induce many response genes that orchestrate the inflammatory cascade. Thus, suppression of TNF-α may represent a general mechanism through which HDIs regulate a selected set of target genes.
Collapse
|
7
|
Wu S, Yin Y, Wang X. The epigenetic regulation of the germinal center response. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194828. [PMID: 35643396 DOI: 10.1016/j.bbagrm.2022.194828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/22/2022] [Indexed: 06/15/2023]
Abstract
In response to T-cell-dependent antigens, antigen-experienced B cells migrate to the center of the B-cell follicle to seed the germinal center (GC) response after cognate interactions with CD4+ T cells. These GC B cells eventually mature into memory and long-lived antibody-secreting plasma cells, thus generating long-lived humoral immunity. Within GC, B cells undergo somatic hypermutation of their B cell receptors (BCR) and positive selection for the emergence of high-affinity antigen-specific B-cell clones. However, this process may be dangerous, as the accumulation of aberrant mutations could result in malignant transformation of GC B cells or give rise to autoreactive B cell clones that can cause autoimmunity. Because of this, better understanding of GC development provides diagnostic and therapeutic clues to the underlying pathologic process. A productive GC response is orchestrated by multiple mechanisms. An emerging important regulator of GC reaction is epigenetic modulation, which has key transcriptional regulatory properties. In this review, we summarize the current knowledge on the biology of epigenetic mechanisms in the regulation of GC reaction and outline its importance in identification of immunotherapy decision making.
Collapse
Affiliation(s)
- Shusheng Wu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuye Yin
- Department of Immunology, State Key Laboratory of Reproductive Medicine, NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Wang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Yadav K, Singh D, Singh MR, Minz S, Sahu KK, Kaurav M, Pradhan M. Dermal nanomedicine: Uncovering the ability of nucleic acid to alleviate autoimmune and other related skin disorders. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Gkoutsias A, Makis A. The role of epigenetics in childhood autoimmune diseases with hematological manifestations. Pediatr Investig 2022; 6:36-46. [PMID: 35382418 PMCID: PMC8960932 DOI: 10.1002/ped4.12309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022] Open
Abstract
Autoimmune diseases with hematological manifestations are often characterized by chronicity and relapses despite treatment, and the underlying pathogenetic mechanisms remain unknown. Epigenetic alterations play a vital role in the deregulation of immune tolerance and the development of autoimmune diseases. In recent years, study of epigenetic mechanisms in both adult and childhood autoimmune disorders has been seeking to explain the pathophysiology of these heterogeneous diseases and to elucidate the interaction between genetic and environmental factors. Various mechanisms, including DNA methylation, histone modifications (chromatin remodeling), and noncoding RNAs (ncRNAs), have been studied extensively in the context of autoimmune diseases. This paper summarizes the epigenetic patterns in some of the most common childhood autoimmune disorders with hematological manifestations, based on epigenetic studies in children with primary immune thrombocytopenia (ITP), systemic lupus erythematosus (SLE), and juvenile idiopathic arthritis (JIA). Research findings indicate that methylation changes in genes expressed on T cells, modifications at a variety of histone sites, and alterations in the expression of several ncRNAs are involved in the pathogenesis of these diseases. These mechanisms not only determine the development of these diseases but also affect the severity of the clinical presentation and biochemical markers. Further studies will provide new tools for the prevention and diagnosis of childhood autoimmune disorders, and possible novel treatment options.
Collapse
Affiliation(s)
- Athanasios Gkoutsias
- Department of PediatricsFaculty of MedicineSchool of Health SciencesUniversity of IoanninaIoanninaGreece
| | - Alexandros Makis
- Department of PediatricsFaculty of MedicineSchool of Health SciencesUniversity of IoanninaIoanninaGreece
| |
Collapse
|
10
|
Histone deacetylase 3 contributes to the antiviral innate immunity of macrophages by interacting with FOXK1 to regulate STAT1/2 transcription. Cell Rep 2022; 38:110302. [PMID: 35081346 DOI: 10.1016/j.celrep.2022.110302] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 11/16/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that interferon (IFN)-α/-β activates the JAK/STAT signaling pathway and suppresses viral replication through the induction of IFN stimulated genes (ISGs). Here, we report that knockout of HDAC3 from macrophages results in the decreased expression of STAT1 and STAT2, leading to defective antiviral immunity in cells and mice. Further studies show that HDAC3 interacts with a conserved transcription factor Forkhead Box K1 (FOXK1), co-localizes with FOXK1 at the promoter of STAT1 and STAT2, and is required for protecting FOXK1 from lysosomal system-mediated degradation. FOXK1-deficient macrophages also show low STAT1 and STAT2 expression with defective responses to viruses. Thus, our studies uncover the biological importance of HDAC3 in regulating the antiviral immunity of macrophages through interacting with FOXK1 to regulate the expression of STAT1 and STAT2.
Collapse
|
11
|
Peng X, Liao G, Sun P, Yu Z, Chen J. An Overview of HDAC Inhibitors and their Synthetic Routes. Curr Top Med Chem 2019; 19:1005-1040. [DOI: 10.2174/1568026619666190227221507] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/19/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Epigenetics play a key role in the origin, development and metastasis of cancer. Epigenetic processes include DNA methylation, histone acetylation, histone methylation, and histone phosphorylation, among which, histone acetylation is the most common one that plays important roles in the regulation of normal cellular processes, and is controlled by histone deacetylases (HDACs) and histone acetyltransferases (HATs). HDACs are involved in the regulation of many key cellular processes, such as DNA damage repair, cell cycle control, autophagy, metabolism, senescence and chaperone function, and can lead to oncogene activation. As a result, HDACs are considered to be an excellent target for anti-cancer therapeutics like histone deacetylase inhibitors (HDACi) which have attracted much attention in the last decade. A wide-ranging knowledge of the role of HDACs in tumorigenesis, and of the action of HDACi, has been achieved. The primary purpose of this paper is to summarize recent HDAC inhibitors and the synthetic routes as well as to discuss the direction for the future development of new HDAC inhibitors.
Collapse
Affiliation(s)
- Xiaopeng Peng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Guochao Liao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Pinghua Sun
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Lu Y, Stuart JH, Talbot-Cooper C, Agrawal-Singh S, Huntly B, Smid AI, Snowden JS, Dupont L, Smith GL. Histone deacetylase 4 promotes type I interferon signaling, restricts DNA viruses, and is degraded via vaccinia virus protein C6. Proc Natl Acad Sci U S A 2019; 116:11997-12006. [PMID: 31127039 PMCID: PMC6575207 DOI: 10.1073/pnas.1816399116] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Interferons (IFNs) represent an important host defense against viruses. Type I IFNs induce JAK-STAT signaling and expression of IFN-stimulated genes (ISGs), which mediate antiviral activity. Histone deacetylases (HDACs) perform multiple functions in regulating gene expression and some class I HDACs and the class IV HDAC, HDAC11, influence type I IFN signaling. Here, HDAC4, a class II HDAC, is shown to promote type I IFN signaling and coprecipitate with STAT2. Pharmacological inhibition of class II HDAC activity, or knockout of HDAC4 from HEK-293T and HeLa cells, caused a defective response to IFN-α. This defect in HDAC4-/- cells was rescued by reintroduction of HDAC4 or catalytically inactive HDAC4, but not HDAC1 or HDAC5. ChIP analysis showed HDAC4 was recruited to ISG promoters following IFN stimulation and was needed for binding of STAT2 to these promoters. The biological importance of HDAC4 as a virus restriction factor was illustrated by the observations that (i) the replication and spread of vaccinia virus (VACV) and herpes simplex virus type 1 (HSV-1) were enhanced in HDAC4-/- cells and inhibited by overexpression of HDAC4; and (ii) HDAC4 is targeted for proteasomal degradation during VACV infection by VACV protein C6, a multifunctional IFN antagonist that coprecipitates with HDAC4 and is necessary and sufficient for HDAC4 degradation.
Collapse
Affiliation(s)
- Yongxu Lu
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Jennifer H Stuart
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Callum Talbot-Cooper
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Shuchi Agrawal-Singh
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, United Kingdom
| | - Brian Huntly
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, United Kingdom
| | - Andrei I Smid
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Joseph S Snowden
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Liane Dupont
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, CB2 1QP Cambridge, United Kingdom;
| |
Collapse
|
13
|
Parira T, Figueroa G, Granado S, Napuri J, Castillo-Chabeco B, Nair M, Agudelo M. Trichostatin A Shows Transient Protection from Chronic Alcohol-Induced Reactive Oxygen Species (ROS) Production in Human Monocyte-Derived Dendritic Cells. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2018; 6:316. [PMID: 30596124 PMCID: PMC6309403 DOI: 10.4172/2329-6488.1000316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objective of this study was to understand whether histone deacetylase (HDACs) inhibitor Trichostatin A or TSA can block and/or reverse chronic alcohol exposure-induced ROS in human monocyte-derived dendritic cells (MDDCs). Additionally, since nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a known regulator of antioxidant responses, we studied the effects of alcohol and TSA on ROS production and modulation of Nrf2 by MDDCs. METHODS Intra-cellular, extra-cellular, and total ROS levels were measured in MDDCs treated chronically with alcohol (0.1 and 0.2 % EtOH) using 2',7'-dichlorofluorescin diacetate (DCF-DA) followed by detection of ROS in microplate reader and imaging flow cytometer. Nrf2 expression was analyzed by qRT- PCR and western blot. In addition, NFE2L2 (Nrf2), class I HDAC genes HDAC1, HDAC2, and histone acetyltransferase genes KAT5 were analyzed in silico using the GeneMania prediction server. RESULTS Our results confirmed alcohol's ability to increase intracellular ROS levels in MDDCs within minutes of treatment. Our findings have also demonstrated, for the first time, that TSA has a transient protective effect on MDDCs treated chronically with alcohol since the ability of TSA to reduce intracellular ROS levels is only detected up to 15 minutes post-chronic alcohol treatment with no significant protective effects by 10 hours. In addition, chronic alcohol treatment was able to increase the expression of the antioxidant regulator Nrf2 in a dose dependent manner, and the effect of the higher amount of alcohol (0.2%) on Nrf2 gene expression was significantly enhanced by TSA. CONCLUSION This study demonstrates that TSA has a transient protective effect against ROS induced by chronic alcohol exposure of human MDDCs and chronic long-term exposure of MDDCs with alcohol and TSA induces cellular toxicity. It also highlights imaging flow cytometry as a novel tool to detect intracellular ROS levels. Overall, the effect of TSA might be mediated through Nrf2; however, further studies are needed to fully understand the molecular mechanisms.
Collapse
Affiliation(s)
- Tiyash Parira
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Gloria Figueroa
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Sherly Granado
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jacqueline Napuri
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Boris Castillo-Chabeco
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Marisela Agudelo
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| |
Collapse
|
14
|
Li Y, Wu T. Proteomic approaches for novel systemic lupus erythematosus (SLE) drug discovery. Expert Opin Drug Discov 2018; 13:765-777. [DOI: 10.1080/17460441.2018.1480718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
15
|
Salvi V, Gianello V, Busatto S, Bergese P, Andreoli L, D'Oro U, Zingoni A, Tincani A, Sozzani S, Bosisio D. Exosome-delivered microRNAs promote IFN-α secretion by human plasmacytoid DCs via TLR7. JCI Insight 2018; 3:98204. [PMID: 29769437 DOI: 10.1172/jci.insight.98204] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/13/2018] [Indexed: 12/28/2022] Open
Abstract
The excessive production of type I IFNs is a hallmark and a main pathogenic mechanism of many autoimmune diseases, including systemic lupus erythematosus (SLE). In these pathologies, the sustained secretion of type I IFNs is dependent on the improper activation of plasmacytoid DCs (pDCs) by self-nucleic acids. However, the nature and origin of pDC-activating self-nucleic acids is still incompletely characterized. Here, we report that exosomes isolated from the plasma of SLE patients can activate the secretion of IFN-α by human blood pDCs in vitro. This activation requires endosomal acidification and is recapitulated by microRNAs isolated from exosomes, suggesting that exosome-delivered microRNAs act as self-ligands of innate single-stranded endosomal RNA sensors. By using synthetic microRNAs, we identified an IFN induction motif that is responsible for the TLR7-dependent activation, maturation, and survival of human pDCs. These findings identify exosome-delivered microRNAs as potentially novel TLR7 endogenous ligands able to induce pDC activation in SLE patients. Therefore, microRNAs may represent novel pathogenic mediators in the onset of autoimmune reactions and potential therapeutic targets in the treatment of type I IFN-mediated diseases.
Collapse
Affiliation(s)
- Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Veronica Gianello
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Busatto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Italian Center for Colloid and Surface Science - CSGI, Firenze, Italy
| | - Laura Andreoli
- Rheumatology and Clinical Immunology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Alessandra Zingoni
- Department of Molecular Medicine-Pasteur Italia Laboratory, Sapienza University of Rome, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
16
|
Foma AM, Aslani S, Karami J, Jamshidi A, Mahmoudi M. Epigenetic involvement in etiopathogenesis and implications in treatment of systemic lupus erythematous. Inflamm Res 2017; 66:1057-1073. [DOI: 10.1007/s00011-017-1082-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/22/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022] Open
|
17
|
Hedrich CM, Mäbert K, Rauen T, Tsokos GC. DNA methylation in systemic lupus erythematosus. Epigenomics 2017; 9:505-525. [PMID: 27885845 PMCID: PMC6040049 DOI: 10.2217/epi-2016-0096] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/12/2016] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease facilitated by aberrant immune responses directed against cells and tissues, resulting in inflammation and organ damage. In the majority of patients, genetic predisposition is accompanied by additional factors conferring disease expression. While the exact molecular mechanisms remain elusive, epigenetic alterations in immune cells have been demonstrated to play a key role in disease pathogenesis through the dysregulation of gene expression. Since epigenetic marks are dynamic, allowing cells and tissues to differentiate and adjust, they can be influenced by environmental factors and also be targeted in therapeutic interventions. Here, we summarize reports on DNA methylation patterns in SLE, underlying molecular defects and their effect on immune cell function. We discuss the potential of DNA methylation as biomarker or therapeutic target in SLE.
Collapse
Affiliation(s)
- Christian M Hedrich
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katrin Mäbert
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Rauen
- Department of Nephrology & Clinical Immunology, RWTH University Hospital, Aachen, Germany
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Potentiation and tolerance of toll-like receptor priming in human endothelial cells. Transl Res 2017; 180:53-67.e4. [PMID: 27567430 PMCID: PMC5253081 DOI: 10.1016/j.trsl.2016.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 11/21/2022]
Abstract
Repeated challenge of lipopolysaccharide (LPS) alters the response to subsequent LPS exposures via modulation of toll-like receptor 4 (TLR4). Whether activation of other TLRs can modulate TLR4 responses, and vice versa, remains unclear. Specifically with regards to endothelial cells, a key component of innate immunity, the impact of TLR cross-modulation is unknown. We postulated that TLR2 priming (via Pam3Csk4) would inhibit TLR4-mediated responses while TLR3 priming (via Poly I:C) would enhance subsequent TLR4-inflammatory signaling. We studied human umbilical vein endothelial cells (HUVECs) and neonatal human dermal microvascular endothelial cells (HMVECs). Cells were primed with a combination of Poly I:C (10 μg/ml), Pam3Csk4 (10 μg/ml), or LPS (100 ng/ml), then washed and allowed to rest. They were then rechallenged with either Poly I:C, Pam3Csk4 or LPS. Endothelial cells showed significant tolerance to repeated LPS challenge. Priming with Pam3Csk4 also reduced the response to secondary LPS challenge in both cell types, despite a reduced proinflammatory response to Pam3Csk4 in HMVECs compared to HUVECs. Poly I:C priming enhanced inflammatory and interferon producing signals upon Poly I:C or LPS rechallenge, respectively. Poly I:C priming induced interferon regulatory factor 7, leading to enhancement of interferon production. Finally, both Poly I:C and LPS priming induced significant changes in receptor-interacting serine/threonine-protein kinase 1 activity. Pharmacological inhibition of receptor-interacting serine/threonine-protein kinase 1 or interferon regulatory factor 7 reduced the potentiated phenotype of TLR3 priming on TLR4 rechallenge. These results demonstrate that in human endothelial cells, prior activation of TLRs can have a significant impact on subsequent exposures and may contribute to the severity of the host response.
Collapse
|
19
|
Abstract
The luciferase (LUC) reporter assay is commonly used to study gene expression at the transcriptional level. It is convenient, fast, sensitive, inexpensive, and provides quantitative data about small changes in transcription. Signal transducer and activator of transcription 1 (STAT1) is a transcription factor that plays a crucial role in signaling by interferons (IFNs). Here, we describe LUC reporter studies that address the role of histone deacetylase (HDAC) activity in STAT1-dependent gene activation. These experiments include overexpression of HDAC1, HDAC2, HDAC3, and HDAC4 as well as silencing of HDAC1, HDAC2, and HDAC3 through RNA interference in mammalian cancer cells.
Collapse
Affiliation(s)
- Benjamin Y Owusu
- Drug Discovery Division, Department of Oncology, Cancer Program, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA
| | - Lidija Klampfer
- Drug Discovery Division, Department of Oncology, Cancer Program, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| |
Collapse
|
20
|
Gong ZJ. Roles of acetylation-dependent regulation in pathogenesis of chronic hepatitis B virus infection: Potential application as targets for antiviral treatment. Shijie Huaren Xiaohua Zazhi 2016; 24:2778-2784. [DOI: 10.11569/wcjd.v24.i18.2778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years, many studies have confirmed that epigenetics and acetylation-dependent regulation play important roles in the pathogenesis of chronic hepatitis and primary hepatocellular carcinoma. This paper systematically introduces the roles and possible mechanisms of acetylation and its related signal pathways in the pathogenesis of chronic hepatitis B virus (HBV) infection, which may provide a potential novel strategy for the therapy of chronic HBV infection.
Collapse
|
21
|
Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases. Pharmacol Res 2016; 107:27-41. [DOI: 10.1016/j.phrs.2016.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
|
22
|
Breakdown of Immune Tolerance in Systemic Lupus Erythematosus by Dendritic Cells. J Immunol Res 2016; 2016:6269157. [PMID: 27034965 PMCID: PMC4789470 DOI: 10.1155/2016/6269157] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/15/2016] [Accepted: 02/07/2016] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DC) play an important role in the pathogenesis of systemic lupus erythematosus (SLE), an autoimmune disease with multiple tissue manifestations. In this review, we summarize recent studies on the roles of conventional DC and plasmacytoid DC in the development of both murine lupus and human SLE. In the past decade, studies using selective DC depletions have demonstrated critical roles of DC in lupus progression. Comprehensive in vitro and in vivo studies suggest activation of DC by self-antigens in lupus pathogenesis, followed by breakdown of immune tolerance to self. Potential treatment strategies targeting DC have been developed. However, many questions remain regarding the mechanisms by which DC modulate lupus pathogenesis that require further investigations.
Collapse
|
23
|
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of unknown aetiology that can be debilitating and life threatening. As new insights are gained into the underlying pathology of SLE, there have been an unprecedented number of new agents under development to treat the disease via a diverse range of targets. One such class of emerging agents target interferon (IFN) signalling. In this article, we review the preclinical evidence that the inhibition of the secretion and downstream effectors of both IFN-α and IFN-γ may be effective for the treatment of SLE. The primary agents that are currently in clinical development to treat SLE via the targeting of interferon pathways are monoclonal neutralising antibodies (Mab) that bind to and neutralise IFN-γ (AMG 811), IFN-α (sifalimumab, rontalizumab and AGS-009) or its receptor (anifrolumab), and IFN-α kinoid, which is a drug composed of inactivated IFN-α molecules coupled to the keyhole limpet haemocyanin protein. Phase I and II trials have demonstrated acceptable short-term safety with no increase in severe viral infections or reactivation, favourable pharmacokinetic profiles and an inhibition of IFN-associated gene overexpression; however, the impact of these drugs on disease activity must still be assessed in phase III clinical trials. This review concludes with a summary of the challenges that are inherent to this approach to managing SLE.
Collapse
|
24
|
Schmitz ML, de la Vega L. New Insights into the Role of Histone Deacetylases as Coactivators of Inflammatory Gene Expression. Antioxid Redox Signal 2015; 23:85-98. [PMID: 24359078 DOI: 10.1089/ars.2013.5750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE The expression and/or activity of histone deacetylases (HDACs) can be regulated by a variety of environmental conditions, including inflammation and oxidative stress. These events result in diminished or exaggerated protein acetylation, both of which can be causative for many ailments. While the anti-inflammatory activity of HDAC inhibitors (HDACis) is well known, recent studies started unraveling details of the molecular mechanisms underlying the pro-inflammatory function of HDACs. RECENT ADVANCES Recent evidence shows that HDACs are found in association with transcribed regions and ensure proper transcription by maintaining acetylation homeostasis. We also discuss current insights in the molecular mechanisms mediating acetylation-dependent inhibition of pro-inflammatory transcription factors of the NF-κB, HIF-1, IRF, and STAT families. CRITICAL ISSUES The high number of acetylations and the complexity of the regulatory consequences make it difficult to assign biological effects directly to a single acetylation event. The vast majority of acetylated proteins are nonhistone proteins, and it remains to be shown whether the therapeutic effects of HDACis are attributable to altered histone acetylation. FUTURE DIRECTIONS In the traditional view, only exaggerated acetylation is harmful and causative for diseases. Recent data show the relevance of acetylation homeostasis and suggest that both diminished and inflated acetylation can enable the development of ailments. Since acetylation of nonhistone proteins is essential for the induction of a substantial part of the inflammatory gene expression program, HDACis are more than "epigenetic drugs." The identification of substrates for individual HDACs will be the prerequisite for the adequate use of highly specific HDACis.
Collapse
Affiliation(s)
- Michael Lienhard Schmitz
- 1 Medical Faculty, Institute of Biochemistry, Justus-Liebig-University , Giessen, Germany .,2 The German Center for Lung Research, Giessen, Germany
| | - Laureano de la Vega
- 3 Division of Cancer Research, Medical Research Institute, Jacqui Wood Cancer Centre, University of Dundee , Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
25
|
Kroesen M, Gielen P, Brok IC, Armandari I, Hoogerbrugge PM, Adema GJ. HDAC inhibitors and immunotherapy; a double edged sword? Oncotarget 2015; 5:6558-72. [PMID: 25115382 PMCID: PMC4196144 DOI: 10.18632/oncotarget.2289] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Epigenetic modifications, like histone acetylation, are essential for regulating gene expression within cells. Cancer cells acquire pathological epigenetic modifications resulting in gene expression patterns that facilitate and sustain tumorigenesis. Epigenetic manipulation therefore is emerging as a novel targeted therapy for cancer. Histone Acetylases (HATs) and Histone Deacetylases (HDACs) regulate histone acetylation and hence gene expression. Histone deacetylase (HDAC) inhibitors are well known to affect cancer cell viability and biology and are already in use for the treatment of cancer patients. Immunotherapy can lead to clinical benefit in selected cancer patients, especially in patients with limited disease after tumor debulking. HDAC inhibitors can potentially synergize with immunotherapy by elimination of tumor cells. The direct effects of HDAC inhibitors on immune cell function, however, remain largely unexplored. Initial data have suggested HDAC inhibitors to be predominantly immunosuppressive, but more recent reports have challenged this view. In this review we will discuss the effects of HDAC inhibitors on tumor cells and different immune cell subsets, synergistic interactions and possible mechanisms. Finally, we will address future challenges and potential application of HDAC inhibitors in immunocombination therapy of cancer.
Collapse
Affiliation(s)
- Michiel Kroesen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Pediatric Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Paul Gielen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; These authors contributed equally to this work
| | - Ingrid C Brok
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; These authors contributed equally to this work
| | - Inna Armandari
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter M Hoogerbrugge
- Department of Pediatric Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands; Princes Máxima Center for Pediatric Oncology, The Bilt, The Netherlands
| | - Gosse J Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Wu H, Zhao M, Chang C, Lu Q. The real culprit in systemic lupus erythematosus: abnormal epigenetic regulation. Int J Mol Sci 2015; 16:11013-33. [PMID: 25988383 PMCID: PMC4463688 DOI: 10.3390/ijms160511013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/08/2015] [Accepted: 05/11/2015] [Indexed: 02/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease involving multiple organs and the presence of anti-nuclear antibodies. The pathogenesis of SLE has been intensively studied but remains far from clear. B and T lymphocyte abnormalities, dysregulation of apoptosis, defects in the clearance of apoptotic materials, and various genetic and epigenetic factors are attributed to the development of SLE. The latest research findings point to the association between abnormal epigenetic regulation and SLE, which has attracted considerable interest worldwide. It is the purpose of this review to present and discuss the relationship between aberrant epigenetic regulation and SLE, including DNA methylation, histone modifications and microRNAs in patients with SLE, the possible mechanisms of immune dysfunction caused by epigenetic changes, and to better understand the roles of aberrant epigenetic regulation in the initiation and development of SLE and to provide an insight into the related therapeutic options in SLE.
Collapse
Affiliation(s)
- Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China.
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China.
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616, USA.
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China.
| |
Collapse
|
27
|
Epigenetics in the treatment of systemic lupus erythematosus: potential clinical application. Clin Immunol 2014; 155:79-90. [PMID: 25218424 DOI: 10.1016/j.clim.2014.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 10/24/2022]
Abstract
The current treatments of systemic lupus erythematosus (SLE) have been based on the use of immunosuppressive drugs which are linked to serious side effects. The more effective therapeutic approaches with minimal or no side effects for SLE patients are hard to develop, mainly due to the complexity of the disease. The discovery of pharmacoepigenetics provides a new way to solve this problem. Epigenetic modifications can influence drug efficacy by altering gene expression via changing chromatin structure. Although still in early development, epigenetic studies in SLE are expected to reveal novel therapeutic targets and disease biomarkers in autoimmunity. For example, miRNAs, which have been identified to govern many genes including drug targets, are altered in disease development and after drug administration. This review aims to present an overview of current epigenetic mechanisms involved in the pathogenesis of SLE, and discuss their potential roles in clinical and pharmacological applications.
Collapse
|
28
|
Arbez J, Lamarthée B, Gaugler B, Saas P. Histone deacetylase inhibitor valproic acid affects plasmacytoid dendritic cells phenotype and function. Immunobiology 2014; 219:637-43. [PMID: 24742998 DOI: 10.1016/j.imbio.2014.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 02/19/2014] [Accepted: 03/24/2014] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Plasmacytoid dendritic cells (PDC) represent a rare subset of dendritic cells specialized in the production of type I IFN in response to microbial pathogens. Recent data suggested that histone deacetylase (HDAC) inhibitors possess potent immunomodulatory properties both in vitro and in vivo. In this study, we assayed the ability of the HDAC inhibitor, valproic acid (VPA), to influence the phenotype and functional properties of human PDC isolated from peripheral blood. METHODS AND RESULTS We showed that VPA inhibited the production of IFN-α and the proinflammatory cytokines TNF-α and IL-6 by CpG-activated PDC. VPA also affected the phenotype of PDC by reducing the expression of costimulatory molecules induced by CpG activation. Moreover, VPA reduced the capacity of CpG-stimulated PDC to promote CD4(+) T cell proliferation and IFN-γ production, while enhancing the proportion of IL-10 positive T cells. CONCLUSION These results suggest that HDAC inhibition by VPA alters essential human PDC functions, highlighting the need for monitoring immune functions in cancer patients receiving HDAC inhibitors, but also making these drugs attractive therapies in inflammatory, and autoimmune diseases implicating PDC.
Collapse
Affiliation(s)
- Jessy Arbez
- INSERM UMR1098, Besançon F25020, France; Université de Franche-Comté, Besançon F25000, France; EFS Bourgogne Franche-Comté, F25020 Besançon Cedex, France
| | - Baptiste Lamarthée
- INSERM UMR1098, Besançon F25020, France; Université de Franche-Comté, Besançon F25000, France; EFS Bourgogne Franche-Comté, F25020 Besançon Cedex, France
| | - Béatrice Gaugler
- INSERM UMR1098, Besançon F25020, France; Université de Franche-Comté, Besançon F25000, France; EFS Bourgogne Franche-Comté, F25020 Besançon Cedex, France.
| | - Philippe Saas
- INSERM UMR1098, Besançon F25020, France; Université de Franche-Comté, Besançon F25000, France; EFS Bourgogne Franche-Comté, F25020 Besançon Cedex, France; Centre d'Investigation Clinique en Biothérapie CIC 1431, Plateforme de Biomonitoring, FHU INCREASE, Besançon F25020, France
| |
Collapse
|
29
|
Primary macrophages rely on histone deacetylase 1 and 2 expression to induce type I interferon in response to gammaherpesvirus infection. J Virol 2013; 88:2268-78. [PMID: 24335310 DOI: 10.1128/jvi.03278-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type I interferon is induced shortly following viral infection and represents a first line of host defense against a majority of viral pathogens. Not surprisingly, both replication and latency of gammaherpesviruses, ubiquitous cancer-associated pathogens, are attenuated by type I interferon, although the mechanism of attenuation remains poorly characterized. Gammaherpesviruses also target histone deacetylases (HDACs), a family of pleiotropic enzymes that modify gene expression and several cell signaling pathways. Specifically, we have previously shown that a conserved gammaherpesvirus protein kinase interacts with HDAC1 and -2 to promote gammaherpesvirus replication in primary macrophages. In the current study, we have used genetic approaches to show that expression of HDAC1 and -2 is critical for induction of a type I interferon response following gammaherpesvirus infection of primary macrophages. Specifically, expression of HDAC1 and -2 was required for phosphorylation of interferon regulatory factor 3 (IRF3) and accumulation of IRF3 at the beta interferon promoter in gammaherpesvirus-infected primary macrophages. To our knowledge, this is the first demonstration of a specific role for HDAC1 and -2 in the induction of type I interferon responses in primary immune cells following virus infection. Furthermore, because HDAC1 and -2 are overexpressed in several types of cancer, our findings illuminate potential side effects of HDAC1- and -2-specific inhibitors that are currently under development as cancer therapy agents. IMPORTANCE Gammaherpesviruses establish chronic infection in a majority of the adult population and are associated with several malignancies. Infected cells counteract gammaherpesvirus infection via innate immune signaling mediated primarily through type I interferon. The induction of type I interferon expression proceeds through several stages using molecular mechanisms that are still incompletely characterized. In this study, we show that expression of HDAC1 and -2 by macrophages is required to mount a type I interferon response to incoming gammaherpesvirus. The involvement of HDAC1 and -2 in the type I interferon response highlights the pleiotropic roles of these enzymes in cellular signaling. Interestingly, HDAC1 and -2 are deregulated in cancer and are attractive targets of new cancer therapies. Due to the ubiquitous and chronic nature of gammaherpesvirus infection, the role of HDAC1 and -2 in the induction of type I interferon responses should be considered during the clinical development of HDAC1- and -2-specific inhibitors.
Collapse
|
30
|
Rogatsky I, Chandrasekaran U, Manni M, Yi W, Pernis AB. Epigenetics and the IRFs: A complex interplay in the control of immunity and autoimmunity. Autoimmunity 2013; 47:242-55. [DOI: 10.3109/08916934.2013.853050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Thabet Y, Cañas F, Ghedira I, Youinou P, Mageed RA, Renaudineau Y. Altered patterns of epigenetic changes in systemic lupus erythematosus and auto-antibody production: is there a link? J Autoimmun 2012; 39:154-60. [PMID: 22709855 DOI: 10.1016/j.jaut.2012.05.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 05/20/2012] [Indexed: 02/07/2023]
Abstract
The prominent feature of immunological defects in systemic lupus erythematosus (SLE) is the production of autoantibodies (auto-Abs) to nuclear antigens including DNA, histones and RNP. In addition, there is growing evidence that epigenetic changes play a key role in the pathogenesis of SLE. Autoreactive CD4(+) T cells and B cells in patients with SLE have evidence of altered patterns of DNA methylation as well as post-translational modifications of histones and ribonucleoproteins (RNP). A key question that has emerged from these two characteristic features of SLE is whether the two processes are linked. New data provide support for such a link. For example, there is evidence that hypomethylated DNA is immunogenic, that anti-histone auto-Abs in patients with SLE bind epigenetic-sensitive hot spots and that epigenetically-modified RNP-derived peptides can modulate lupus disease. All in all, the available evidence indicates that a better understanding of dysregulation in epigenetics in SLE may offer opportunities to develop new biomarkers and novel therapeutic strategies.
Collapse
Affiliation(s)
- Yosra Thabet
- EA2216 Immunology, Pathology and Immunotherapy, European University of Brittany, Brest, France
| | | | | | | | | | | |
Collapse
|
32
|
Drews K, Tavernier G, Demeester J, Lehrach H, De Smedt SC, Rejman J, Adjaye J. The cytotoxic and immunogenic hurdles associated with non-viral mRNA-mediated reprogramming of human fibroblasts. Biomaterials 2012; 33:4059-68. [DOI: 10.1016/j.biomaterials.2012.02.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 02/09/2012] [Indexed: 11/28/2022]
|
33
|
|
34
|
Abstract
Recent advances in epigenetics have enhanced our knowledge of how environmental factors (UV radiation, drugs, infections, etc.) contribute to the development of autoimmune diseases (AID) in genetically predisposed individuals. Studies conducted in monozygotic twins discordant for AID and spontaneous autoimmune animal models have highlighted the importance of DNA methylation changes and histone modifications. Alterations in the epigenetic pattern seem to be cell specific, as CD4+ T cells and B cells are dysregulated in systemic lupus erythematosus, synovial fibroblasts in rheumatoid arthritis and cerebral cells in multiple sclerosis. With regard to lymphocytes, the control of tolerance is affected, leading to the development of autoreactive cells. Other epigenetic processes, such as the newly described miRNAs, and post-translational protein modifications may also be suspected. Altogether, a conceptual revolution is in progress, in AID, with potential new therapeutic strategies targeting epigenetic patterns.
Collapse
|
35
|
Shakespear MR, Halili MA, Irvine KM, Fairlie DP, Sweet MJ. Histone deacetylases as regulators of inflammation and immunity. Trends Immunol 2011; 32:335-43. [PMID: 21570914 DOI: 10.1016/j.it.2011.04.001] [Citation(s) in RCA: 404] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/01/2011] [Accepted: 04/03/2011] [Indexed: 12/31/2022]
Abstract
Histone deacetylases (HDACs) remove an acetyl group from lysine residues of target proteins to regulate cellular processes. Small-molecule inhibitors of HDACs cause cellular growth arrest, differentiation and/or apoptosis, and some are used clinically as anticancer drugs. In animal models, HDAC inhibitors are therapeutic for several inflammatory diseases, but exacerbate atherosclerosis and compromise host defence. Loss of HDAC function has also been linked to chronic lung diseases in humans. These contrasting effects might reflect distinct roles for individual HDACs in immune responses. Here, we review the current understanding of innate and adaptive immune pathways that are regulated by classical HDAC enzymes. The objective is to provide a rationale for targeting (or not targeting) individual HDAC enzymes with inhibitors for future immune-related applications.
Collapse
Affiliation(s)
- Melanie R Shakespear
- The University of Queensland, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
36
|
Reilly CM, Regna N, Mishra N. HDAC inhibition in lupus models. Mol Med 2011; 17:417-25. [PMID: 21327298 DOI: 10.2119/molmed.2011.00055] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/10/2011] [Indexed: 12/25/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune inflammatory disease characterized by the production of autoantibodies directed against nuclear antigens such as nucleosomes, DNA and histone proteins found within the body's cells and plasma. Autoantibodies may induce disease by forming immune complexes that lodge in target organs or by crossreacting with targeted antigens and damaging tissue. In addition to autoantibody production, apoptotic defects and impaired removal of apoptotic cells contribute to an overload of autoantigens that initiate an autoimmune response. Besides the well-recognized genetic susceptibility to SLE, environmental and epigenetic factors play a crucial role in disease pathogenesis as evidenced by monozygotic twins typically being discordant for disease. Changes in DNA methylation and histone acetylation alter gene expression and are thought to contribute to the epigenetic deregulation in disease. In SLE, global and gene-specific DNA methylation changes have been demonstrated to occur. Additionally, aberrant histone acetylation is evident in individuals with SLE. Moreover, histone deacetylase inhibitors (HDACi) have been shown to reverse the skewed expression of multiple genes involved in SLE. In this review, we discuss the implications of epigenetic alterations in the development and progression of SLE, and how therapeutics designed to alter histone acetylation status may constitute a promising avenue to target disease.
Collapse
Affiliation(s)
- Christopher M Reilly
- Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA.
| | | | | |
Collapse
|