1
|
Saban Güler M, Arslan S, Ağagündüz D, Cerqua I, Pagano E, Berni Canani R, Capasso R. Butyrate: A potential mediator of obesity and microbiome via different mechanisms of actions. Food Res Int 2025; 199:115420. [PMID: 39658184 DOI: 10.1016/j.foodres.2024.115420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Butyrate, a short-chain fatty acid, is a crucial product of gut microbial fermentation with significant implications for various metabolic and physiological processes. Dietary sources of butyrate are limited, primarily derived from the fermentation of dietary fibers by butyrate-producing gut bacteria. Butyrate exerts its effects primarily as a histone deacetylase (HDAC) inhibitor and through signaling pathways involving G protein-coupled receptors (GPCRs). Its diverse benefits include promoting gut health, enhancing energy metabolism, and potentially alleviating complications associated with obesity. However, the exact role of butyrate in obesity is still under investigation, with a limited number of human trials necessitating further research to determine its efficacy and safety profile. Moreover, butyrate impact on the gut-brain axis and its modulation of microbiome effect on behavior highlight its broader importance in regulating host physiology. A thorough understanding of the metabolic pathways and mechanisms of butyrate is essential for developing targeted interventions for metabolic disorders. Continued research is crucial to fully realize its therapeutic potential and optimize its clinical applications in human health. In summary, this review illuminates the multifaceted role of butyrate as a potential mediator of obesity and related metabolic changes.
Collapse
Affiliation(s)
- Meryem Saban Güler
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey.
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE Biotechnologies Research Center and Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055 Naples, Italy.
| |
Collapse
|
2
|
Lin R, Wang X, Ni C, Fu C, Yang C, Dong D, Wu X, Chen X, Wang L, Hou J. Echinococcus granulosus cyst fluid inhibits KDM6B-mediated demethylation of trimethylated histone H3 lysine 27 and interleukin-1β production in macrophages. Parasit Vectors 2023; 16:422. [PMID: 37974225 PMCID: PMC10652454 DOI: 10.1186/s13071-023-06041-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Echinococcus granulosus can manipulate its host's immune response to ensure its own survival. However, the effect of histone modifications on the regulation of the NOD-like receptor protein 3 (NLRP3) inflammasome and downstream interleukin-1β (IL-1β) production in response to the parasite is not fully understood. METHODS We evaluated IL-1β secretion through enzyme-linked immunosorbent assay and assessed reactive oxygen species levels using the dichlorodihydrofluorescein diacetate probe. Western blotting and quantitative real-time polymerase chain reaction were performed to examine the expression of NLRP3 and IL-1β in mouse peritoneal macrophages and Tohoku Hospital Pediatrics-1 cells, a human macrophage cell line. The presence of trimethylated histone H3 lysine 27 (H3K27me3) modification on NLRP3 and IL-1β promoters was studied by chromatin immunoprecipitation. RESULTS Treatment with E. granulosus cyst fluid (EgCF) considerably reduced IL-1β secretion in mouse and human macrophages, although reactive oxygen species production increased. EgCF also suppressed the expression of NLRP3 and IL-1β. Mechanistically, EgCF prompted the enrichment of repressive H3K27me3 modification on the promoters of both NLRP3 and IL-1β in macrophages. Notably, the presence of EgCF led to a significant reduction in the expression of the H3K27me3 demethylase KDM6B. CONCLUSIONS Our study revealed that EgCF inhibits KDM6B expression and H3K27me3 demethylation, resulting in the transcriptional inhibition of NLRP3 and IL-1β. These results provide new insights into the immune evasion mechanisms of E. granulosus.
Collapse
Affiliation(s)
- Ruolin Lin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiaopeng Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Caiya Ni
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chunxue Fu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chun Yang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | | | - Xiangwei Wu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lianghai Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Jun Hou
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
3
|
Mann MW, Fu Y, Gearhart RL, Xu X, Roberts DS, Li Y, Zhou J, Ge Y, Brasier AR. Bromodomain-containing Protein 4 regulates innate inflammation via modulation of alternative splicing. Front Immunol 2023; 14:1212770. [PMID: 37435059 PMCID: PMC10331468 DOI: 10.3389/fimmu.2023.1212770] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Bromodomain-containing Protein 4 (BRD4) is a transcriptional regulator which coordinates gene expression programs controlling cancer biology, inflammation, and fibrosis. In the context of airway viral infection, BRD4-specific inhibitors (BRD4i) block the release of pro-inflammatory cytokines and prevent downstream epithelial plasticity. Although the chromatin modifying functions of BRD4 in inducible gene expression have been extensively investigated, its roles in post-transcriptional regulation are not well understood. Given BRD4's interaction with the transcriptional elongation complex and spliceosome, we hypothesize that BRD4 is a functional regulator of mRNA processing. Methods To address this question, we combine data-independent analysis - parallel accumulation-serial fragmentation (diaPASEF) with RNA-sequencing to achieve deep and integrated coverage of the proteomic and transcriptomic landscapes of human small airway epithelial cells exposed to viral challenge and treated with BRD4i. Results We discover that BRD4 regulates alternative splicing of key genes, including Interferon-related Developmental Regulator 1 (IFRD1) and X-Box Binding Protein 1 (XBP1), related to the innate immune response and the unfolded protein response (UPR). We identify requirement of BRD4 for expression of serine-arginine splicing factors, splicosome components and the Inositol-Requiring Enzyme 1 IREα affecting immediate early innate response and the UPR. Discussion These findings extend the transcriptional elongation-facilitating actions of BRD4 in control of post-transcriptional RNA processing via modulating splicing factor expression in virus-induced innate signaling.
Collapse
Affiliation(s)
- Morgan W. Mann
- Department of Medicine, University of Wisconsin – Madison, Madison, WI, United States
| | - Yao Fu
- Department of Medicine, University of Wisconsin – Madison, Madison, WI, United States
| | - Robert L. Gearhart
- Department of Chemistry, University of Wisconsin – Madison, Madison, WI, United States
| | - Xiaofang Xu
- Department of Medicine, University of Wisconsin – Madison, Madison, WI, United States
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin – Madison, Madison, WI, United States
| | - Yi Li
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ying Ge
- Department of Chemistry, University of Wisconsin – Madison, Madison, WI, United States
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, United States
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin – Madison, Madison, WI, United States
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
4
|
Mann M, Fu Y, Xu X, Roberts DS, Li Y, Zhou J, Ge Y, Brasier AR. Bromodomain-containing Protein 4 Regulates Innate Inflammation in Airway Epithelial Cells via Modulation of Alternative Splicing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524257. [PMID: 36711789 PMCID: PMC9882210 DOI: 10.1101/2023.01.17.524257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bromodomain-containing Protein 4 (BRD4) is a transcriptional regulator which coordinates gene expression programs controlling cancer biology, inflammation, and fibrosis. In airway viral infection, non-toxic BRD4-specific inhibitors (BRD4i) block the release of pro-inflammatory cytokines and prevent downstream remodeling. Although the chromatin modifying functions of BRD4 in inducible gene expression have been extensively investigated, its roles in post-transcriptional regulation are not as well understood. Based on its interaction with the transcriptional elongation complex and spliceosome, we hypothesize that BRD4 is a functional regulator of mRNA processing. To address this question, we combine data-independent analysis - parallel accumulation-serial fragmentation (diaPASEF) with RNA-sequencing to achieve deep and integrated coverage of the proteomic and transcriptomic landscapes of human small airway epithelial cells exposed to viral challenge and treated with BRD4i. The transcript-level data was further interrogated for alternative splicing analysis, and the resulting data sets were correlated to identify pathways subject to post-transcriptional regulation. We discover that BRD4 regulates alternative splicing of key genes, including Interferon-related Developmental Regulator 1 ( IFRD1 ) and X-Box Binding Protein 1 ( XBP1 ), related to the innate immune response and the unfolded protein response, respectively. These findings extend the transcriptional elongation-facilitating actions of BRD4 in control of post-transcriptional RNA processing in innate signaling.
Collapse
Affiliation(s)
- Morgan Mann
- Department of Medicine, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| | - Yao Fu
- Department of Medicine, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| | - Xiaofang Xu
- Department of Medicine, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| | - Yi Li
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, 77550, Texas, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, 77550, Texas, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA,Human Proteomics Program, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA,Department of Cell and Regenerative Biology, University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| | - Allan R. Brasier
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin – Madison, Madison, 53705, Wisconsin, USA
| |
Collapse
|
5
|
Sen R, Sarkar S, Chlamydas S, Garbati M, Barnes C. Epigenetic features, methods, and implementations associated with COVID-19. OMICS APPROACHES AND TECHNOLOGIES IN COVID-19 2023:161-175. [DOI: 10.1016/b978-0-323-91794-0.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Delshad M, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. Host genetic diversity and genetic variations of SARS-CoV-2 in COVID-19 pathogenesis and the effectiveness of vaccination. Int Immunopharmacol 2022; 111:109128. [PMID: 35963158 PMCID: PMC9359488 DOI: 10.1016/j.intimp.2022.109128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 08/03/2022] [Indexed: 12/14/2022]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for the outbreak of coronavirus disease 2019 (COVID-19), has shown a vast range of clinical manifestations from asymptomatic to life-threatening symptoms. To figure out the cause of this heterogeneity, studies demonstrated the trace of genetic diversities whether in the hosts or the virus itself. With this regard, this review provides a comprehensive overview of how host genetic such as those related to the entry of the virus, the immune-related genes, gender-related genes, disease-related genes, and also host epigenetic could influence the severity of COVID-19. Besides, the mutations in the genome of SARS-CoV-2 __leading to emerging of new variants__ per se affect the affinity of the virus to the host cells and enhance the immune escape capacity. The current review discusses these variants and also the latest data about vaccination effectiveness facing the most important variants.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Liu L, Li Q, Yang Y, Guo A. Biological Function of Short-Chain Fatty Acids and Its Regulation on Intestinal Health of Poultry. Front Vet Sci 2021; 8:736739. [PMID: 34733901 PMCID: PMC8558227 DOI: 10.3389/fvets.2021.736739] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are metabolites generated by bacterial fermentation of dietary fiber (DF) in the hindgut. SCFAs are mainly composed of acetate, propionate and butyrate. Many studies have shown that SCFAs play a significant role in the regulation of intestinal health in poultry. SCFAs are primarily absorbed from the intestine and used by enterocytes as a key substrate for energy production. SCFAs can also inhibit the invasion and colonization of pathogens by lowering the intestinal pH. Additionally, butyrate inhibits the expression of nitric oxide synthase (NOS), which encodes inducible nitric oxide synthase (iNOS) in intestinal cells via the PPAR-γ pathway. This pathway causes significant reduction of iNOS and nitrate, and inhibits the proliferation of Enterobacteriaceae to maintain overall intestinal homeostasis. SCFAs can enhance the immune response by stimulating cytokine production (e.g. TNF-α, IL-2, IL-6, and IL-10) in the immune cells of the host. Similarly, it has been established that SCFAs promote the differentiation of T cells into T regulatory cells (Tregs) and expansion by binding to receptors, such as Toll-like receptors (TLR) and G protein-coupled receptors (GPRs), on immune cells. SCFAs have been shown to repair intestinal mucosa and alleviate intestinal inflammation by activating GPRs, inhibiting histone deacetylases (HDACs), and downregulating the expression of pro-inflammatory factor genes. Butyrate improves tight-junction-dependent intestinal barrier function by promoting tight junction (TJ) assembly. In recent years, the demand for banning antibiotics has increased in poultry production. Therefore, it is extremely important to maintain the intestinal health and sustainable production of poultry. Taking nutrition strategies is important to regulate SCFA production by supplementing dietary fiber and prebiotics, SCFA-producing bacteria (SPB), and additives in poultry diet. However, excessive SCFAs will lead to the enteritis in poultry production. There may be an optimal level and proportion of SCFAs in poultry intestine, which benefits to gut health of poultry. This review summarizes the biological functions of SCFAs and their role in gut health, as well as nutritional strategies to regulate SCFA production in the poultry gut.
Collapse
Affiliation(s)
- Lixuan Liu
- Faculty of Life Sciences, Southwest Forestry University, Kunming, China
| | - Qingqing Li
- Faculty of Life Sciences, Southwest Forestry University, Kunming, China
- Kunming Xianghao Technology Co., Ltd., Kunming, China
| | - Yajin Yang
- Faculty of Life Sciences, Southwest Forestry University, Kunming, China
| | - Aiwei Guo
- Faculty of Life Sciences, Southwest Forestry University, Kunming, China
| |
Collapse
|
8
|
Choudhary S, Sreenivasulu K, Mitra P, Misra S, Sharma P. Role of Genetic Variants and Gene Expression in the Susceptibility and Severity of COVID-19. Ann Lab Med 2021; 41:129-138. [PMID: 33063674 PMCID: PMC7591285 DOI: 10.3343/alm.2021.41.2.129] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/20/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023] Open
Abstract
Since its first report in December 2019, coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly emerged as a pandemic affecting nearly all countries worldwide. As the COVID-19 pandemic progresses, the need to identify genetic risk factors for susceptibility to this serious illness has emerged. Host genetic factors, along with other risk factors may help determine susceptibility to respiratory tract infections. It is hypothesized that the ACE2 gene, encoding angiotensin-converting enzyme 2 (ACE2), is a genetic risk factor for SARS-CoV-2 infection and is required by the virus to enter cells. Together with ACE2, transmembrane protease serine 2 (TMPRSS2) and dipeptidyl peptidase-4 (DPP4) also play an important role in disease severity. Evaluating the role of genetic variants in determining the direction of respiratory infections will help identify potential drug target candidates for further study in COVID-19 patients. We have summarized the latest reports demonstrating that ACE2 variants, their expression, and epigenetic factors may influence an individual's susceptibility to SARS-CoV-2 infection and disease outcome.
Collapse
Affiliation(s)
- Sarita Choudhary
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Karli Sreenivasulu
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Prasenjit Mitra
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Sanjeev Misra
- Department of Surgical Oncology, All India Institute of Medical Sciences, Jodhpur, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| |
Collapse
|
9
|
Abstract
The COVID-19 pandemic is one of the most significant public health threats in recent history and has impacted the lives of almost everyone worldwide. Epigenetic mechanisms contribute to many aspects of the SARS-CoV-2 replication cycle, including expression levels of viral receptor ACE2, expression of cytokine genes as part of the host immune response, and the implication of various histone modifications in several aspects of COVID-19. SARS-CoV-2 proteins physically associate with many different host proteins over the course of infection, and notably there are several interactions between viral proteins and epigenetic enzymes such as HDACs and bromodomain-containing proteins as shown by correlation-based studies. The many contributions of epigenetic mechanisms to the viral life cycle and the host immune response to infection have resulted in epigenetic factors being identified as emerging biomarkers for COVID-19, and project epigenetic modifiers as promising therapeutic targets to combat COVID-19. This review article highlights the major epigenetic pathways at play during COVID-19 disease and discusses ongoing clinical trials that will hopefully contribute to slowing the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Rwik Sen
- Active Motif, Incorporated, 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
| | - Michael Garbati
- Active Motif, Incorporated, 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
| | - Kevin Bryant
- Active Motif, Incorporated, 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
| | - Yanan Lu
- Active Motif, Incorporated, 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
| |
Collapse
|
10
|
Couto MR, Gonçalves P, Magro F, Martel F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol Res 2020; 159:104947. [DOI: 10.1016/j.phrs.2020.104947] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/04/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
|
11
|
Stazi G, Taglieri L, Nicolai A, Romanelli A, Fioravanti R, Morrone S, Sabatino M, Ragno R, Taurone S, Nebbioso M, Carletti R, Artico M, Valente S, Scarpa S, Mai A. Dissecting the role of novel EZH2 inhibitors in primary glioblastoma cell cultures: effects on proliferation, epithelial-mesenchymal transition, migration, and on the pro-inflammatory phenotype. Clin Epigenetics 2019; 11:173. [PMID: 31791385 PMCID: PMC6889222 DOI: 10.1186/s13148-019-0763-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most lethal and aggressive malignant primary brain tumor in adults. After surgical resection of the tumor, the patient typically should be subjected to chemotherapy (temozolomide, TMZ) and concomitant radiotherapy. Since the TMZ treatment does not lead to complete remission and often develops resistance, the identification of efficacious therapeutics is strongly to pursue. Among the epigenetic players, the H3K27 methyltransferase (MT) EZH2 (enhancer of zeste homologue 2) has been found overexpressed or mutated in several human cancers including gliomas, and its overexpression is associated with poor outcome in GBM. Two EZH2 inhibitors (EZH2i), UNC1999 and GSK343, suppressed GBM growth in vitro and in vivo indicating that EZH2i can be potential drugs against GBM. RESULTS Two new EZH2i, MC4040 and MC4041, were designed, prepared, and tested by us to determine their effects in primary GBM cell cultures. MC4040 and MC4041 displayed single-digit micromolar inhibition of EZH2, 10-fold less potency against EZH1, and no activity towards other MTs. In primary GBM cells as well as in U-87 GBM cells, the two compounds reduced H3K27me3 levels, and dose- and time-dependently impaired GBM cell viability without inducing apoptosis and arresting the cell cycle in the G0/G1 phase, with increased p21 and p27 levels. In combination with TMZ, MC4040 and MC4041 displayed stronger, but not additive, effects on cell viability. The potent clinical candidate as EZH2i tazemetostat, alone or in combination with TMZ, exhibited a similar potency of inhibition of GBM cell growth when compared to MC4040 and MC4041. At the molecular level, MC4040 and MC4041 reduced the VEGFR1/VEGF expression, reversed the epithelial-mesenchymal transition (EMT), and hampered cell migration and invasion attenuating the cancer malignant phenotype. Treatment of GBM cells with MC4040 and MC4041 also impaired the GBM pro-inflammatory phenotype, with a significant decrease of TGF-β, TNF-α, and IL-6, joined to an increase of the anti-inflammatory cytokine IL-10. CONCLUSIONS The two novel EZH2i MC4040 and MC4041 impaired primary GBM cell viability, showing even stronger effects in combination with TMZ. They also weakened the aggressive malignant phenotype by reducing angiogenesis, EMT, cell migration/invasion and inflammation, thus they may be considered potential candidates against GBM also for combination therapies.
Collapse
Affiliation(s)
- Giulia Stazi
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Ludovica Taglieri
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alice Nicolai
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Annalisa Romanelli
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Rossella Fioravanti
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Stefania Morrone
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | | | - Rino Ragno
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Alchemical Dynamics s.r.l, 00125 Roma, Italy
| | - Samanta Taurone
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Marcella Nebbioso
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Raffaella Carletti
- Department of Radiologic, Oncological, and Anatomical and Pathological Sciences, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Marco Artico
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Susanna Scarpa
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
12
|
Chen J, Zhang M, Zhang X, Fan L, Liu P, Yu L, Cao X, Qiu S, Xu Y. EZH2 inhibitor DZNep modulates microglial activation and protects against ischaemic brain injury after experimental stroke. Eur J Pharmacol 2019; 857:172452. [PMID: 31202798 DOI: 10.1016/j.ejphar.2019.172452] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 01/04/2023]
Abstract
Enhancer of zeste homolog-2 (EZH2), a histone methyltransferase, has been recognized to play a pivotal role in regulating the immune response in various diseases. However, its role in the inflammatory response induced by ischaemic stroke remains to be further investigated. The aim of this study was to determine the role of EZH2 in microglia-associated inflammation in ischaemic stroke and to further detect the effects of the EZH2 inhibitor, 3-deazaadenosine A (DZNep), in ischaemic brain injury. Here, we found that both in vivo ischemic/reperfusion (I/R) injury and in vitro oxygen-glucose deprivation (OGD) treatment induced a marked upregulation of EZH2 in microglia. The administration of the EZH2 inhibitor DZNep improved behavioural performance and reduced the infarct volume in mice after experimental stroke. Furthermore, we showed that DZNep blocked pro-inflammatory (CD86+) microglial activation and triggered anti-inflammatory (CD206+) microglial polarization in experimental stroke. Pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and CXCL10 were also significantly downregulated by DZNep. In addition, it was found that DZNep blocked the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in microglia, which was increased by I/R injury and OGD. Collectively, we demonstrated that EZH2 is implicated in regulating microglial activation and exacerbates neurological deficits after ischaemic stroke, probably via activating STAT3, and that the EZH2 inhibitor DZNep can exert neuroprotective effects after ischaemic stroke.
Collapse
Affiliation(s)
- Jian Chen
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Meijuan Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Xi Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Lizhen Fan
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Pinyi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Linjie Yu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Shuwei Qiu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
13
|
Kim MJ, Yun JM. Molecular Mechanism of the Protective Effect of Zerumbone on Lipopolysaccharide-Induced Inflammation of THP-1 Cell-Derived Macrophages. J Med Food 2019; 22:62-73. [DOI: 10.1089/jmf.2018.4253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Min-Ju Kim
- Department of Food and Nutrition, Chonnam National University, Gwangju, South Korea
| | - Jung-Mi Yun
- Department of Food and Nutrition, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
14
|
Borghini L, Hibberd M, Davila S. Changes in H3K27ac following lipopolysaccharide stimulation of nasopharyngeal epithelial cells. BMC Genomics 2018; 19:969. [PMID: 30587130 PMCID: PMC6307289 DOI: 10.1186/s12864-018-5295-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The epithelium is the first line of defense against pathogens. Notably the epithelial cells lining the respiratory track are crucial in sensing airborne microbes and mounting an effective immune response via the expression of target genes such as cytokines and chemokines. Gene expression regulation following microbial recognition is partly regulated by chromatin re-organization and has been described in immune cells but data from epithelial cells is not as detailed. Here, we report genome-wide changes of the H3K27ac mark, characteristic of activated enhancers and promoters, after stimulation of nasopharyngeal epithelial cells with the bacterial endotoxin Lipopolysaccharide (LPS). RESULTS In this study, we have identified 626 regions where the H3K27ac mark showed reproducible increase following LPS induction in epithelial cells. This indicated that sensing of LPS led to opening of the chromatin in our system. Moreover, this phenomenon seemed to happen extensively at enhancers regions and we could observe instances of Super-enhancer formation. As expected, LPS-increased H3K27ac regions were found in the vicinity of genes relevant for LPS response and these changes correlated with up-regulation of their expression. In addition, we found the induction of H3K27ac mark to overlap with the binding of one of the NF-kB members and key regulator of the innate immune response, RELA, following LPS sensing. Indeed, inhibiting the NF-kB pathway abolished the deposition of H3K27ac at the TNF locus, a target of RELA, suggesting that these two phenomena are associated. CONCLUSIONS Enhancers' selection and activation following microbial or inflammatory stimuli has been described previously and shown to be mediated via the NF-kB pathway. Here, we demonstrate that this is also likely to occur in the case of LPS-sensing by nasopharyngeal epithelial cells as well. In addition to validating previous findings, we generated a valuable data set relevant to the host immune response to epithelial cell colonizing or infecting pathogens.
Collapse
Affiliation(s)
- Lisa Borghini
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Infectious Disease, Genome Institute of Singapore, Singapore, 138672, Singapore.
| | - Martin Hibberd
- Infectious Disease, Genome Institute of Singapore, Singapore, 138672, Singapore.,Present Address: Pathogen Molecular Biology, Infectious & Tropical Disease, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore.,Present Address: SingHealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore, 169609, Singapore
| |
Collapse
|
15
|
Downregulated expression of miR-223 promotes Toll-like receptor-activated inflammatory responses in macrophages by targeting RhoB. Mol Immunol 2017; 91:42-48. [PMID: 28881218 DOI: 10.1016/j.molimm.2017.08.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/10/2017] [Accepted: 08/28/2017] [Indexed: 11/21/2022]
Abstract
Toll-like receptors (TLRs) induced-inflammatory response must be tightly regulated to avoid impairment in host itself. Numerous factors have been identified in regulation of TLR-triggered inflammatory response. Among these, microRNAs (miRNAs) are small non-coding RNA molecules which have got much attention. MiR-223, which highly expresses in myeloid cells of the bone marrow, has reported to participate in kinds of inflammatory responses by targeting inflammasome sensor-NLRP3 to repress production of IL-6 and IL-1β, and thus attenuate inflammatory response. However, the function of miR-223 in TLRs-activated inflammatory response of macrophages is not clear. Here we found miR-223 expression is dramatically reduced in macrophages by TLR ligand stimulation (e.g. LPS, CpG and poly (I:C)). The down-regulated miR-223 leads to the increase in the RhoB expression, which induce the activation of NF-κB and MAPK signaling, promoting TNF-α, IL-6 and IL-1β production upon LPS stimulation. In addition, the histone deacetylase inhibitor trichostatin A increased miR-223 expression obviously in TLR-triggered macrophages, which in turn suppressed RhoB expression and downstream IL-6 production, suggesting that the inhibition of miR-223 by histone deacetylation may be involved in the regulation of TLR-activated inflammatory response. Herein, our findings suggest that miR-223-RhoB axis might be a novel target for the treatment of inflammatory diseases.
Collapse
|
16
|
Arifuzzaman S, Das A, Kim SH, Yoon T, Lee YS, Jung KH, Chai YG. Selective inhibition of EZH2 by a small molecule inhibitor regulates microglial gene expression essential for inflammation. Biochem Pharmacol 2017; 137:61-80. [PMID: 28431938 DOI: 10.1016/j.bcp.2017.04.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022]
Abstract
Multiple studies have documented that Enhancer of zeste homolog 2 (EZH2) could play a role in inflammation and a wide range of malignancies; however, the underlying mechanisms remain largely unaddressed. Microglial activation is a key process in the production and release of numerous pro-inflammatory mediators that play important roles in inflammation and neurodegeneration in the central nervous system (CNS). Therefore, our aim was to investigate whether inhibition of EZH2 with the selective small molecule inhibitor EPZ-6438 protects against neonatal microglial activation. First, in mouse primary microglial cells and a microglial cell line, we found that LPS can rapidly increase EZH2 mRNA level and we subsequently performed gene expression profiling and constructed networks in resting, EPZ-6438-treated, LPS-treated and LPS+EPZ-6438-treated primary microglial cells and a microglial cell line using transcriptome RNA sequencing and bioinformatics analyses. By examining the RNA sequencing, we identified EPZ-6438 target genes and co-regulated modules that were critical for inflammation. We also identified unexpected relationships between the inducible transcription factors (TFs), motif strength, and the transcription of key inflammatory mediators. Furthermore, we showed that EPZ-6438 controls important inflammatory gene targets by modulating interferon regulatory factor (IRF) 1, IRF8, and signal transducer and activator of transcription (STAT) 1 levels at their promoter sites. Our unprecedented findings demonstrate that pharmacological interventions built upon EZH2 inhibition by EPZ-6438 could be a useful therapeutic approach for the treatment of neuroinflammatory diseases associated with microglial activation.
Collapse
Affiliation(s)
- Sarder Arifuzzaman
- Department of Bionanotechnology, Hanyang University, Seoul 04673, Republic of Korea.
| | - Amitabh Das
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, Republic of Korea.
| | - Sun Hwa Kim
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Republic of Korea.
| | - Taeho Yoon
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Republic of Korea.
| | - Young Seek Lee
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Republic of Korea.
| | - Kyoung Hwa Jung
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, Republic of Korea.
| | - Young Gyu Chai
- Department of Bionanotechnology, Hanyang University, Seoul 04673, Republic of Korea; Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Republic of Korea.
| |
Collapse
|
17
|
Abstract
Coronaviruses (CoV) comprise a large group of emerging human and animal pathogens, including the highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) strains. The molecular mechanisms regulating emerging coronavirus pathogenesis are complex and include virus–host interactions associated with entry, replication, egress and innate immune control. Epigenetics research investigates the genetic and non-genetic factors that regulate phenotypic variation, usually caused by external and environmental factors that alter host expression patterns and performance without any change in the underlying genotype. Epigenetic modifications, such as histone modifications, DNA methylation, chromatin remodeling, and non-coding RNAs, function as important regulators that remodel host chromatin, altering host expression patterns and networks in a highly flexible manner. For most of the past two and a half decades, research has focused on the molecular mechanisms by which RNA viruses antagonize the signaling and sensing components that regulate induction of the host innate immune and antiviral defense programs upon infection. More recently, a growing body of evidence supports the hypothesis that viruses, even lytic RNA viruses that replicate in the cytoplasm, have developed intricate, highly evolved, and well-coordinated processes that are designed to regulate the host epigenome, and control host innate immune antiviral defense processes, thereby promoting robust virus replication and pathogenesis. In this article, we discuss the strategies that are used to evaluate the mechanisms by which viruses regulate the host epigenome, especially focusing on highly pathogenic respiratory RNA virus infections as a model. By combining measures of epigenome reorganization with RNA and proteomic datasets, we articulate a spatial-temporal data integration approach to identify regulatory genomic clusters and regions that play a crucial role in the host’s innate immune response, thereby defining a new viral antagonism mechanism following emerging coronavirus infection.
Collapse
|
18
|
Yıldırım-Buharalıoğlu G, Bond M, Sala-Newby GB, Hindmarch CCT, Newby AC. Regulation of Epigenetic Modifiers, Including KDM6B, by Interferon-γ and Interleukin-4 in Human Macrophages. Front Immunol 2017; 8:92. [PMID: 28228757 PMCID: PMC5296311 DOI: 10.3389/fimmu.2017.00092] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/19/2017] [Indexed: 11/25/2022] Open
Abstract
Background Interferon-γ (IFN-γ) or interleukin-4 (IL-4) drives widely different transcriptional programs in macrophages. However, how IFN-γ and IL-4 alter expression of histone-modifying enzymes involved in epigenetic regulation and how this affects the resulting phenotypic polarization is incompletely understood. Methods and results We investigated steady-state messenger RNA levels of 84 histone-modifying enzymes and related regulators in colony-stimulating factor-1 differentiated primary human macrophages using quantitative polymerase chain reaction. IFN-γ or IL-4 treatment for 6–48 h changed 11 mRNAs significantly. IFN-γ increased CIITA, KDM6B, and NCOA1, and IL-4 also increased KDM6B by 6 h. However, either cytokine decreased AURKB, ESCO2, SETD6, SUV39H1, and WHSC1, whereas IFN-γ alone decreased KAT2A, PRMT7, and SMYD3 mRNAs only after 18 h, which coincided with decreased cell proliferation. Rendering macrophages quiescent by growth factor starvation or adenovirus-mediated overexpression of p27kip1 inhibited expression of AURKB, ESCO2, SUV39H1, and WHSC1, and mRNA levels were restored by overexpressing the S-phase transcription factor E2F1, implying their expression, at least partly, depended on proliferation. However, CIITA, KDM6B, NCOA1, KAT2A, PRMT7, SETD6, and SMYD3 were regulated independently of effects on proliferation. Silencing KDM6B, the only transcriptional activator upregulated by both IFN-γ and IL-4, pharmacologically or with short hairpin RNA, blunted a subset of responses to each cytokine. Conclusion These findings demonstrate that IFN-γ or IL-4 can regulate the expression of histone acetyl transferases and histone methyl transferases independently of effects on proliferation and that upregulation of the histone demethylase, KDM6B, assists phenotypic polarization by both cytokines.
Collapse
Affiliation(s)
| | - Mark Bond
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| | - Graciela B Sala-Newby
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| | - Charles C T Hindmarch
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Andrew C Newby
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| |
Collapse
|
19
|
De Luca M, Pels K, Moleirinho S, Curtale G. The epigenetic landscape of innate immunity. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.1.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
20
|
Emerging Roles for Epigenetic Programming in the Control of Inflammatory Signaling Integration in Heath and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1024:63-90. [PMID: 28921465 DOI: 10.1007/978-981-10-5987-2_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Macrophages and dendritic cells initiate the innate immune response to infection and injury and contribute to inflammatory signaling to maintain the homeostasis of various tissues, which includes resident macrophages for the elimination of invading microorganisms and tissue damage. Inappropriate inflammatory signaling can lead to persistent inflammation and further develop into autoimmune and inflammation-associated diseases. Inflammatory signaling pathways have been well characterized, but how these signaling pathways are converted into sustained and diverse patterns of expression of cytokines, chemokines, and other genes in response to environmental challenges is unclear. Emerging evidence suggests the important role of epigenetic mechanisms in finely tuning the outcome of the host innate immune response. An understanding of epigenetic regulation of innate immune cell identity and function will enable the identification of the mechanism between gene-specific host defenses and inflammatory disease and will also allow for exploration of the program of innate immune memory in health and disease. This information could be used to develop therapeutic agents to enhance the host response, preventing chronic inflammation through preserving tissues and signaling integrity.
Collapse
|
21
|
Kuban KCK, Joseph RM, O’Shea TM, Heeren T, Fichorova RN, Douglass L, Jara H, Frazier JA, Hirtz D, Rollins JV, Paneth N. Circulating Inflammatory-Associated Proteins in the First Month of Life and Cognitive Impairment at Age 10 Years in Children Born Extremely Preterm. J Pediatr 2017; 180:116-123.e1. [PMID: 27788929 PMCID: PMC5183478 DOI: 10.1016/j.jpeds.2016.09.054] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/19/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To evaluate whether in children born extremely preterm, indicators of sustained systemic inflammation in the first month of life are associated with cognitive impairment at school age. STUDY DESIGN A total of 873 of 966 eligible children previously enrolled in the multicenter Extremely Low Gestational Age Newborn Study from 2002 to 2004 were evaluated at age 10 years. We analyzed the relationship between elevated blood concentrations of inflammation-associated proteins in the first 2 weeks ("early elevations"; n = 812) and the third and fourth week ("late elevations"; n = 532) of life with neurocognition. RESULTS Early elevations of C-reactive protein, tumor necrosis factor-α, interleukin (IL)-8, intercellular adhesion molecule (ICAM)-1, and erythropoietin were associated with IQ values >2 SD below the expected mean (ORs: 2.0-2.3) and with moderate to severe cognitive impairment on a composite measure of IQ and executive function (ORs: 2.1-3.6). Additionally, severe cognitive impairment was associated with late protein elevations of C-reactive protein (OR: 4.0; 95% CI 1.5, 10), IL-8 (OR: 5.0; 1.9, 13), ICAM-1 (OR: 6.5; 2.6, 16), vascular endothelial growth factor-receptor 2 (OR: 3.2; 1.2, 8.3), and thyroid-stimulating hormone (OR: 3.1; 1.3, 7.3). Moderate cognitive impairment was most strongly associated with elevations of IL-8, ICAM-1, and vascular endothelial growth factor-receptor 2. When 4 or more inflammatory proteins were elevated early, the risk of having an IQ <70 and having overall impaired cognitive ability was more than doubled (ORs: 2.1-2.4); the presence of 4 or more inflammatory protein elevated late was strongly linked to adverse cognitive outcomes (ORs: 2.9-4.8). CONCLUSIONS Extremely preterm children who had sustained elevations of inflammation-related proteins in the first postnatal month are more likely than extremely preterm peers without such elevations to have cognitive impairment at 10 years.
Collapse
Affiliation(s)
- Karl C. K. Kuban
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Robert M. Joseph
- Department of Anatomy and Neuroanatomy, Boston University School of Medicine, Boston, MA, USA
| | - Thomas M. O’Shea
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy Heeren
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Laurie Douglass
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Hernan Jara
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
| | - Jean A. Frazier
- Department of Psychiatry, UMASS Medical School/ University of Massachusetts Memorial Health Care, Worcester, MA, USA
| | - Deborah Hirtz
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | | | - Nigel Paneth
- Department of Epidemiology and Biostatistics and Pediatrics, Michigan State University
| | | |
Collapse
|
22
|
Innate immune cell activation and epigenetic remodeling in symptomatic and asymptomatic atherosclerosis in humans in vivo. Atherosclerosis 2016; 254:228-236. [PMID: 27764724 DOI: 10.1016/j.atherosclerosis.2016.10.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS We have recently reported that monocytes can undergo functional and transcriptional reprogramming towards a long-term pro-inflammatory phenotype after brief in vitro exposure to atherogenic stimuli such as oxidized LDL. This process is termed 'trained immunity', and is mediated by epigenetic remodeling and a metabolic switch towards increased aerobic glycolysis. We hypothesize that trained immunity contributes to atherogenesis. Therefore, we investigated the inflammatory phenotype and epigenetic remodeling of monocytes from patients with and without established atherosclerosis. METHODS Monocytes were isolated from 20 patients with severe symptomatic coronary atherosclerosis (total plaque score >4 on coronary computed tomography angiography) and 17 patients with asymptomatic carotid atherosclerosis and matched controls for both groups. Ex vivo stimulation, RNA analysis and chromatin immunoprecipitation were performed. RESULTS Monocytes from patients with symptomatic atherosclerosis have a higher production of pro-inflammatory cytokines upon LPS stimulation than healthy controls (TNFα 499 ± 102 vs. 267 ± 45 pg/ml, p = 0.01). This was associated with lower histone 3 lysine 4 trimethylation (H3K4me3) (19% vs. 33%, p = 0.002), and lower H3K27me3 (0.005% vs. 0.8%, p < 0.0001) on the TNFα promoter. Furthermore, relative mRNA expression of the glycolytic rate limiting enzymes hexokinase 2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 was higher in patients (0.7 ± 0.2 vs. 0.3 ± 0.1 resp. 1.7 ± 0.2 vs. 1.0 ± 0.1, p = 0.007 resp. 0.003) compared to control individuals. Interestingly, this pro-inflammatory phenotype was only present in patients with symptomatic atherosclerosis, and not in patients with asymptomatic carotid atherosclerosis. CONCLUSIONS Circulating monocytes of patients with symptomatic, but not asymptomatic, atherosclerosis have a pro-inflammatory phenotype and increased expression of glycolytic enzymes, associated with epigenetic remodeling at the level of histone methylation.
Collapse
|
23
|
Histone Deacetylase Inhibition Protects Mice Against Lethal Postinfluenza Pneumococcal Infection. Crit Care Med 2016; 44:e980-7. [DOI: 10.1097/ccm.0000000000001821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
24
|
The transcriptional repressor Hes1 attenuates inflammation by regulating transcription elongation. Nat Immunol 2016; 17:930-7. [PMID: 27322654 PMCID: PMC4955730 DOI: 10.1038/ni.3486] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 05/05/2016] [Indexed: 12/15/2022]
Abstract
Most of the known regulatory mechanisms that curb inflammatory gene expression target pre-transcription-initiation steps, and evidence for post-initiation regulation of inflammatory gene expression remains scarce. We found that the transcriptional repressor Hes1 suppressed production of CXCL1, a chemokine that is crucial for recruiting neutrophils. Hes1 negatively regulated neutrophil recruitment in vivo in a manner that was dependent on macrophage-produced CXCL1, and it attenuated the severity of inflammatory arthritis. Mechanistically, inhibition of Cxcl1 expression by Hes1 did not involve modification of transcription initiation. Instead, Hes1 inhibited signal-induced recruitment of the positive transcription-elongation complex P-TEFb and thereby prevented phosphorylation of RNA polymerase II at Ser2 and productive elongation. Thus, our results identify Hes1 as a homeostatic suppressor of inflammatory responses that exerts its suppressive function by regulating transcription elongation.
Collapse
|
25
|
Kapellos TS, Iqbal AJ. Epigenetic Control of Macrophage Polarisation and Soluble Mediator Gene Expression during Inflammation. Mediators Inflamm 2016; 2016:6591703. [PMID: 27143818 PMCID: PMC4842078 DOI: 10.1155/2016/6591703] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/28/2016] [Indexed: 12/14/2022] Open
Abstract
Macrophages function as sentinel cells, which constantly monitor the host environment for infection or injury. Macrophages have been shown to exhibit a spectrum of activated phenotypes, which can often be categorised under the M1/M2 paradigm. M1 macrophages secrete proinflammatory cytokines and chemokines, such as TNF-α, IL-6, IL-12, CCL4, and CXCL10, and induce phagocytosis and oxidative dependent killing mechanisms. In contrast, M2 macrophages support wound healing and resolution of inflammation. In the past decade, interest has grown in understanding the mechanisms involved in regulating macrophage activation. In particular, epigenetic control of M1 or M2 activation states has been shown to rely on posttranslational modifications of histone proteins adjacent to inflammatory-related genes. Changes in methylation and acetylation of histones by methyltransferases, demethylases, acetyltransferases, and deacetylases can all impact how macrophage phenotypes are generated. In this review, we summarise the latest advances in the field of epigenetic regulation of macrophage polarisation to M1 or M2 states, with particular focus on the cytokine and chemokine profiles associated with these phenotypes.
Collapse
Affiliation(s)
| | - Asif J. Iqbal
- Sir William Dunn school of Pathology, South Parks Road, Oxford OX1 3RE, UK
| |
Collapse
|
26
|
El Aidy S, Stilling R, Dinan TG, Cryan JF. Microbiome to Brain: Unravelling the Multidirectional Axes of Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:301-36. [PMID: 26589226 DOI: 10.1007/978-3-319-20215-0_15] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gut microbiome plays a crucial role in host physiology. Disruption of its community structure and function can have wide-ranging effects making it critical to understand exactly how the interactive dialogue between the host and its microbiota is regulated to maintain homeostasis. An array of multidirectional signalling molecules is clearly involved in the host-microbiome communication. This interactive signalling not only impacts the gastrointestinal tract, where the majority of microbiota resides, but also extends to affect other host systems including the brain and liver as well as the microbiome itself. Understanding the mechanistic principles of this inter-kingdom signalling is fundamental to unravelling how our supraorganism function to maintain wellbeing, subsequently opening up new avenues for microbiome manipulation to favour desirable mental health outcome.
Collapse
Affiliation(s)
- Sahar El Aidy
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Roman Stilling
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.,Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland. .,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
27
|
Tarantino G, Finelli C. Lipids, Low-Grade Chronic Inflammation and NAFLD. HANDBOOK OF LIPIDS IN HUMAN FUNCTION 2016:731-759. [DOI: 10.1016/b978-1-63067-036-8.00028-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Tarantino G, Finelli C. Lipids Nutrition and Epigenetic Modification in Obesity-Related Co-Morbitities * *All authors equally contributed to draft the manuscript. All authors gave final approval of the version to be published. Disclosure statement: The authors declare that there are no conflicts of interest. HANDBOOK OF LIPIDS IN HUMAN FUNCTION 2016:85-110. [DOI: 10.1016/b978-1-63067-036-8.00004-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Tripathi S, Garcia-Sastre A. Antiviral innate immunity through the lens of systems biology. Virus Res 2015; 218:10-7. [PMID: 26657882 DOI: 10.1016/j.virusres.2015.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022]
Abstract
Cellular innate immunity poses the first hurdle against invading viruses in their attempt to establish infection. This antiviral response is manifested with the detection of viral components by the host cell, followed by transduction of antiviral signals, transcription and translation of antiviral effectors and leads to the establishment of an antiviral state. These events occur in a rather branched and interconnected sequence than a linear path. Traditionally, these processes were studied in the context of a single virus and a host component. However, with the advent of rapid and affordable OMICS technologies it has become feasible to address such questions on a global scale. In the discipline of Systems Biology', extensive omics datasets are assimilated using computational tools and mathematical models to acquire deeper understanding of complex biological processes. In this review we have catalogued and discussed the application of Systems Biology approaches in dissecting the antiviral innate immune responses.
Collapse
Affiliation(s)
- Shashank Tripathi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Adolfo Garcia-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, NY, USA.
| |
Collapse
|
30
|
Picascia A, Grimaldi V, Iannone C, Soricelli A, Napoli C. Innate and adaptive immune response in stroke: Focus on epigenetic regulation. J Neuroimmunol 2015; 289:111-20. [PMID: 26616880 DOI: 10.1016/j.jneuroim.2015.10.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/17/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Inflammation and immune response play a pivotal role in the pathophysiology of ischemic stroke giving their contribution to tissue damage and repair. Emerging evidence supports the involvement of epigenetic mechanisms such as methylation, histone modification and miRNAs in the pathogenesis of stroke. Interestingly, epigenetics can influence the molecular events involved in ischemic injury by controlling the switch from pro- to anti-inflammatory response, however, this is still a field to be fully explored. The knowledge of epigenetic processes could to allow for the discovery of more sensitive and specific biomarkers for risk, onset, and progression of disease as well as further novel tools to be used in both primary prevention and therapy of stroke. Indeed, studies performed in vitro and in small animal models seem to suggest a neuroprotective role of HDAC inhibitors (e.g. valproic acid) and antagomir (e.g. anti-miR-181a) in ischemic condition by modulation of both immune and inflammatory pathways. Thus, the clinical implications of altered epigenetic mechanisms for the prevention of stroke are very promising but clinical prospective studies and translational approaches are still warranted.
Collapse
Affiliation(s)
- Antonietta Picascia
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy
| | - Vincenzo Grimaldi
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy.
| | - Carmela Iannone
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy
| | - Andrea Soricelli
- IRCCS Research Institute SDN, Naples, Italy; Department of Studies of Institutions and Territorial Systems, University of Naples Parthenope, Naples, Italy
| | - Claudio Napoli
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy; IRCCS Research Institute SDN, Naples, Italy
| |
Collapse
|
31
|
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: 1.1] [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
|
32
|
Epigenetic pathways in macrophages emerge as novel targets in atherosclerosis. Eur J Pharmacol 2015; 763:79-89. [PMID: 26004034 DOI: 10.1016/j.ejphar.2015.03.101] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/25/2015] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disorder. Monocytes and macrophages are key immune cells in the development of disease and clinical outcome. It is becoming increasingly clear that epigenetic pathways govern many aspects of monocyte and macrophage differentiation and activation. The dynamic regulation of epigenetic patterns provides opportunities to alter disease-associated epigenetic states. Therefore, pharmaceutical companies have embraced the targeting of epigenetic processes as new approaches for interventions. Particularly histone deacetylase (Hdac) inhibitors and DNA-methyltransferase inhibitors have long received attention and several of them have been approved for clinical use in relation to hematological malignancies. The key focus is still on oncology, but Alzheimer's disease, Huntington's disease and inflammatory disorders are coming in focus as well. These developments raise opportunities for the epigenetic targeting in cardiovascular disease (CVD). In this review we discuss the epigenetic regulation of the inflammatory pathways in relation to atherosclerosis with a specific attention to monocyte- and macrophage-related processes. What are the opportunities for future therapy of atherosclerosis by epigenetic interventions?
Collapse
|
33
|
Zhang X, Xu J, Ke X, Zhang S, Huang P, Xu T, Huang L, Lou J, Shi X, Sun R, Wang F, Pan S. Expression and function of Toll-like receptors in peripheral blood mononuclear cells from patients with ovarian cancer. Cancer Immunol Immunother 2015; 64:275-86. [PMID: 25376541 PMCID: PMC11029442 DOI: 10.1007/s00262-014-1632-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
Abstract
Inflammation has been implicated in the initiation and progression of ovarian cancer (OC), the underlying mechanisms of which are still unclear. We hypothesized that the abnormal expression of Toll-like receptors (TLRs), which were potential activators of nuclear factor-kappa B p65 (NF-κB p65), could promote inflammation and tumorigenesis in OC. In this study, we characterized the expression of TLRs in peripheral blood mononuclear cells (PBMCs) and found TLR2 and TLR6 mRNAs levels to be higher in PBMCs from OC patients than in those from benign disease (BC) or healthy normal controls (NC). Flow cytometry analysis showed that TLR1, TLR2 and TLR6 were highly expressed in monocytes from OC patients, but not in those from control subjects. Consistently, inflammatory cytokines interleukin (IL)-1β and IL-6 were up-regulated in PBMCs from OC patients upon stimulation with Pam3CSK4 (TLR1 ligand) and HKLM (TLR2 ligand), compared with unstimulated PBMCs. Stimulation of PBMCs with TLR ligands led to activation of downstream signaling molecules in TLRs (MyD88, TRAF6, TANK, NF-κB p65 and p-NF-κB p65). We also discovered that SK-OV-3-secreted factors were potent PBMCs activators, leading to the production of IL-1β, IL-6 and IL-8 through activation of TLRs and downstream signaling molecules in PBMCs. Before coculturing with SK-OV-3, pretreatment of THP-1 cells or PBMCs with monoclonal antibodies against TLR1, TLR2 or TLR6 inhibited the production of IL-1β and IL-6 and activation of MyD88, TRAF6, TANK, NF-κB p65 and p-NF-κB p65. Our results provided new evidence that TLR1, TLR2 and TLR6 signaling was linked with inflammation in OC microenvironment.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Juan Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Xing Ke
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Shuping Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Peijun Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Ting Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Lei Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Jianfang Lou
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Xinhui Shi
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Ruihong Sun
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| | - Shiyang Pan
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
- National Key Clinical Department of Laboratory Medicine, Nanjing, 210029 China
| |
Collapse
|
34
|
Saccani S, Trabucchi M. Regulation of stimulus-inducible gene expression in myeloid cells. Semin Immunol 2015; 27:33-43. [DOI: 10.1016/j.smim.2015.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 02/14/2015] [Accepted: 02/19/2015] [Indexed: 12/16/2022]
|
35
|
Heward JA, Roux BT, Lindsay MA. Divergent signalling pathways regulate lipopolysaccharide-induced eRNA expression in human monocytic THP1 cells. FEBS Lett 2014; 589:396-406. [PMID: 25554418 PMCID: PMC4306547 DOI: 10.1016/j.febslet.2014.12.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/08/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022]
Abstract
eRNAs are expressed from enhancers and have been shown to regulate gene expression. Expression of eRNAs is widespread upon activation of the innate immune response. We show that the NF-κB signalling pathway regulates LPS-induced eRNAs. Expression of individual eRNAs is also dependent on ERK-1/2 and p38.
Recent studies have indicated that non-coding RNAs transcribed from enhancer regions are important regulators of enhancer function and gene expression. In this report, we have characterised the expression of six enhancer RNAs (eRNAs) induced in human monocytic THP1 cells following activation of the innate immune response by lipopolysaccharide (LPS). Specifically, we have demonstrated that LPS-induced expression of individual eRNAs is mediated through divergent intracellular signalling pathways that includes NF-κB and the mitogen activated protein kinases, extracellular regulated kinase-1/2 and p38.
Collapse
Affiliation(s)
- James A Heward
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Benoit T Roux
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Mark A Lindsay
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom.
| |
Collapse
|
36
|
Abstract
Systems-level analysis of biological processes strives to comprehensively and quantitatively evaluate the interactions between the relevant molecular components over time, thereby enabling development of models that can be employed to ultimately predict behavior. Rapid development in measurement technologies (omics), when combined with the accessible nature of the cellular constituents themselves, is allowing the field of innate immunity to take significant strides toward this lofty goal. In this review, we survey exciting results derived from systems biology analyses of the immune system, ranging from gene regulatory networks to influenza pathogenesis and systems vaccinology.
Collapse
|
37
|
Malik P, Zuleger N, de las Heras JI, Saiz-Ros N, Makarov AA, Lazou V, Meinke P, Waterfall M, Kelly DA, Schirmer EC. NET23/STING promotes chromatin compaction from the nuclear envelope. PLoS One 2014; 9:e111851. [PMID: 25386906 PMCID: PMC4227661 DOI: 10.1371/journal.pone.0111851] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022] Open
Abstract
Changes in the peripheral distribution and amount of condensed chromatin are observed in a number of diseases linked to mutations in the lamin A protein of the nuclear envelope. We postulated that lamin A interactions with nuclear envelope transmembrane proteins (NETs) that affect chromatin structure might be altered in these diseases and so screened thirty-one NETs for those that promote chromatin compaction as determined by an increase in the number of chromatin clusters of high pixel intensity. One of these, NET23 (also called STING, MITA, MPYS, ERIS, Tmem173), strongly promoted chromatin compaction. A correlation between chromatin compaction and endogenous levels of NET23/STING was observed for a number of human cell lines, suggesting that NET23/STING may contribute generally to chromatin condensation. NET23/STING has separately been found to be involved in innate immune response signaling. Upon infection cells make a choice to either apoptose or to alter chromatin architecture to support focused expression of interferon genes and other response factors. We postulate that the chromatin compaction induced by NET23/STING may contribute to this choice because the cells expressing NET23/STING eventually apoptose, but the chromatin compaction effect is separate from this as the condensation was still observed when cells were treated with Z-VAD to block apoptosis. NET23/STING-induced compacted chromatin revealed changes in epigenetic marks including changes in histone methylation and acetylation. This indicates a previously uncharacterized nuclear role for NET23/STING potentially in both innate immune signaling and general chromatin architecture.
Collapse
Affiliation(s)
- Poonam Malik
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Nikolaj Zuleger
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Jose I. de las Heras
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Natalia Saiz-Ros
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandr A. Makarov
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Vassiliki Lazou
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Meinke
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin Waterfall
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Kelly
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Eric C. Schirmer
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Van den Bossche J, Neele AE, Hoeksema MA, de Winther MPJ. Macrophage polarization: the epigenetic point of view. Curr Opin Lipidol 2014; 25:367-73. [PMID: 25188918 DOI: 10.1097/mol.0000000000000109] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW The first functions of macrophages to be identified by Metchnikoff were phagocytosis and microbial killing. Although these are important features, macrophages are functionally very complex and involved in virtually all aspects of life, from immunity and host defense, to homeostasis, tissue repair and development. To accommodate for this, macrophages adopt a plethora of polarization states. Understanding their transcriptional regulation and phenotypic heterogeneity is vital because macrophages are critical in many diseases and have emerged as attractive targets for therapy. Here, we review how epigenetic mechanisms control macrophage polarization. RECENT FINDINGS It is becoming increasingly clear that chromatin remodelling governs multiple aspects of macrophage differentiation, activation and polarization. In recent years, independent research groups highlighted the importance of epigenetic mechanisms to regulate enhancer activity. Moreover, distinct histone-modifying enzymes were identified that control macrophage activation and polarization. SUMMARY We recap epigenetic features of distinct enhancers and describe the role of Jumonji domain-containing protein 3 (Jmjd3) and Hdac3 as crucial mediators of macrophage differentiation, activation and polarization. We hypothesize that epigenetic enzymes could serve as the link between environment, cellular metabolism and macrophage phenotype. To conclude, we propose epigenetic intervention as a future pharmacological target to modulate macrophage polarization and to treat inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Jan Van den Bossche
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands *Both Jan Van den Bossche and Annette E. Neele contributed equally to the article
| | | | | | | |
Collapse
|
39
|
Histone demethylase Jumonji D3 (JMJD3/KDM6B) at the nexus of epigenetic regulation of inflammation and the aging process. J Mol Med (Berl) 2014; 92:1035-43. [DOI: 10.1007/s00109-014-1182-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/27/2014] [Accepted: 06/05/2014] [Indexed: 02/03/2023]
|
40
|
Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. GENES BRAIN AND BEHAVIOR 2013; 13:69-86. [PMID: 24286462 DOI: 10.1111/gbb.12109] [Citation(s) in RCA: 437] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 11/13/2013] [Accepted: 11/25/2013] [Indexed: 12/12/2022]
Abstract
To date, there is rapidly increasing evidence for host-microbe interaction at virtually all levels of complexity, ranging from direct cell-to-cell communication to extensive systemic signalling, and involving various organs and organ systems, including the central nervous system. As such, the discovery that differential microbial composition is associated with alterations in behaviour and cognition has significantly contributed to establishing the microbiota-gut-brain axis as an extension of the well-accepted gut-brain axis concept. Many efforts have been focused on delineating a role for this axis in health and disease, ranging from stress-related disorders such as depression, anxiety and irritable bowel syndrome to neurodevelopmental disorders such as autism. There is also a growing appreciation of the role of epigenetic mechanisms in shaping brain and behaviour. However, the role of epigenetics in informing host-microbe interactions has received little attention to date. This is despite the fact that there are many plausible routes of interaction between epigenetic mechanisms and the host-microbiota dialogue. From this new perspective we put forward novel, yet testable, hypotheses. Firstly, we suggest that gut-microbial products can affect chromatin plasticity within their host's brain that in turn leads to changes in neuronal transcription and eventually alters host behaviour. Secondly, we argue that the microbiota is an important mediator of gene-environment interactions. Finally, we reason that the microbiota itself may be viewed as an epigenetic entity. In conclusion, the fields of (neuro)epigenetics and microbiology are converging at many levels and more interdisciplinary studies are necessary to unravel the full range of this interaction.
Collapse
|