1
|
Ma R, Zhang C, Zhang Y, Tan H, Zhang Y, Li Q, Bai Y, Sun X. The Impact of Respiratory Syncytial Virus on Asthma Development and Exacerbation. Ann Allergy Asthma Immunol 2025:S1081-1206(25)00236-4. [PMID: 40389152 DOI: 10.1016/j.anai.2025.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025]
Abstract
Asthma is a chronic inflammatory disorder of the lower airways clinically characterized by recurrent wheezing, breathlessness, cough, and dyspnea, and is the most prevalent chronic disease among children and adolescents. Respiratory viral infections are implicated in asthma inception and exacerbation, with respiratory syncytial virus (RSV) emerging as a key contributor. RSV is a leading cause of acute lower respiratory tract infections (LRTIs), particularly infant bronchiolitis, and is associated with a type-2-biased immune response, diminished interferon activity, epithelial barrier dysfunction, and altered airway microbiome. While the causal relationship between RSV and asthma remains debated, early-life RSV LRTIs are increasingly recognized as a significant risk factor for recurrent wheezing and asthma-like symptoms in childhood. This review comprehensively evaluates existing evidence on the long-term respiratory outcomes of infant RSV infection, elucidates the pathophysiological mechanisms connecting RSV infection to asthma development-such as immune dysregulation, chronic airway inflammation, and gene-environment interplay-and highlights novel preventive strategies. Recent advancements, such as maternal RSV vaccines and long-acting monoclonal antibodies, demonstrate efficacy in reducing severe RSV disease burden and subsequent wheeze in high-risk infants. By bridging clinical observations with mechanistic insights, this review underpins the development of future clinical therapies.
Collapse
Affiliation(s)
- Ruixue Ma
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Chenyu Zhang
- School of Basic Medicine, the Fourth Military Medical University, Xi'an, China
| | - Yi Zhang
- School of Basic Medicine, the Fourth Military Medical University, Xi'an, China
| | - Hong Tan
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yao Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Qiuhong Li
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yumei Bai
- Department of Oncology, the Central Theater Command Air Force Hospital of People's Liberation Army, Datong, China.
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Montano MM, Yeh I, Ketchart W. cGAS/STING-Independent Induction of Type I Interferon by Inhibitors of the Histone Methylase KDM5B. FASEB J 2025; 39:e70629. [PMID: 40353728 PMCID: PMC12068183 DOI: 10.1096/fj.202500628r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/16/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Studies support the role of hexamethylene bis-acetamide [HMBA] induced protein 1 (HEXIM1) as a tumor suppressor. We previously reported that the histone demethylase, KDM5B, inhibits the expression of HEXIM1, and KDM5B inhibitors (KDM5Bi) upregulate HEXIM1 expression. As a consequence, KDM5Bi inhibited cell proliferation, induced differentiation, potentiated sensitivity to cancer chemotherapy, and inhibited breast tumor metastasis. HEXIM1 is crucial for the regulation of triple-negative breast cancer (TNBC) phenotype by KDM5Bi. Type I Interferon (IFN-I) employs the immune system in the tumor microenvironment to restrict tumor growth. Moreover, therapeutic approaches (including mainstay chemotherapy) engage IFN-I signaling. We report herein that HEXIM1 and KDM5Bi induce IFN-I in TNBC. HEXIM1 and KDM5Bi downregulate the expression of polyribonucleotide nucleotidyltransferase 1 (PNPT1) resulting in the release of mitochondrial dsRNA (mt-dsRNA) into the cytoplasm. HEXIM1 also upregulates melanoma differentiation-associated protein 5 (MDA5), a cytoplasmic viral RNA receptor in the innate immune system. MDA5 is required for HEXIM1 and KDM5Bi to induce IFN-I and downstream signaling factors. We observed the augmentation of DNA damage response to Doxorubicin in the presence of KDM5Bi, and this action is a contributing factor in KDM5Bi-induced IFN-I. These actions of HEXIM1 and KDM5Bi occur independently of Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (cGAS/STING), a major DNA sensing pathway and inducer of innate immunity. Via the upregulation of HEXIM1, KDM5Bi represent pharmacologically induced and tumor intrinsic IFN-I production that is cGAS/STING independent. This is critical because cGAS/STING induce an inflammatory response that promotes the survival of cancer cells, and STING is often impaired in malignant cancers.
Collapse
Affiliation(s)
- Monica M. Montano
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
| | - I‐Ju Yeh
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Wannarasmi Ketchart
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
- Department of Pharmacology, Faculty of MedicineChulalongkorn UniversityBangkokThailand
| |
Collapse
|
3
|
Lian X, Wang W, Li Y, Zhou X, Li X, Zuo J, Song L, Wang L, Song L. Histone lysine demethylase 5 regulates haemocyte proliferation during immune priming in the oyster Crassostreagigas. FISH & SHELLFISH IMMUNOLOGY 2025; 160:110236. [PMID: 40010616 DOI: 10.1016/j.fsi.2025.110236] [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: 11/27/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
Histone lysine demethylase 5 (KDM5) is a ketoglutarate-dependent dioxygenase in histone lysine demethylation, playing a vital role in immunological memory by modulating H3K4me3. Investigating on the role of invertebrate KDM5 in the immune priming, a novel form of immunological memory recently verified in invertebrates, will further our knowledge of epigenetic regulation for innate immune memory. In the present study, a KDM5A was identified in Pacific oyster Crassostrea gigas, and its role in regulating haemocyte proliferation during immune priming was assessed. The deduced amino acid sequence of CgKDM5A harbors a complete JmjC domain featuring a conserved αKG binding site. The mRNA expression of CgKDM5A in the haemolymph was significantly higher than that in the tested tissues of the mature oysters. After Vibrio splendidus stimulation, CgKDM5A transcripts and KDM5 enzymatic activity in haemocytes significantly decreased, accompanying with increased H3K4me3 levels. Moreover, H3K4me3 modifications at the CgBMP7 and CgGATA2/3 promoters were elevated at 7 d after V. splendidus stimulation (p < 0.05), and the haemocyte proliferation index increased significantly at 12 h after the secondary stimulation (p < 0.05). Treatment with KDM5 activator DM-αKG further led to a significant increase in H3K4me3 enrichment levels at the CgBMP7 and CgGATA2/3 promoters at 7 d after the primary stimulation (p < 0.05). Subsequently, the expression of CgBMP7 and CgGATA2/3, as well as the haemocyte proliferation index decreased significantly after the secondary stimulation (p < 0.05). In contrast, CgKDM5A-RNAi oysters exhibited an enriched H3K4me3 at the CgBMP7 and CgGATA2/3 promoters at 7 d after the primary stimulation and an increased haemocyte proliferation index at 12 h after the secondary stimulation (p < 0.05). These findings suggest that CgKDM5A plays a critical role in haemocyte proliferation by affect H3K4me3 enrichment at the CgBMP7 and CgGATA2/3 promoters during immune priming in C. gigas, highlighting the potential of epigenetic regulation in innate immune memory of mollusks.
Collapse
Affiliation(s)
- Xingye Lian
- School of Life Science, Liaoning Normal University, Dalian, 116029, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Yinan Li
- School of Life Science, Liaoning Normal University, Dalian, 116029, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoxu Zhou
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xuesong Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Jiajun Zuo
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingyuan Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
4
|
Kraus L, Fredericks S, Scheeler K. The epigenetic regulation of crosstalk between cardiac fibroblasts and other cardiac cell types during stress. Front Cardiovasc Med 2025; 12:1539826. [PMID: 40264508 PMCID: PMC12011845 DOI: 10.3389/fcvm.2025.1539826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
With the global impact of cardiovascular disease, there is a dire need to understand the mechanisms in the heart during injury and stress. It has been shown that the regulation of the extracellular matrix via cardiac fibroblasts plays a major role in the progression of heart failure and worsening function of the heart. Importantly, it has been suggested that crosstalk between other cardiac cells like cardiomyocytes, immune cells, and endothelial cells are influenced by the pathological function of the fibroblasts. This decline in function across all cardiac cells is seemingly irreversible. However, epigenetic mechanisms have been shown to regulate functionality across cardiac cells and improve outcomes during stress or injury. This epigenetic regulation has also been shown to control communication between different cell types and influence the role of multiple cardiac cell types during injury. The goal of this review is to summarize and discuss the current research of epigenetic regulation of cardiac fibroblasts and the subsequent crosstalk with other cardiac cell types in cardiovascular disease states.
Collapse
Affiliation(s)
- Lindsay Kraus
- Department of Biology, College of Science, Technology, Engineering, Arts, and Mathematics, Alvernia University, Reading, PA, United States
| | | | | |
Collapse
|
5
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
6
|
Das S, Lavine KJ. Role of Trained Immunity in Heath and Disease. Curr Cardiol Rep 2025; 27:18. [PMID: 39804563 DOI: 10.1007/s11886-024-02167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 05/02/2025]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of immune memory and trained immunity, focusing on how innate immune cells like monocytes, macrophages, and natural killer cells undergo long-term epigenetic and metabolic rewiring. Specifically, it examines the mechanisms by which trained immunity, often triggered by infection or vaccination, could impact cardiac processes and contribute to both protective and pathological responses within the cardiovascular system. RECENT FINDINGS Recent research demonstrates that vaccination and infection not only activate immune responses in circulating monocytes and tissue macrophages but also affect immune progenitor cells within the bone marrow environment, conferring lasting protection against heterologous infections. These protective effects are attributed to epigenetic and metabolic reprogramming, which enable a heightened immune response upon subsequent encounters with pathogens. However, while trained immunity is beneficial in combating infections, it has been linked to exacerbated inflammation, which may increase susceptibility to cardiovascular diseases, including atherosclerosis and heart failure. Our review highlights the dual nature of trained immunity: while it offers protective advantages against infections, it also poses potential risks for cardiovascular health by promoting chronic inflammation. Understanding the molecular mechanisms underlying immune memory's impact on cardiac processes could lead to new therapeutic strategies to mitigate cardiovascular diseases, such as atherosclerosis, heart failure, and diabetes. These insights build the grounds for future research to balance the benefits of trained immunity with its potential risks in cardiovascular disease management.
Collapse
Affiliation(s)
- Shibali Das
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA.
| |
Collapse
|
7
|
Ma Y, Lv W, Guo Y, Yin T, Bai Y, Liu Z, Chen C, WenjuanYang, Feng J, Qian W, Tang R, Su Y, Shan S, Dong H, Bao Y, Qu L. Histone demethylases in autophagy and inflammation. Cell Commun Signal 2025; 23:24. [PMID: 39806430 PMCID: PMC11727796 DOI: 10.1186/s12964-024-02006-w] [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: 06/21/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy dysfunction is associated with changes in autophagy-related genes. Various factors are connected to autophagy, and the mechanism regulating autophagy is highly complicated. Epigenetic changes, such as aberrant expression of histone demethylase, are actively associated not only with oncogenesis but also with inflammatory responses. Among post-translational modifications, histone lysine methylation holds significant importance. There are over 30 members of histone lysine demethylases (KDMs), which act as epigenetic regulators in physiological processes and diseases. Importantly, KDMs are abnormally expressed in the regulation of cellular autophagy and inflammation, representing a crucial mechanism affecting inflammation-related diseases. This article reviewed the function of KDMs proteins in autophagy and inflammation. Specifically, It focused on the specific regulatory mechanisms underlying the activation or inhibition of autophagy, as well as their abnormal expression in inflammatory responses. By analyzing each KDM in epigenetic modification, this review provides a reliable theoretical basis for clinical decision marking regarding autophagy abnormalities and inflammatory diseases.
Collapse
Affiliation(s)
- Yaoyao Ma
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Hubei, 437000, China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Hubei, 437000, China
| | - Wenting Lv
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China
| | - Yi Guo
- School of Basic Medical Sciences, Hubei University of Science and Technology, Hubei, 437000, China
| | - Tong Yin
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China
| | - Yujie Bai
- Department of Scientific Research and Education, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Ziqi Liu
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China
| | - Chao Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - WenjuanYang
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China
| | - Jiayi Feng
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China
| | - Wenbin Qian
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Hubei, 437000, China
| | - Ruiling Tang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Hubei, 437000, China
| | - Yanting Su
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Hubei, 437000, China
| | - Shigang Shan
- School of Public Health and Nursing, Hubei University of Science and Technology, Hubei, 437000, China
| | - Huifen Dong
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China.
| | - Yongfen Bao
- School of Basic Medical Sciences, Hubei University of Science and Technology, Hubei, 437000, China.
| | - Lihua Qu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Hubei, 437000, China.
- 3Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, 430071, China.
| |
Collapse
|
8
|
Steigmann JC, Zhou X, Suttenberg LN, Salman I, Rehmathullah ZF, Weinberg JB. Effects of immunoproteasome inhibition on acute respiratory infection with murine hepatitis virus strain 1. J Virol 2024; 98:e0123824. [PMID: 39508578 PMCID: PMC11650983 DOI: 10.1128/jvi.01238-24] [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: 07/12/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
The immunoproteasome (IP) is a predominantly inducible component of the ubiquitin proteasome system that plays key roles in multiple aspects of immune function, inflammation, and protein homeostasis. We used murine hepatitis virus strain 1 (MHV-1), a mouse coronavirus, to define the role of IP activity during acute coronavirus respiratory infection. Expression of the β5i subunit of the IP and cytokines that induce IP activity, including IFN-γ, TNF-α, and IFN-β, increased in lungs and livers of CH3/HeJ mice following intranasal infection with MHV-1. IP inhibition using ONX-0914 did not affect MHV-1 replication in bone marrow-derived dendritic cells in vitro. IP inhibition in vivo exacerbated virus-induced weight loss and mortality but had no effect on virus replication in lungs or livers. IP inhibition had minimal effect on virus-induced pulmonary inflammation but led to substantially increased liver pathology, including greater upregulation of pro-inflammatory cytokines and histological evidence of inflammation and necrosis. Those findings were associated with evidence of increased endoplasmic reticulum stress although not with accumulation of ubiquitinated protein. Our results indicate that the IP is a protective host factor during acute MHV-1 infection. IMPORTANCE Inflammatory responses triggered by acute infection by respiratory viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) drive morbidity and mortality. Infection of mice with murine hepatitis virus strain 1 (MHV-1), a mouse coronavirus, is a useful model to study the pathogenesis of coronavirus respiratory infections. The immunoproteasome is an inducible component of the ubiquitin proteasome system that is poised to contribute to multiple aspects of immune function, inflammation, and protein homeostasis during an infection. We used the MHV-1 model to define the role of the immunoproteasome in coronavirus pathogenesis. We found that immunoproteasome subunit expression increases in the lungs and the liver during acute MHV-1 respiratory infection. Inhibition of immunoproteasome activity did not affect MHV-1 replication but increased MHV-1-induced weight loss, mortality, and inflammation in lungs and livers. Thus, our findings indicate that the immunoproteasome is a critical protective host factor during coronavirus respiratory infection.
Collapse
Affiliation(s)
- Jacob C. Steigmann
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaofeng Zhou
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lauren N. Suttenberg
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Irha Salman
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Zainab F. Rehmathullah
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason B. Weinberg
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Hojjatipour T, Ajeli M, Maali A, Azad M. Epigenetic-modifying agents: The potential game changers in the treatment of hematologic malignancies. Crit Rev Oncol Hematol 2024; 204:104498. [PMID: 39244179 DOI: 10.1016/j.critrevonc.2024.104498] [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: 06/10/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
Hematologic malignancies are lethal diseases arising from accumulated leukemic cells with substantial genetic or epigenetic defects in their natural development. Epigenetic modifications, including DNA methylation and histone modifications, are critical in hematologic malignancy formation, propagation, and treatment response. Both mutations and aberrant recruitment of epigenetic modifiers are reported in different hematologic malignancies, which regarding the reversible nature of epigenetic regulations, make them a potential target for cancer treatment. Here, we have first outlined a comprehensive overview of current knowledge related to epigenetic regulation's impact on the development and prognosis of hematologic malignancies. Furthermore, we have presented an updated overview regarding the current status of epigenetic-based drugs in hematologic malignancies treatment. And finally, discuss current challenges and ongoing clinical trials based on the manipulation of epigenetic modifies in hematologic malignancies.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Cancer Immunology Group, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Mina Ajeli
- Department of Medical Laboratory Sciences, Guilan University of Medical Sciences, Guilan, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
10
|
Zar HJ, Cacho F, Kootbodien T, Mejias A, Ortiz JR, Stein RT, Hartert TV. Early-life respiratory syncytial virus disease and long-term respiratory health. THE LANCET. RESPIRATORY MEDICINE 2024; 12:810-821. [PMID: 39265601 DOI: 10.1016/s2213-2600(24)00246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/14/2024]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection (LRTI), hospital admission, and mortality in children worldwide. Early-life RSV LRTI has also been associated with subsequent long-term respiratory sequelae, including recurrent LRTI, recurrent wheezing, asthma, and lung function impairment, and these effects can persist into adulthood as chronic respiratory disease. New preventive measures (maternal vaccine or long-acting monoclonal antibodies) have been licensed to reduce the burden of acute RSV LRTI in infants and children at high risk through passive immunisation. Studies of these RSV prevention products show high efficacy and effectiveness, particularly for preventing severe RSV LRTI, with implementation in many high-income countries, but limited access in low-income and middle-income countries (LMICs). These interventions might also reduce the risk of additional health outcomes and long-term morbidity. This Series paper provides the evidence for the long-term effects of early-life RSV disease, discusses mechanisms of disease development, and addresses the potential full public health value of prevention of RSV illness. Further research is needed to determine whether prevention of RSV LRTI or delay of RSV illness in early life might prevent or ameliorate the development of associated long-term respiratory disease. This potential further underscores the urgency for access and availability of new interventions to prevent early-life RSV LRTI in LMICs.
Collapse
Affiliation(s)
- Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa.
| | - Ferdinand Cacho
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tahira Kootbodien
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Asuncion Mejias
- Department of Infectious Disease, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Renato T Stein
- Department of Pediatrics, School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tina V Hartert
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
11
|
Li TW, Park Y, Watters EG, Wang X, Zhou D, Fiches GN, Wu Z, Badley AD, Sacha JB, Ho WZ, Santoso NG, Qi J, Zhu J. KDM5A/B contribute to HIV-1 latent infection and survival of HIV-1 infected cells. Antiviral Res 2024; 228:105947. [PMID: 38925368 PMCID: PMC11927087 DOI: 10.1016/j.antiviral.2024.105947] [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: 12/26/2023] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Combinational antiretroviral therapy (cART) suppresses human immunodeficiency virus type 1 (HIV-1) viral replication and pathogenesis in acquired immunodeficiency syndrome (AIDS) patients. However, HIV-1 remains in the latent stage of infection by suppressing viral transcription, which hinders an HIV-1 cure. One approach for an HIV-1 cure is the "shock and kill" strategy. The strategy focuses on reactivating latent HIV-1, inducing the viral cytopathic effect and facilitating the immune clearance for the elimination of latent HIV-1 reservoirs. Here, we reported that the H3K4 trimethylation (H3K4me3)-specific demethylase KDM5A/B play a role in suppressing HIV-1 Tat/LTR-mediated viral transcription in HIV-1 latent cells. Furthermore, we evaluated the potential of KDM5-specific inhibitor JQKD82 as an HIV-1 "shock and kill" agent. Our results showed that JQKD82 increases the H3K4me3 level at HIV-1 5' LTR promoter regions, HIV-1 reactivation, and the cytopathic effects in an HIV-1-latent T cell model. In addition, we identified that the combination of JQKD82 and AZD5582, a non-canonical NF-κB activator, generates a synergistic impact on inducing HIV-1 lytic reactivation and cell death in the T cell. The latency-reversing potency of the JQKD82 and AZD5582 pair was also confirmed in peripheral blood mononuclear cells (PBMCs) isolated from HIV-1 aviremic patients and in an HIV-1 latent monocyte. In latently infected microglia (HC69) of the brain, either deletion or inhibition of KDM5A/B results in a reversal of the HIV-1 latency. Overall, we concluded that KDM5A/B function as a host repressor of the HIV-1 lytic reactivation and thus promote the latency and the survival of HIV-1 infected reservoirs.
Collapse
Affiliation(s)
- Tai-Wei Li
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Youngmin Park
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Emily G Watters
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Dawei Zhou
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Guillaume N Fiches
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhenyu Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, 55902, USA
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Netty G Santoso
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jun Qi
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Jian Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Maity J, Majumder S, Pal R, Saha B, Mukhopadhyay PK. Ascorbic acid modulates immune responses through Jumonji-C domain containing histone demethylases and Ten eleven translocation (TET) methylcytosine dioxygenase. Bioessays 2023; 45:e2300035. [PMID: 37694689 DOI: 10.1002/bies.202300035] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023]
Abstract
Ascorbic acid is a redox regulator in many physiological processes. Besides its antioxidant activity, many intriguing functions of ascorbic acid in the expression of immunoregulatory genes have been suggested. Ascorbic acid acts as a co-factor for the Fe+2 -containing α-ketoglutarate-dependent Jumonji-C domain-containing histone demethylases (JHDM) and Ten eleven translocation (TET) methylcytosine dioxygenasemediated epigenetic modulation. By influencing JHDM and TET, ascorbic acid facilitates the differentiation of double negative (CD4- CD8- ) T cells to double positive (CD4+ CD8+ ) T cells and of T-helper cells to different effector subsets. Ascorbic acid modulates plasma cell differentiation and promotes early differentiation of hematopoietic stem cells (HSCs) to NK cells. These findings indicate that ascorbic acid plays a significant role in regulating both innate and adaptive immune cells, opening up new research areas in Immunonutrition. Being a water-soluble vitamin and a safe micro-nutrient, ascorbic acid can be used as an adjunct therapy for many disorders of the immune system.
Collapse
Affiliation(s)
- Jeet Maity
- Department of Life Sciences, Presidency University, Kolkata, India
| | | | - Ranjana Pal
- Department of Life Sciences, Presidency University, Kolkata, India
| | | | | |
Collapse
|
13
|
Ge L, Wang Y, Liu Z, Du H, Zhao D. Chitinase 3-like 1 plays a pivotal role in airway response of RSV infection via regulating DC functional transition. Int Immunopharmacol 2023; 124:110819. [PMID: 37607465 DOI: 10.1016/j.intimp.2023.110819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Dendritic cells (DCs) contribute to immune imbalance and airway hyperresponsiveness (AHR) induced by respiratory syncytial virus (RSV). The aim of present study was to explore the mechanism of RSV regulating naive T cell differentiation through DCs. METHODS We generated a Lentivirus shRNA expression vector to knock down CHI3L1 in mouse lungs and bone marrow-derived dendritic cells (BMDCs). Then we investigated the effect of CHI3L1 knockdown on MAPK/ERK pathway, PI3K/AKT pathway, mature DCs represented by molecular markers, naive T cell differentiation and related cytokine expression in vitro and in vivo models of RSV. RESULTS RSV elevated CHI3L1 expression in lung DCs and BMDCs. Knockdown of CHI3L1 impeded RSV-induced activation of MAPK/ERK and PI3K/AKT signaling pathways, attenuated CD86 and OX40L expression in mature DCs, reduced the proportion of Th2 and Th17 cells, and increased the proportion of Treg cells. In addition, by blocking CHI3L1, RSV-infected mice shown relief of airway resistance, the downregulation of Th2/Th17 like cytokines IL-4, IL-13 and IL-17 levels, and the upregulation of IL-10. CONCLUSION Our data show that CHI3L1 promotes RSV induced immune imbalance and airway hyperresponsiveness by regulating the functional transformation of DCs.
Collapse
Affiliation(s)
- Lingli Ge
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
| | - Yuxin Wang
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhi Liu
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Hui Du
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Dongchi Zhao
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Children's digital health and data Center of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Fazazi MR, Ruda GF, Brennan PE, Rangachari M. The X-linked histone demethylases KDM5C and KDM6A as regulators of T cell-driven autoimmunity in the central nervous system. Brain Res Bull 2023; 202:110748. [PMID: 37657612 DOI: 10.1016/j.brainresbull.2023.110748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023]
Abstract
T cell-driven autoimmune responses are subject to striking sex-dependent effects. While the contributions of sex hormones are well-understood, those of sex chromosomes are meeting with increased appreciation. Here, we outline what is known about the contribution of sex chromosome-linked factors to experimental autoimmune encephalomyelitis (EAE), a mouse model that recapitulates many of the T cell-driven mechanisms of multiple sclerosis (MS) pathology. Particular attention is paid to the KDM family of histone demethylases, several of which - KDM5C, KDM5D and KDM6A - are sex chromosome encoded. Finally, we provide evidence that functional inhibition of KDM5 molecules can suppress interferon (IFN)γ production from murine male effector T cells, and that an increased ratio of inflammatory Kdm6a to immunomodulatory Kdm5c transcript is observed in T helper 17 (Th17) cells from women with the autoimmune disorder ankylosing spondylitis (AS). Histone lysine demethlyases thus represent intriguing targets for the treatment of T cell-driven autoimmune disorders.
Collapse
Affiliation(s)
- Mohamed Reda Fazazi
- axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Gian Filippo Ruda
- Centre for Medicines Discovery and NIHR, Oxford Biomedical Research Centre, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Paul E Brennan
- Centre for Medicines Discovery and NIHR, Oxford Biomedical Research Centre, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; Alzheimer's Research UK, Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Manu Rangachari
- axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
15
|
Malinczak CA, Fonseca W, Mire MM, Parolia A, Chinnaiyan A, Rasky AJ, Morris S, Yagi K, Bermick JR, Lukacs NW. Sex-associated early-life viral innate immune response is transcriptionally associated with chromatin remodeling of type-I IFN-inducible genes. Mucosal Immunol 2023; 16:578-592. [PMID: 37302711 PMCID: PMC10646734 DOI: 10.1016/j.mucimm.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/15/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
This study investigates sex-associated systemic innate immune differences by examining bone marrow-derived dendritic cells (BMDCs). BMDC grown from 7-day-old mice show enhanced type-I interferon (IFN) signaling in female compared to male BMDC. Upon respiratory syncytial virus (RSV) infection of 7-day-old mice, a significantly altered phenotype of BMDC at 4 weeks post-infection is observed in a sex-dependent manner. The alterations include heightened Ifnb/ interleukin (Il12a) and enhanced IFNAR1+ expression in BMDC from early-life RSV-infected female mice that leads to increased IFN-γ production by T cells. Phenotypic differences were verified upon pulmonary sensitization whereby EL-RSV male-derived BMDC promoted enhanced T helper 2/17 responses and exacerbated disease upon RSV infection while EL-RSV/F BMDC sensitization was relatively protective. Assay for transposase-accessible chromatin using sequencing analysis (ATAC-seq) demonstrated that EL-RSV/F BMDC had enhanced chromatin accessibility near type-I immune genes with JUN, STAT1/2, and IRF1/8 transcription factors predicted to have binding sites in accessible regions. Importantly, ATAC-seq of human cord blood-derived monocytes displayed a similar sex-associated chromatin landscape with female-derived monocytes having more accessibility in type-I immune genes. These studies enhance our understanding of sex-associated differences in innate immunity by epigenetically controlled transcriptional programs amplified by early-life infection in females via type-I immunity.
Collapse
Affiliation(s)
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Mohamed M Mire
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, USA
| | - Arul Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, USA; Howard Hughes Medical Institute, University of Michigan, Ann Arbor, USA
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Susan Morris
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Kazuma Yagi
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | | | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, USA; Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
16
|
Qu L, Yin T, Zhao Y, Lv W, Liu Z, Chen C, Liu K, Shan S, Zhou R, Li X, Dong H. Histone demethylases in the regulation of immunity and inflammation. Cell Death Discov 2023; 9:188. [PMID: 37353521 DOI: 10.1038/s41420-023-01489-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023] Open
Abstract
Pathogens or danger signals trigger the immune response. Moderate immune response activation removes pathogens and avoids excessive inflammation and tissue damage. Histone demethylases (KDMs) regulate gene expression and play essential roles in numerous physiological processes by removing methyl groups from lysine residues on target proteins. Abnormal expression of KDMs is closely associated with the pathogenesis of various inflammatory diseases such as liver fibrosis, lung injury, and autoimmune diseases. Despite becoming exciting targets for diagnosing and treating these diseases, the role of these enzymes in the regulation of immune and inflammatory response is still unclear. Here, we review the underlying mechanisms through which KDMs regulate immune-related pathways and inflammatory responses. In addition, we also discuss the future applications of KDMs inhibitors in immune and inflammatory diseases.
Collapse
Affiliation(s)
- Lihua Qu
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Tong Yin
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yijin Zhao
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wenting Lv
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Ziqi Liu
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chao Chen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kejun Liu
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Shigang Shan
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Rui Zhou
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaoqing Li
- Biological Targeted Therapy Key Laboratory in Hubei, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Huifen Dong
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei, China.
- Department of Pathogenic Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
17
|
Zhang Y, Gao Y, Jiang Y, Ding Y, Chen H, Xiang Y, Zhan Z, Liu X. Histone demethylase KDM5B licenses macrophage-mediated inflammatory responses by repressing Nfkbia transcription. Cell Death Differ 2023; 30:1279-1292. [PMID: 36914768 PMCID: PMC10154333 DOI: 10.1038/s41418-023-01136-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 03/16/2023] Open
Abstract
Macrophages play a critical role in the immune homeostasis and host defense against invading pathogens. However, uncontrolled activation of inflammatory macrophages leads to tissue injury and even fuels autoimmunity. Hence the molecular mechanisms underlying macrophage activation need to be further elucidated. The effects of epigenetic modifications on the function of immune cells draw increasing attention. Here, we demonstrated that lysine-specific demethylase 5B (KDM5B), a classical transcriptional repressor in stem cell development and cancer, was required for the full activation of NF-κB signaling cascade and pro-inflammatory cytokine production in macrophages. KDM5B deficiency or inhibitor treatment protected mice from immunologic injury in both collagen-induced arthritis (CIA) model and endotoxin shock model. Genome-wide analysis of KDM5B-binding peaks identified that KDM5B was selectively recruited to the promoter of Nfkbia, the gene encoding IκBα, in activated macrophages. KDM5B mediated the H3K4me3 modification erasing and decreased chromatin accessibility of Nfkbia gene locus, coordinating the elaborate suppression of IκBα expression and the enhanced NF-κB-mediated macrophage activation. Our finding identifies the indispensable role of KDM5B in macrophage-mediated inflammatory responses and provides a candidate therapeutic target for autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Yunkai Zhang
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of Medical Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ying Gao
- Department of Rheumatology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuyu Jiang
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Yingying Ding
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Huiying Chen
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Yan Xiang
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Zhenzhen Zhan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of Liver Surgery, Shanghai Institute of Transplantation, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Xingguang Liu
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China.
- National Key Laboratory of Medical Immunology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Tovo PA, Garazzino S, Savino F, Daprà V, Pruccoli G, Dini M, Filisetti G, Funiciello E, Galliano I, Bergallo M. Expressions of Type I and III Interferons, Endogenous Retroviruses, TRIM28, and SETDB1 in Children with Respiratory Syncytial Virus Bronchiolitis. Curr Issues Mol Biol 2023; 45:1197-1217. [PMID: 36826024 PMCID: PMC9954910 DOI: 10.3390/cimb45020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Interferons (IFNs) and IFN-stimulated genes (ISGs) play essential roles for the control of viral infections. Their expression in infants with respiratory syncytial virus (RSV) bronchiolitis is poorly defined. Human endogenous retroviruses (HERVs) represent 8% of our genome and modulate inflammatory and immune reactions. TRIM28 and SETDB1 participate in the epigenetic regulation of genes involved in the immune response, including IFNs and HERVs. No study has explored the expression of HERVs, TRIM28, and SETDB1 during RSV bronchiolitis. We assessed, through a PCR real-time Taqman amplification assay, the transcription levels of six IFN-I ISGs, four IFNλs, the pol genes of HERV-H, -K, and -W families, the env genes of Syncytin (SYN)1 and SYN2, and of TRIM28/SETDB1 in whole blood from 37 children hospitalized for severe RSV bronchiolitis and in healthy children (HC). The expression of most IFN-I ISGs was significantly higher in RSV+ patients than in age-matched HC, but it was inhibited by steroid therapy. The mRNA concentrations of IFN-λs were comparable between patients and age-matched HC. This lack of RSV-driven IFN-III activation may result in the defective protection of the airway mucosal surface leading to severe bronchiolitis. The expression of IFN-III showed a positive correlation with age in HC, that could account for the high susceptibility of young children to viral respiratory tract infections. The transcription levels of every HERV gene were significantly lower in RSV+ patients than in HC, while the expressions of TRIM28/SETDB1 were overlapping. Given the negative impact of HERVs and the positive effects of TRIM28/SETDB1 on innate and adaptive immune responses, the downregulation of the former and the normal expression of the latter may contribute to preserving immune functions against infection.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Correspondence: Correspondence: (P.-A.T.); (M.B.)
| | - Silvia Garazzino
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Francesco Savino
- Early Infancy Special Care Unit, Department of Pediatric Care, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Valentina Daprà
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Giulia Pruccoli
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Maddalena Dini
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Giacomo Filisetti
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Early Infancy Special Care Unit, Department of Pediatric Care, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Elisa Funiciello
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Ilaria Galliano
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Massimiliano Bergallo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Correspondence: Correspondence: (P.-A.T.); (M.B.)
| |
Collapse
|
19
|
Baghel VS, Shinde S, Sinha V, Dixit V, Tiwari AK, Saxena S, Vishvakarma NK, Shukla D, Bhatt P. Inhibitors targeting epigenetic modifications in cancer. TRANSCRIPTION AND TRANSLATION IN HEALTH AND DISEASE 2023:287-324. [DOI: 10.1016/b978-0-323-99521-4.00007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Zhang SM, Cao J, Yan Q. KDM5 Lysine Demethylases in Pathogenesis, from Basic Science Discovery to the Clinic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1433:113-137. [PMID: 37751138 DOI: 10.1007/978-3-031-38176-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The histone lysine demethylase 5 (KDM5) family proteins are Fe2+ and α-ketoglutarate-dependent dioxygenases, with jumonji C (JmjC) domain as their catalytic core and several plant homeodomains (PHDs) to bind different histone methylation marks. These enzymes are capable of demethylating tri-, di- and mono-methylated lysine 4 in histone H3 (H3K4me3/2/1), the key epigenetic marks for active chromatin. Thus, this H3K4 demethylase family plays critical roles in cell fate determination during development as well as malignant transformation. KDM5 demethylases have both oncogenic and tumor suppressive functions in a cancer type-dependent manner. In solid tumors, KDM5A/B are generally oncogenic, whereas KDM5C/D have tumor suppressive roles. Their involvement in de-differentiation, cancer metastasis, drug resistance, and tumor immunoevasion indicated that KDM5 family proteins are promising drug targets for cancer therapy. Significant efforts from both academia and industry have led to the development of potent and selective KDM5 inhibitors for preclinical experiments and phase I clinical trials. However, a better understanding of the roles of KDM5 demethylases in different physiological and pathological conditions is critical for further developing KDM5 modulators for clinical applications.
Collapse
Affiliation(s)
- Shang-Min Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Jian Cao
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
| | - Qin Yan
- Department of Pathology, Yale Cancer Center, Yale Stem Cell Center, Yale Center for Immuno-Oncology, Yale Center for Research on Aging, Yale School of Medicine, P.O. Box 208023, New Haven, CT, 06520-8023, USA.
| |
Collapse
|
21
|
Loss of KDM5B ameliorates pathological cardiac fibrosis and dysfunction by epigenetically enhancing ATF3 expression. Exp Mol Med 2022; 54:2175-2187. [PMID: 36481938 PMCID: PMC9794816 DOI: 10.1038/s12276-022-00904-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/26/2022] [Accepted: 10/24/2022] [Indexed: 12/13/2022] Open
Abstract
Excessive cardiac fibrosis is central to adverse cardiac remodeling and dysfunction leading to heart failure in many cardiac diseases. Histone methylation plays a crucial role in various pathophysiological events. However, the role of histone methylation modification enzymes in pathological cardiac fibrosis needs to be fully elucidated. Here, we identified lysine demethylase 5B (KDM5B), a histone H3K4me2/me3 demethylase, as a key epigenetic mediator of pathological cardiac fibrosis. KDM5B expression was upregulated in cardiac fibroblasts and myocardial tissues in response to pathological stress. KDM5B deficiency markedly ameliorated cardiac fibrosis, improved cardiac function, and prevented adverse cardiac remodeling following myocardial infarction (MI) or pressure overload. KDM5B knockout or inhibitor treatment constrained the transition of cardiac fibroblasts to profibrogenic myofibroblasts and suppressed fibrotic responses. KDM5B deficiency also facilitated the transformation of cardiac fibroblasts to endothelial-like cells and promoted angiogenesis in response to myocardial injury. Mechanistically, KDM5B bound to the promoter of activating transcription factor 3 (Atf3), an antifibrotic regulator of cardiac fibrosis, and inhibited ATF3 expression by demethylating the activated H3K4me2/3 modification, leading to the enhanced activation of TGF-β signaling and excessive expression of profibrotic genes. Our study indicates that KDM5B drives pathological cardiac fibrosis and represents a candidate target for intervention in cardiac dysfunction and heart failure.
Collapse
|
22
|
Gilmore SA, Tam D, Cheung TL, Snyder C, Farand J, Dick R, Matles M, Feng JY, Ramirez R, Li L, Yu H, Xu Y, Barnes D, Czerwieniec G, Brendza KM, Appleby TC, Birkus G, Willkom M, Kobayashi T, Paoli E, Labelle M, Boesen T, Tay CH, Delaney WE, Notte GT, Schmitz U, Feierbach B. Characterization of a KDM5 small molecule inhibitor with antiviral activity against hepatitis B virus. PLoS One 2022; 17:e0271145. [PMID: 36477212 PMCID: PMC9728921 DOI: 10.1371/journal.pone.0271145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatitis B (CHB) is a global health care challenge and a major cause of liver disease. To find new therapeutic avenues with a potential to functionally cure chronic Hepatitis B virus (HBV) infection, we performed a focused screen of epigenetic modifiers to identify potential inhibitors of replication or gene expression. From this work we identified isonicotinic acid inhibitors of the histone lysine demethylase 5 (KDM5) with potent anti-HBV activity. To enhance the cellular permeability and liver accumulation of the most potent KDM5 inhibitor identified (GS-080) an ester prodrug was developed (GS-5801) that resulted in improved bioavailability and liver exposure as well as an increased H3K4me3:H3 ratio on chromatin. GS-5801 treatment of HBV-infected primary human hepatocytes reduced the levels of HBV RNA, DNA and antigen. Evaluation of GS-5801 antiviral activity in a humanized mouse model of HBV infection, however, did not result in antiviral efficacy, despite achieving pharmacodynamic levels of H3K4me3:H3 predicted to be efficacious from the in vitro model. Here we discuss potential reasons for the disconnect between in vitro and in vivo efficacy, which highlight the translational difficulties of epigenetic targets for viral diseases.
Collapse
Affiliation(s)
- Sarah A. Gilmore
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Danny Tam
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Tara L. Cheung
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Chelsea Snyder
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Julie Farand
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Ryan Dick
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Mike Matles
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Joy Y. Feng
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Ricardo Ramirez
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Li Li
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Helen Yu
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Yili Xu
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Dwight Barnes
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Gregg Czerwieniec
- Gilead Sciences, Inc., Foster City, California, United States America
| | | | - Todd C. Appleby
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Gabriel Birkus
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Madeleine Willkom
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Tetsuya Kobayashi
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Eric Paoli
- Gilead Sciences, Inc., Foster City, California, United States America
| | | | - Thomas Boesen
- EpiTherapeutics ApS, Copenhagen, Denmark
- Novo Nordisk A/S, Bagsvaerd, Denmark
| | - Chin H. Tay
- Gilead Sciences, Inc., Foster City, California, United States America
| | | | - Gregory T. Notte
- Gilead Sciences, Inc., Foster City, California, United States America
| | - Uli Schmitz
- Gilead Sciences, Inc., Foster City, California, United States America
- * E-mail:
| | - Becket Feierbach
- Gilead Sciences, Inc., Foster City, California, United States America
| |
Collapse
|
23
|
Pischedda S, Rivero-Calle I, Gómez-Carballa A, Cebey-López M, Barral-Arca R, Gómez-Rial J, Pardo-Seco J, Curras-Tuala MJ, Viz-Lasheras S, Bello X, Crujeiras AB, Diaz-Lagares A, González-López MT, Martinón-Torres F, Salas A. Role and Diagnostic Performance of Host Epigenome in Respiratory Morbidity after RSV Infection: The EPIRESVi Study. Front Immunol 2022; 13:875691. [PMID: 35619695 PMCID: PMC9128527 DOI: 10.3389/fimmu.2022.875691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) infection has been associated with the subsequent development of recurrent wheezing and asthma, although the mechanisms involved are still unknown. We investigate the role of epigenetics in the respiratory morbidity after infection by comparing methylation patterns from children who develop recurrent wheezing (RW-RSV), subsequent asthma (AS-RVS), and those experiencing complete recovery (CR-RSV). Methods Prospective, observational study of infants aged < 2 years with RSV respiratory infection admitted to hospital and followed-up after discharge for at least three years. According to their clinical course, patients were categorized into subgroups: RW-RSV (n = 36), AS-RSV (n = 9), and CR-RSV (n = 32). The DNA genome-wide methylation pattern was analyzed in whole blood samples, collected during the acute phase of the infection, using the Illumina Infinium Methylation EPIC BeadChip (850K CpG sites). Differences in methylation were determined through a linear regression model adjusted for age, gender and cell composition. Results Patients who developed respiratory sequelae showed a statistically significant higher proportion of NK and CD8T cells (inferred through a deconvolution approach) than those with complete recovery. We identified 5,097 significant differentially methylated positions (DMPs) when comparing RW-RSV and AS-RVS together against CR-RSV. Methylation profiles affect several genes involved in airway inflammation processes. The most significant DMPs were found to be hypomethylated in cases and therefore generally leading to overexpression of affected genes. The lead CpG position (cg24509398) falls at the gene body of EYA3 (P-value = 2.77×10-10), a tyrosine phosphatase connected with pulmonary vascular remodeling, a key process in the asthma pathology. Logistic regression analysis resulted in a diagnostic epigenetic signature of 3-DMPs (involving genes ZNF2698, LOC102723354 and RPL15/NKIRAS1) that allows to efficiently differentiate sequelae cases from CR-RSV patients (AUC = 1.00). Enrichment pathway analysis reveals the role of the cell cycle checkpoint (FDR P-value = 4.71×10-2), DNA damage (FDP-value = 2.53×10-2), and DNA integrity checkpoint (FDR P-value = 2.56×10-2) in differentiating sequelae from CR-RSV patients. Conclusions Epigenetic mechanisms might play a fundamental role in the long-term sequelae after RSV infection, contributing to explain the different phenotypes observed.
Collapse
Affiliation(s)
- Sara Pischedda
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Miriam Cebey-López
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain
| | - Ruth Barral-Arca
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain
| | - Jose Gómez-Rial
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - María-José Curras-Tuala
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Sandra Viz-Lasheras
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Ana B Crujeiras
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto De Investigación Sanitaria De Santiago De Compostela (IDIS), Complejo Hospitalario Universitario De Santiago De Compostela (CHUS/SERGAS), Santiago de Compostela, Spain.,Centro De Investigación Biomédica En Red Fisiopatología De La Obesidad Y Nutrición (Ciberobn), Madrid, Spain
| | - Angel Diaz-Lagares
- Cancer Epigenomics, Epigenomics Unit, Translational Medical Oncology (Oncomet), Instituto De Investigación Sanitaria De Santiago De Compostela (IDIS), Complejo Hospitalario Universitario De Santiago De Compostela (CHUS/SERGAS), Santiago De Compostela, Spain.,Centro De Investigación Biomédica En Red Cancer (CIBERONC), Madrid, Spain
| | | | - Federico Martinón-Torres
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Antonio Salas
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain.,Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | |
Collapse
|
24
|
Liu Z, Ren Y, Weng S, Xu H, Li L, Han X. A New Trend in Cancer Treatment: The Combination of Epigenetics and Immunotherapy. Front Immunol 2022; 13:809761. [PMID: 35140720 PMCID: PMC8818678 DOI: 10.3389/fimmu.2022.809761] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
In recent years, immunotherapy has become a hot spot in the treatment of tumors. As an emerging treatment, it solves many problems in traditional cancer treatment and has now become the main method for cancer treatment. Although immunotherapy is promising, most patients do not respond to treatment or develop resistance. Therefore, in order to achieve a better therapeutic effect, combination therapy has emerged. The combination of immune checkpoint inhibition and epigenetic therapy is one such strategy. In this review, we summarize the current understanding of the key mechanisms of how epigenetic mechanisms affect cancer immune responses and reveal the key role of epigenetic processes in regulating immune cell function and mediating anti-tumor immunity. In addition, we highlight the outlook of combined epigenetic and immune regimens, particularly the combination of immune checkpoint blockade with epigenetic agents, to address the limitations of immunotherapy alone.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Lifeng Li
- Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, China
- Medical School, Huanghe Science and Technology University, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Lifeng Li,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Lifeng Li,
| |
Collapse
|
25
|
Xu CJ, Scheltema NM, Qi C, Vedder R, Klein LBC, Nibbelke EE, van der Ent CK, Bont LJ, Koppelman GH. Infant RSV immunoprophylaxis changes nasal epithelial DNA methylation at 6 years of age. Pediatr Pulmonol 2021; 56:3822-3831. [PMID: 34473906 DOI: 10.1002/ppul.25643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/17/2021] [Accepted: 08/22/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection has been associated with childhood wheeze and asthma, and potential mechanisms include persistent epigenetic effects. METHODS In the randomized, placebo-controlled MAKI trial, 429 preterm infants randomly received RSV immunoprophylaxis with palivizumab or placebo during their first RSV season. Children were followed until age 6 for asthma evaluation. DNA methylation in cells obtained by nasal brushes at age 6 was measured by Illumina MethylationEPIC array. RESULTS RSV immunoprophylaxis in infancy had a significant impact on global methylation patterns in nasal cells at age 6. The first principal component (PC) related to the immunoprophylaxis intervention was enriched for the pathway "detection of chemical stimulus involved in sensory perception of smell" and "T cell differentiation." Subsequent analysis of these PCs indicated an effect of RSV immunoprophylaxis on cell type composition of nasal brushed cells. Three CpG sites, cg18040241, cg08243963, and cg19555973 which are annotated to genes GLB1L2, SC5D, and BPIFB1, were differentially methylated at genome-wide significance, but were not associated with asthma. CONCLUSION The study provides the first proof of concept that RSV immunoprophylaxis during infancy has long-term effects on nasal epigenetic signatures at age 6, relating to host sensory perception, epidermal growth factor receptor signaling, and adaptive immune responses.
Collapse
Affiliation(s)
- Cheng-Jian Xu
- Centre for Individualised Infection Medicine, CiiM, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,Research Group of Bioinformatics and Computational Genomics, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nienke M Scheltema
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cancan Qi
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rolf Vedder
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Laura B C Klein
- Department of Molecular Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Elisabeth E Nibbelke
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelis K van der Ent
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
| | - Louis J Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
26
|
Wang X, Xia H, Liu S, Cao L, You F. Epigenetic regulation in antiviral innate immunity. Eur J Immunol 2021; 51:1641-1651. [PMID: 33964027 DOI: 10.1002/eji.202048975] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/03/2021] [Indexed: 12/24/2022]
Abstract
Emerging life-threatening viruses have posed great challenges to public health. It is now increasingly clear that epigenetics plays a role in shaping host-virus interactions and there is a great need for a more thorough understanding of these intricate interactions through the epigenetic lens, which may represent potential therapeutic opportunities in the clinic. In this review, we highlight the current understanding of the roles of key epigenetic regulators - chromatin remodeling and histone modification - in modulating chromatin openness during host defense against virus. We also discuss how the RNA modification m6A (N6-methyladenosine) affects fundamental aspects of host-virus interactions. We conclude with future directions for uncovering more detailed functions that epigenetic regulation exerts on both host cells and viruses during infection.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Immunology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, P. R. China
| | - Huawei Xia
- Department of Immunology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, P. R. China
| | - Shengde Liu
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Lili Cao
- Department of Immunology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, P. R. China
| | - Fuping You
- Department of Immunology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, P. R. China
| |
Collapse
|
27
|
Hafler D, Sumida T, Dulberg S, Schupp J, Stillwell H, Axisa PP, Comi M, Lincoln M, Unterman A, Kaminski N, Madi A, Kuchroo V. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. RESEARCH SQUARE 2021:rs.3.rs-133494. [PMID: 34127967 PMCID: PMC8202434 DOI: 10.21203/rs.3.rs-133494/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
While inhibition of T cell co-inhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. Type 1 interferon (IFN-I) modulates T cell immunity in viral infection, autoimmunity, and cancer, and may facilitate induction of T cell exhaustion in chronic viral infection. Here we show that IFN-I regulates co-inhibitory receptor expression on human T cells, inducing PD-1/TIM-3/LAG-3 while surprisingly inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses enabled the construction of dynamic transcriptional regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors on human primary T cells revealed unique regulators that control expression of co-inhibitory receptors. We found that the dynamic IFN-I response in vitro closely mirrored T cell features with IFN-I linked acute SARS-CoV-2 infection in human, with high LAG3 and decreased TIGIT expression. Finally, our gene regulatory network identified SP140 as a key regulator for differential LAG3 and TIGIT expression, which were validated at the level of protein expression. The construction of IFN-I regulatory networks with identification of unique transcription factors controlling co-inhibitory receptor expression may provide targets for enhancement of immunotherapy in cancer, infectious diseases, and autoimmunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michela Comi
- Department of Immunobiology, Yale University School of Medicine; Department of Neurology, Yale University School of Medicine
| | | | - Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine
| | | | | | | |
Collapse
|
28
|
Narayanan S, Elesela S, Rasky AJ, Morris SH, Kumar S, Lombard D, Lukacs NW. ER stress protein PERK promotes inappropriate innate immune responses and pathogenesis during RSV infection. J Leukoc Biol 2021; 111:379-389. [PMID: 33866604 DOI: 10.1002/jlb.3a0520-322rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The activation of dendritic cells (DC) during respiratory viral infections is central to directing the immune response and the pathologic outcome. In these studies, the effect of RSV infection on development of ER stress responses and the impact on innate immunity was examined. The upregulation of ER stress was closely associated with the PERK pathway through the upregulation of CHOP in RSV infected DC. The inhibition of PERK corresponded with decreased EIF2a phosphorylation but had no significant effect on Nrf2 in DC, two primary pathways regulated by PERK. Subsequent studies identified that by blocking PERK activity in infected DC an altered ER stress response and innate cytokine profile was observed with the upregulation of IFNβ and IL-12, coincident to the down regulation of IL-1β. When mitochondria respiration was assessed in PERK deficient DC there were increased dysfunctional mitochondria after RSV infection that resulted in reduced oxygen consumption rates (OCR) and ATP production indicating altered cellular metabolism. Use of a CD11c targeted genetic deleted murine model, RSV infection was characterized by reduced inflammation and diminished mucus staining as well as reduced mucus-associated gene gob5 expression. The assessment of the cytokine responses showed decreased IL-13 and IL-17 along with diminished IL-1β in the lungs of PERK deficient infected mice. When PERK-deficient animals were assessed in parallel for lung leukocyte numbers, animals displayed significantly reduced myeloid and activated CD4 and CD8 T cell numbers. Thus, the PERK activation pathway may provide a rational target for altering the severe outcome of an RSV infection through modifying immune responses.
Collapse
Affiliation(s)
- Samanthi Narayanan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Srikanth Elesela
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan H Morris
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Surinder Kumar
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - David Lombard
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Toor SM, Sasidharan Nair V, Saleh R, Taha RZ, Murshed K, Al-Dhaheri M, Khawar M, Ahmed AA, Kurer MA, Abu Nada M, Elkord E. Transcriptome of Tumor-Infiltrating T Cells in Colorectal Cancer Patients Uncovered a Unique Gene Signature in CD4 + T Cells Associated with Poor Disease-Specific Survival. Vaccines (Basel) 2021; 9:vaccines9040334. [PMID: 33916009 PMCID: PMC8065799 DOI: 10.3390/vaccines9040334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is influenced by infiltration of immune cell populations in the tumor microenvironment. While elevated levels of cytotoxic T cells are associated with improved prognosis, limited studies have reported associations between CD4+ T cells and disease outcomes. We recently performed transcriptomic profiling and comparative analyses of sorted CD4+ and CD8+ tumor-infiltrating lymphocytes (TILs) from bulk tumors of CRC patients with varying disease stages. In this study, we compared the transcriptomes of CD4+ with CD8+ TILs. Functional annotation pathway analyses revealed the downregulation of inflammatory response-related genes, while T cell activation and angiogenesis-related genes were upregulated in CD4+ TILs. The top 200 deregulated genes in CD4+ TILs were aligned with the cancer genome atlas (TCGA) CRC dataset to identify a unique gene signature associated with poor prognosis. Moreover, 69 upregulated and 20 downregulated genes showed similar trends of up/downregulation in the TCGA dataset and were used to calculate "poor prognosis score" (ppScore), which was significantly associated with disease-specific survival. High ppScore patients showed lower expression of Treg-, Th1-, and Th17-related genes, and higher expression of Th2-related genes. Our data highlight the significance of T cells within the TME and identify a unique candidate prognostic gene signature for CD4+ TILs in CRC patients.
Collapse
Affiliation(s)
- Salman M. Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Rowaida Z. Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Khaled Murshed
- Department of Pathology, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar;
| | - Mahmood Al-Dhaheri
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mahwish Khawar
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Ayman A. Ahmed
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mohamed A. Kurer
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mohamed Abu Nada
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester M5 4WT, UK
- Correspondence: ; Tel.: +44-161-295-5736
| |
Collapse
|
30
|
Acevedo N, Alashkar Alhamwe B, Caraballo L, Ding M, Ferrante A, Garn H, Garssen J, Hii CS, Irvine J, Llinás-Caballero K, López JF, Miethe S, Perveen K, Pogge von Strandmann E, Sokolowska M, Potaczek DP, van Esch BCAM. Perinatal and Early-Life Nutrition, Epigenetics, and Allergy. Nutrients 2021; 13:724. [PMID: 33668787 PMCID: PMC7996340 DOI: 10.3390/nu13030724] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Epidemiological studies have shown a dramatic increase in the incidence and the prevalence of allergic diseases over the last several decades. Environmental triggers including risk factors (e.g., pollution), the loss of rural living conditions (e.g., farming conditions), and nutritional status (e.g., maternal, breastfeeding) are considered major contributors to this increase. The influences of these environmental factors are thought to be mediated by epigenetic mechanisms which are heritable, reversible, and biologically relevant biochemical modifications of the chromatin carrying the genetic information without changing the nucleotide sequence of the genome. An important feature characterizing epigenetically-mediated processes is the existence of a time frame where the induced effects are the strongest and therefore most crucial. This period between conception, pregnancy, and the first years of life (e.g., first 1000 days) is considered the optimal time for environmental factors, such as nutrition, to exert their beneficial epigenetic effects. In the current review, we discussed the impact of the exposure to bacteria, viruses, parasites, fungal components, microbiome metabolites, and specific nutritional components (e.g., polyunsaturated fatty acids (PUFA), vitamins, plant- and animal-derived microRNAs, breast milk) on the epigenetic patterns related to allergic manifestations. We gave insight into the epigenetic signature of bioactive milk components and the effects of specific nutrition on neonatal T cell development. Several lines of evidence suggest that atypical metabolic reprogramming induced by extrinsic factors such as allergens, viruses, pollutants, diet, or microbiome might drive cellular metabolic dysfunctions and defective immune responses in allergic disease. Therefore, we described the current knowledge on the relationship between immunometabolism and allergy mediated by epigenetic mechanisms. The knowledge as presented will give insight into epigenetic changes and the potential of maternal and post-natal nutrition on the development of allergic disease.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Bilal Alashkar Alhamwe
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - James Irvine
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kevin Llinás-Caballero
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Juan Felipe López
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
31
|
Anderson J, Do LAH, Wurzel D, Quan Toh Z, Mulholland K, Pellicci DG, Licciardi PV. Severe respiratory syncytial virus disease in preterm infants: a case of innate immaturity. Thorax 2021; 76:942-950. [PMID: 33574121 DOI: 10.1136/thoraxjnl-2020-216291] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/27/2021] [Indexed: 11/03/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common viral pathogen associated with acute lower respiratory tract infection (LRTI) in children under 5 years of age. Severe RSV disease is associated with the development of chronic respiratory complications such as recurrent wheezing and asthma. A common risk factor for developing severe RSV disease is premature gestation and this is largely due to an immature innate immune system. This increases susceptibility to RSV since the innate immune system is less able to protect against pathogens at a time when adaptive immunity has not fully developed. This review focuses on comparing different aspects of innate immunity between preterm and term infants to better understand why preterm infants are more susceptible to severe RSV disease. Identifying early life innate immune biomarkers associated with the development of severe RSV disease, and understanding how these compare between preterm and term infants, remains a critically important question that would aid the development of interventions to reduce the burden of disease in this vulnerable population.
Collapse
Affiliation(s)
- Jeremy Anderson
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Lien Anh Ha Do
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Danielle Wurzel
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia.,The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Zheng Quan Toh
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Kim Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia.,Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Daniel G Pellicci
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia .,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Shibata T, Makino A, Ogata R, Nakamura S, Ito T, Nagata K, Terauchi Y, Oishi T, Fujieda M, Takahashi Y, Ato M. Respiratory syncytial virus infection exacerbates pneumococcal pneumonia via Gas6/Axl-mediated macrophage polarization. J Clin Invest 2021; 130:3021-3037. [PMID: 32364537 DOI: 10.1172/jci125505] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with respiratory syncytial virus (RSV) infection exhibit enhanced susceptibility to subsequent pneumococcal infections. However, the underlying mechanisms involved in this increased susceptibility remain unclear. Here, we identified potentially novel cellular and molecular cascades triggered by RSV infection to exacerbate secondary pneumococcal pneumonia. RSV infection stimulated the local production of growth arrest-specific 6 (Gas6). The Gas6 receptor Axl was crucial for attenuating pneumococcal immunity in that the Gas6/Axl blockade fully restored antibacterial immunity. Mechanistically, Gas6/Axl interaction regulated the conversion of alveolar macrophages from an antibacterial phenotype to an M2-like phenotype that did not exhibit antibacterial activity, and the attenuation of caspase-1 activation and IL-18 production in response to pneumococcal infection. The attenuated IL-18 production failed to drive both NK cell-mediated IFN-γ production and local NO and TNF-α production, which impair the control of bacterial infection. Hence, the RSV-mediated Gas6/Axl activity attenuates the macrophage-mediated protection against pneumococcal infection. The Gas6/Axl axis could be a potentially novel therapeutic target for RSV-associated secondary bacterial infection.
Collapse
Affiliation(s)
- Takehiko Shibata
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Airi Makino
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, Japan
| | - Ruiko Ogata
- Department of Immunology, Nara Medical University, Nara, Japan
| | - Shigeki Nakamura
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Nara, Japan
| | - Kisaburo Nagata
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, Japan
| | - Yoshihiko Terauchi
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Pediatrics, National Hospital Organization Kochi Hospital, Kochi, Japan
| | - Taku Oishi
- Department of Pediatrics, National Hospital Organization Kochi Hospital, Kochi, Japan
| | - Mikiya Fujieda
- Department of Pediatrics, National Hospital Organization Kochi Hospital, Kochi, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Mycobacteriology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
33
|
Malinczak CA, Parolia A, Fonseca W, Morris S, Rasky AJ, Bawa P, Zhang Y, Mire MM, Ziegler SF, Ptaschinski C, Chinnaiyan AM, Lukacs NW. TSLP-Driven Chromatin Remodeling and Trained Systemic Immunity after Neonatal Respiratory Viral Infection. THE JOURNAL OF IMMUNOLOGY 2021; 206:1315-1328. [PMID: 33514510 DOI: 10.4049/jimmunol.2001205] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Our studies have previously shown a role for persistent TSLP production in the lungs of mice after early-life respiratory syncytial virus (RSV) infection that leads to an altered immune phenotype, including accumulation of "inflammatory" dendritic cells (DC). This study investigates the role of TSLP driving systemic trained immunity in DC in early-life RSV-infected mice. Bone marrow-derived DCs (BMDC) from early-life RSV-infected mice at 4 wk postinfection showed enhanced expression of costimulatory molecules and cytokines, including Tslp, that regulate immune cell function. The adoptive transfer of BMDC grown from early-life RSV-infected mice was sufficient to exacerbate allergic disease development. The addition of recombinant TSLP during differentiation of BMDC from naive mice induced a similar altered phenotype as BMDC grown from early-life RSV-infected mice, suggesting a role for TSLP in the phenotypic changes. To assess the role of TSLP in these changes, global transcriptomic characterization of TSLPR-/- BMDC infected with RSV was performed and showed a higher upregulation of type 1 IFN genes and concomitant downregulation of inflammatory genes. Assay for transposase-accessible chromatin using sequencing analysis demonstrated that TSLPR-/- BMDC had a parallel gain in physical chromatin accessibility near type 1 genes and loss in accessibility near genes related to RSV pathology, with IFN regulatory factor 4 (IRF4) and STAT3 predicted as top transcription factors binding within differentially accessible regions in wild-type. Importantly, these studies show that in the absence of TSLP signaling, BMDC are able to mount an appropriate type 1 IFN-associated antiviral response to RSV. In summary, RSV-induced TSLP alters chromatin structure in DC to drive trained innate immunity and activates pathogenic gene programs in mice.
Collapse
Affiliation(s)
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Susan Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Pushpinder Bawa
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed M Mire
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Steven F Ziegler
- Department of Immunology, Benaroya Research Institute, Seattle, WA 98101
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109; and.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
34
|
Ramos-Lopez O, Milagro FI, Riezu-Boj JI, Martinez JA. Epigenetic signatures underlying inflammation: an interplay of nutrition, physical activity, metabolic diseases, and environmental factors for personalized nutrition. Inflamm Res 2021; 70:29-49. [PMID: 33231704 PMCID: PMC7684853 DOI: 10.1007/s00011-020-01425-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
AIM AND OBJECTIVE Emerging translational evidence suggests that epigenetic alterations (DNA methylation, miRNA expression, and histone modifications) occur after external stimuli and may contribute to exacerbated inflammation and the risk of suffering several diseases including diabetes, cardiovascular diseases, cancer, and neurological disorders. This review summarizes the current knowledge about the harmful effects of high-fat/high-sugar diets, micronutrient deficiencies (folate, manganese, and carotenoids), obesity and associated complications, bacterial/viral infections, smoking, excessive alcohol consumption, sleep deprivation, chronic stress, air pollution, and chemical exposure on inflammation through epigenetic mechanisms. Additionally, the epigenetic phenomena underlying the anti-inflammatory potential of caloric restriction, n-3 PUFA, Mediterranean diet, vitamin D, zinc, polyphenols (i.e., resveratrol, gallic acid, epicatechin, luteolin, curcumin), and the role of systematic exercise are discussed. METHODS Original and review articles encompassing epigenetics and inflammation were screened from major databases (including PubMed, Medline, Science Direct, Scopus, etc.) and analyzed for the writing of the review paper. CONCLUSION Although caution should be exercised, research on epigenetic mechanisms is contributing to understand pathological processes involving inflammatory responses, the prediction of disease risk based on the epigenotype, as well as the putative design of therapeutic interventions targeting the epigenome.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana, Baja California, Mexico
| | - Fermin I Milagro
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.
| | - Jose I Riezu-Boj
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - J Alfredo Martinez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Madrid, Spain
| |
Collapse
|
35
|
Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020. [DOI: 10.3390/vaccines8040783
expr 839529059 + 832255227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
|
36
|
Lukacs NW, Malinczak CA. Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020; 8:783. [PMID: 33371275 PMCID: PMC7766447 DOI: 10.3390/vaccines8040783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
Affiliation(s)
- Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
37
|
Xu Q, Tang Y, Huang G. Innate immune responses in RNA viral infection. Front Med 2020; 15:333-346. [PMID: 33263837 PMCID: PMC7862985 DOI: 10.1007/s11684-020-0776-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 03/14/2020] [Indexed: 12/17/2022]
Abstract
RNA viruses cause a multitude of human diseases, including several pandemic events in the past century. Upon viral invasion, the innate immune system responds rapidly and plays a key role in activating the adaptive immune system. In the innate immune system, the interactions between pathogen-associated molecular patterns and host pattern recognition receptors activate multiple signaling pathways in immune cells and induce the production of pro-inflammatory cytokines and interferons to elicit antiviral responses. Macrophages, dendritic cells, and natural killer cells are the principal innate immune components that exert antiviral activities. In this review, the current understanding of innate immunity contributing to the restriction of RNA viral infections was briefly summarized. Besides the main role of immune cells in combating viral infection, the intercellular transfer of pathogen and host-derived materials and their epigenetic and metabolic interactions associated with innate immunity was discussed. This knowledge provides an enhanced understanding of the innate immune response to RNA viral infections in general and aids in the preparation for the existing and next emerging viral infections.
Collapse
Affiliation(s)
- Qian Xu
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuting Tang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
38
|
Sumida TS, Dulberg S, Schupp J, Stillwell HA, Axisa PP, Comi M, Lincoln M, Unterman A, Kaminski N, Madi A, Kuchroo VK, Hafler DA. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.10.30.362947. [PMID: 33140047 PMCID: PMC7605554 DOI: 10.1101/2020.10.30.362947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
While inhibition of T cell co-inhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. Type 1 interferon (IFN-I) modulates T cell immunity in viral infection, autoimmunity, and cancer, and may facilitate induction of T cell exhaustion in chronic viral infection 1,2 . Here we show that IFN-I regulates co-inhibitory receptors expression on human T cells, inducing PD-1/TIM-3/LAG-3 while surprisingly inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses enabled the construction of dynamic transcriptional regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors on human primary T cells revealed both canonical and non-canonical IFN-I transcriptional regulators, and identified unique regulators that control expression of co-inhibitory receptors. To provide direct in vivo evidence for the role of IFN-I on co-inhibitory receptors, we then performed single cell RNA-sequencing in subjects infected with SARS-CoV-2, where viral load was strongly associated with T cell IFN-I signatures. We found that the dynamic IFN-I response in vitro closely mirrored T cell features with acute IFN-I linked viral infection, with high LAG3 and decreased TIGIT expression. Finally, our gene regulatory network identified SP140 as a key regulator for differential LAG3 and TIGIT expression. The construction of co-inhibitory regulatory networks induced by IFN-I with identification of unique transcription factors controlling their expression may provide targets for enhancement of immunotherapy in cancer, infectious diseases, and autoimmunity.
Collapse
Affiliation(s)
- Tomokazu S. Sumida
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Shai Dulberg
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonas Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Helen A. Stillwell
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Pierre-Paul Axisa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Michela Comi
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Matthew Lincoln
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Asaf Madi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
39
|
Terada-Ikeda C, Kitabatake M, Hiraku A, Kato K, Yasui S, Imakita N, Ouji-Sageshima N, Iwabuchi N, Hamada K, Ito T. Maternal supplementation with Bifidobacterium breve M-16V prevents their offspring from allergic airway inflammation accelerated by the prenatal exposure to an air pollutant aerosol. PLoS One 2020; 15:e0238923. [PMID: 32915886 PMCID: PMC7485856 DOI: 10.1371/journal.pone.0238923] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Bifidobacterium breve M-16V is a probiotic bacterial strain with efficacy in infants achieved by suppressing T-helper type (Th) 2 immune responses and modulating the systemic Th1/Th2 balance. Exposure to air pollution during pregnancy increases asthma susceptibility in offspring. The aim of this study was to investigate the effects of the maternal intake of B. breve M-16V on susceptibility to asthma accelerated by prenatal exposure to air pollution. The intake of B. breve M-16V in residual oil fly ash (ROFA)-exposed pregnant mice resulted in fewer eosinophils in the bronchoalveolar lavage fluid of neonatal mice and reduced allergic lung inflammation. The expressions of Th2 cytokines including IL-5 and IL-13 were decreased in neonatal mice from ROFA-exposed mothers fed B. breve M-16V. The analysis of fecal microbiota from neonatal mice revealed that the intake of B. breve M-16V by mothers changed the composition of fecal microbiota in neonatal mice, which resulted in a decreased population of Firmicutes. Moreover, several bacterial strains of fecal microbiota from neonatal mice had a strong correlation with Th2 cytokines and histological score. These results suggest that the maternal intake of M-16V might have beneficial effects in neonates by preventing and/or alleviating allergic reactions accelerated by prenatal exposure to air pollution.
Collapse
Affiliation(s)
| | | | - Akari Hiraku
- R&D Division, Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Kumiko Kato
- R&D Division, Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Satsuki Yasui
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan
| | - Natsuko Imakita
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan
| | | | - Noriyuki Iwabuchi
- R&D Division, Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Kaoru Hamada
- Department of Clinical and Investigative Medicine, Faculty of Nursing, Nara Medical University, Kashihara, Nara, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan
- * E-mail:
| |
Collapse
|
40
|
Decoding Susceptibility to Respiratory Viral Infections and Asthma Inception in Children. Int J Mol Sci 2020; 21:ijms21176372. [PMID: 32887352 PMCID: PMC7503410 DOI: 10.3390/ijms21176372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023] Open
Abstract
Human Respiratory Syncytial Virus and Human Rhinovirus are the most frequent cause of respiratory tract infections in infants and children and are major triggers of acute viral bronchiolitis, wheezing and asthma exacerbations. Here, we will discuss the application of the powerful tools of systems biology to decode the molecular mechanisms that determine risk for infection and subsequent asthma. An important conceptual advance is the understanding that the innate immune system is governed by a Bow-tie architecture, where diverse input signals converge onto a few core pathways (e.g., IRF7), which in turn generate diverse outputs that orchestrate effector and regulatory functions. Molecular profiling studies in children with severe exacerbations of asthma/wheeze have identified two major immunological phenotypes. The IRF7hi phenotype is characterised by robust upregulation of antiviral response networks, and the IRF7lo phenotype is characterised by upregulation of markers of TGFβ signalling and type 2 inflammation. Similar phenotypes have been identified in infants and children with severe viral bronchiolitis. Notably, genome-wide association studies supported by experimental validation have identified key pathways that increase susceptibility to HRV infection (ORMDL3 and CHDR3) and modulate TGFβ signalling (GSDMB, TGFBR1, and SMAD3). Moreover, functional deficiencies in the activation of type I and III interferon responses are already evident at birth in children at risk of developing febrile lower respiratory tract infections and persistent asthma/wheeze, suggesting that the trajectory to asthma begins at birth or in utero. Finally, exposure to microbes and their products reprograms innate immunity and provides protection from the development of allergies and asthma in children, and therefore microbial products are logical candidates for the primary prevention of asthma.
Collapse
|
41
|
Early-Life Respiratory Syncytial Virus Infection, Trained Immunity and Subsequent Pulmonary Diseases. Viruses 2020; 12:v12050505. [PMID: 32375305 PMCID: PMC7290378 DOI: 10.3390/v12050505] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) is often the first clinically relevant pathogen encountered in life, with nearly all children infected by two years of age. Many studies have also linked early-life severe respiratory viral infection with more pathogenic immune responses later in life that lead to pulmonary diseases like childhood asthma. This phenomenon is thought to occur through long-term immune system alterations following early-life respiratory viral infection and may include local responses such as unresolved inflammation and/or direct structural or developmental modifications within the lung. Furthermore, systemic responses that could impact the bone marrow progenitors may be a significant cause of long-term alterations, through inflammatory mediators and shifts in metabolic profiles. Among these alterations may be changes in transcriptional and epigenetic programs that drive persistent modifications throughout life, leaving the immune system poised toward pathogenic responses upon secondary insult. This review will focus on early-life severe RSV infection and long-term alterations. Understanding these mechanisms will not only lead to better treatment options to limit initial RSV infection severity but also protect against the development of childhood asthma linked to severe respiratory viral infections.
Collapse
|
42
|
Parmar N, Chandrakar P, Kar S. Leishmania donovani Subverts Host Immune Response by Epigenetic Reprogramming of Macrophage M(Lipopolysaccharides + IFN-γ)/M(IL-10) Polarization. THE JOURNAL OF IMMUNOLOGY 2020; 204:2762-2778. [DOI: 10.4049/jimmunol.1900251] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/16/2020] [Indexed: 01/19/2023]
|
43
|
Cao J, Yan Q. Cancer Epigenetics, Tumor Immunity, and Immunotherapy. Trends Cancer 2020; 6:580-592. [PMID: 32610068 DOI: 10.1016/j.trecan.2020.02.003] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Epigenetic mechanisms, including DNA methylation, histone post-translational modifications, and chromatin structure regulation, are critical for the interactions between tumor and immune cells. Emerging evidence shows that tumors commonly hijack various epigenetic mechanisms to escape immune restriction. As a result, the pharmaceutical modulation of epigenetic regulators, including 'writers', 'readers', 'erasers', and 'remodelers', is able to normalize the impaired immunosurveillance and/or trigger antitumor immune responses. Thus, epigenetic targeting agents are attractive immunomodulatory drugs and will have major impacts on immuno-oncology. Here, we discuss epigenetic regulators of the cancer-immunity cycle and current advances in developing epigenetic therapies to boost anticancer immune responses, either alone or in combination with current immunotherapies.
Collapse
Affiliation(s)
- Jian Cao
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Qin Yan
- Department of Pathology, Yale Cancer Center, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
44
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Allergic diseases are prototypic examples for gene × environment-wide interactions. This review considers the current evidence for genetic and epigenetic mechanisms in allergic diseases and highlights barriers and facilitators for the implementation of these novel tools both for research and clinical practice. RECENT FINDINGS The value of whole-genome sequencing studies and the use of polygenic risk score analysis in homogeneous well characterized populations are currently being tested. Epigenetic mechanisms are known to play a crucial role in the pathogenesis of allergic disorders, especially through mediating the effects of the environmental factors, well recognized risk modifiers. There is emerging evidence for the immune-modulatory role of probiotics through epigenetic changes. Direct or indirect targeting of epigenetic mechanisms affect expression of the genes favouring the development of allergic diseases and can improve tissue biology. The ability to specifically edit the epigenome, especially using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 technology, holds the promise of enhancing understanding of how epigenetic modifications function and enabling manipulation of cell phenotype for research or therapeutic purposes. SUMMARY Additional research in the role of genetic and epigenetic mechanisms in relation to allergic diseases' endotypes is needed. An international project characterizing the human epigenome in relation to allergic diseases is warranted.
Collapse
|
46
|
Nehme Z, Pasquereau S, Herbein G. Targeting histone epigenetics to control viral infections. HISTONE MODIFICATIONS IN THERAPY 2020. [PMCID: PMC7453269 DOI: 10.1016/b978-0-12-816422-8.00011-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past decades, many studies have significantly broadened our understanding of complex virus-host interactions to control chromatin structure and dynamics.1, 2 However, the role and impact of such modifications during viral infections is not fully revealed. Indeed, this type of regulation is bidirectional between the virus and the host. While viral replication and gene expression are significantly impacted by histone modifications on the viral chromatin,3 studies have shown that some viral pathogens dynamically manipulate cellular epigenetic factors to enhance their own survival and pathogenesis, as well as escape the immune system defense lines.4 In this dynamic, histone posttranslational modifications (PTMs) appear to play fundamental roles in the regulation of chromatin structure and recruitment of other factors.5 Genuinely, those PTMs play a vital role in lytic infection, latency reinforcement, or, conversely, viral reactivation.6 In this chapter, we will examine and review the involvement of histone modifications as well as their potential manipulation to control infections during various viral life cycle stages, highlighting their prospective implications in the clinical management of human immunodeficiency virus (HIV), herpes simplex virus (HSV), human cytomegalovirus (HCMV), hepatitis B and C viruses (HBV and HCV, respectively), Epstein–Barr virus (EBV), and other viral diseases. Targeting histone modifications is critical in setting the treatment of chronic viral infections with both lytic and latent stages (HIV, HCMV, HSV, RSV), virus-induced cancers (HBV, HCV, EBV, KSHV, HPV), and epidemic/emerging viruses (e.g. influenza virus, arboviruses).
Collapse
|
47
|
Malinczak CA, Rasky AJ, Fonseca W, Schaller MA, Allen RM, Ptaschinski C, Morris S, Lukacs NW. Upregulation of H3K27 Demethylase KDM6 During Respiratory Syncytial Virus Infection Enhances Proinflammatory Responses and Immunopathology. THE JOURNAL OF IMMUNOLOGY 2019; 204:159-168. [PMID: 31748348 DOI: 10.4049/jimmunol.1900741] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022]
Abstract
Severe disease following respiratory syncytial virus (RSV) infection has been linked to enhanced proinflammatory cytokine production that promotes a Th2-type immune environment. Epigenetic regulation in immune cells following viral infection plays a role in the inflammatory response and may result from upregulation of key epigenetic modifiers. In this study, we show that RSV-infected bone marrow-derived dendritic cells (BMDC) as well as pulmonary dendritic cells (DC) from RSV-infected mice upregulated the expression of Kdm6b/Jmjd3 and Kdm6a/Utx, H3K27 demethylases. KDM6-specific chemical inhibition (GSK J4) in BMDC led to decreased production of chemokines and cytokines associated with the inflammatory response during RSV infection (i.e., CCL-2, CCL-3, CCL-5, IL-6) as well as decreased MHC class II and costimulatory marker (CD80/86) expression. RSV-infected BMDC treated with GSK J4 altered coactivation of T cell cytokine production to RSV as well as a primary OVA response. Airway sensitization of naive mice with RSV-infected BMDCs exacerbate a live challenge with RSV infection but was inhibited when BMDCs were treated with GSK J4 prior to sensitization. Finally, in vivo treatment with the KDM6 inhibitor, GSK J4, during RSV infection reduced inflammatory DC in the lungs along with IL-13 levels and overall inflammation. These results suggest that KDM6 expression in DC enhances proinflammatory innate cytokine production to promote an altered Th2 immune response following RSV infection that leads to more severe immunopathology.
Collapse
Affiliation(s)
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Matthew A Schaller
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL 32610; and
| | - Ronald M Allen
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | | | - Susan Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109; .,Mary H. Weiser Food and Allergy Center, Ann Arbor, MI 48109
| |
Collapse
|
48
|
Qi C, Xu CJ, Koppelman GH. The role of epigenetics in the development of childhood asthma. Expert Rev Clin Immunol 2019; 15:1287-1302. [PMID: 31674254 DOI: 10.1080/1744666x.2020.1686977] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The development of childhood asthma is caused by a combination of genetic factors and environmental exposures. Epigenetics describes mechanisms of (heritable) regulation of gene expression that occur without changes in DNA sequence. Epigenetics is strongly related to aging, is cell-type specific, and includes DNA methylation, noncoding RNAs, and histone modifications.Areas covered: This review summarizes recent epigenetic studies of childhood asthma in humans, which mostly involve studies of DNA methylation published in the recent five years. Environmental exposures, in particular cigarette smoking, have significant impact on epigenetic changes, but few of these epigenetic signals are also associated with asthma. Several asthma-associated genetic variants relate to DNA methylation. Epigenetic signals can be better understood by studying their correlation with gene expression, which revealed higher presence and activation of blood eosinophils in asthma. Strong associations of nasal methylation signatures and atopic asthma were identified, which were replicable across different populations.Expert commentary: Epigenetic markers have been strongly associated with asthma, and might serve as biomarker of asthma. The causal and longitudinal relationships between epigenetics and disease, and between environmental exposures and epigenetic changes need to be further investigated. Efforts should be made to understand cell-type-specific epigenetic mechanisms in asthma.
Collapse
Affiliation(s)
- Cancan Qi
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cheng-Jian Xu
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Gastroenterology, Hepatology and Endocrinology, CiiM, Centre for individualised infection medicine, A joint venture between Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Gerard H Koppelman
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
49
|
McAllister CS, Ansaldi D, Growcott EJ, Zhong Y, Quackenbush D, Wolff KC, Chen Z, Tanaseichuk O, Lelais G, Barnes SW, Federe GC, Luna F, Walker JR, Zhou Y, Kuhen KL. Dexamethasone inhibits respiratory syncytial virus-driven mucus production while increasing viral replication without altering antiviral interferon signaling. Virology 2019; 540:195-206. [PMID: 31929001 DOI: 10.1016/j.virol.2019.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/23/2019] [Accepted: 10/18/2019] [Indexed: 01/03/2023]
Abstract
Respiratory syncytial virus (RSV) infection can cause mucus overproduction and bronchiolitis in infants leading to severe disease and hospitalization. As a therapeutic strategy, immune modulatory agents may help prevent RSV-driven immune responses that cause severe airway disease. We developed a high throughput screen to identify compounds that reduced RSV-driven mucin 5AC (Muc5AC) expression and identified dexamethasone. Despite leading to a pronounced reduction in RSV-driven Muc5AC, dexamethasone increased RSV infection in vitro and delayed viral clearance in mice. This correlated with reduced expression of a subset of immune response genes and reduced lymphocyte infiltration in vivo. Interestingly, dexamethasone increased RSV infection levels without altering antiviral interferon signaling. In summary, the immunosuppressive activities of dexamethasone had favorable inhibitory effects on RSV-driven mucus production yet prevented immune defense activities that limit RSV infection in vitro and in vivo. These findings offer an explanation for the lack of efficacy of glucocorticoids in RSV-infected patients.
Collapse
Affiliation(s)
| | - Dan Ansaldi
- Novartis Institute for BioMedical Research, Emeryville, CA, USA
| | | | - Yang Zhong
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Doug Quackenbush
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Karen C Wolff
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Zhong Chen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Olga Tanaseichuk
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Gerald Lelais
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - S Whitney Barnes
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Glenn C Federe
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Fabio Luna
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - John R Walker
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Yingyao Zhou
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Kelli L Kuhen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| |
Collapse
|
50
|
Boukhaled GM, Corrado M, Guak H, Krawczyk CM. Chromatin Architecture as an Essential Determinant of Dendritic Cell Function. Front Immunol 2019; 10:1119. [PMID: 31214161 PMCID: PMC6557980 DOI: 10.3389/fimmu.2019.01119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
Epigenetics has widespread implications in a variety of cellular processes ranging from cell identity and specification, to cellular adaptation to environmental stimuli. While typically associated with heritable changes in gene expression, epigenetic mechanisms are now appreciated to regulate dynamic changes in gene expression—even in post-mitotic cells. Cells of the innate immune system, including dendritic cells (DC), rapidly integrate signals from their microenvironment and respond accordingly, undergoing massive changes in transcriptional programming. This dynamic transcriptional reprogramming relies on epigenetic changes mediated by numerous enzymes and their substrates. This review highlights our current understanding of epigenetic regulation of DC function. Epigenetic mechanisms contribute to the maintenance of the steady state and are important for precise responses to proinflammatory stimuli. Interdependence between epigenetic modifications and the delicate balance of metabolites present another layer of complexity. In addition, dynamic regulation of the expression of proteins that modify chromatin architecture in DCs significantly impacts DC function. Environmental factors, including inflammation, aging, chemicals, nutrients, and lipid mediators, are increasingly appreciated to affect the epigenome in DCs, and, in doing so, regulate host immunity. Our understanding of how epigenetic mechanisms regulate DC function is in its infancy, and it must be expanded in order to discern the mechanisms underlying the balance between health and disease states.
Collapse
Affiliation(s)
- Giselle M Boukhaled
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Mario Corrado
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Hannah Guak
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Connie M Krawczyk
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada.,Center for Cancer and Cell Biology, Program in Metabolic and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, United States
| |
Collapse
|