1
|
Serradas ML, Ding Y, Martorell PV, Kulińska I, Castro-Gomez S. Therapeutic Targets in Innate Immunity to Tackle Alzheimer's Disease. Cells 2024; 13:1426. [PMID: 39272998 PMCID: PMC11394242 DOI: 10.3390/cells13171426] [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: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
There is an urgent need for effective disease-modifying therapeutic interventions for Alzheimer's disease (AD)-the most prevalent cause of dementia with a profound socioeconomic burden. Most clinical trials targeting the classical hallmarks of this disease-β-amyloid plaques and neurofibrillary tangles-failed, showed discrete clinical effects, or were accompanied by concerning side effects. There has been an ongoing search for novel therapeutic targets. Neuroinflammation, now widely recognized as a hallmark of all neurodegenerative diseases, has been proven to be a major contributor to AD pathology. Here, we summarize the role of neuroinflammation in the pathogenesis and progression of AD and discuss potential targets such as microglia, TREM2, the complement system, inflammasomes, and cytosolic DNA sensors. We also present an overview of ongoing studies targeting specific innate immune system components, highlighting the progress in this field of drug research while bringing attention to the delicate nature of innate immune modulations in AD.
Collapse
Affiliation(s)
- Maria L Serradas
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Yingying Ding
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Paula V Martorell
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Ida Kulińska
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Sergio Castro-Gomez
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
2
|
Yang S, Guo R, Meng X, Zhang Y. AIM2 participates in house dust mite (HDM)-induced epithelial dysfunctions and ovalbumin (OVA)-induced allergic asthma in infant mice. J Asthma 2024; 61:479-490. [PMID: 38078661 DOI: 10.1080/02770903.2023.2289157] [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: 07/28/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 01/13/2024]
Abstract
Objective: Allergen sensitization and high rates of concomitant allergic diseases are characteristic of severe pediatric asthma. The present study was aimed to explore the mechanism of allergic asthma via bioinformatics and experiment investigation. Methods: The GSE27011 dataset contained the expression profiles of normal and pediatric asthma white blood cells was downloaded for analyzing the different expression genes and function enrichment. The allergic asthma model in infant mice was established by ovalbumin (OVA) stimulation. The cellular model was established by house dust mite (HDM)-stimulation in human bronchial epithelial cells. The absent in melanoma 2 (AIM2) knockdown was achieved by intranasal lentivirus injection or cell infection. The bronchoalveolar lavage fluid (BALF) was collected for cell counting and ELISA assessment of cytokines. Lung tissues were collected for HE staining and immunohistochemical (IHC) staining. Real-time PCR and immunoblotting were used for the determination of key gene expressions in mouse and cell models. Results: upregulation of AIM2 gene expression was observed in pediatric asthma patients based on GSE27011 and OVA-induced infant mouse allergic asthma model. AIM2 knockdown ameliorated OVA caused elevation in airway hyper-responsiveness (AHR), elevation in cell quantities (eosinophils, neutrophils, lymphocytes), and levels of cytokines (IL-4, IL-13, TNF-α, and OVA-specific IgE) in BALF. Moreover, AIM2 knockdown relieved OVA-caused histopathological alterations in mouse lungs, up-regulation of AIM2 levels, and NOD1 and receptor-interacting protein 2 (RIP2) protein levels, as well as p65 phosphorylation. In the cell model, AIM2 knockdown partially ameliorated HDM-induced epithelial dysfunctions by promoting cell viability, down-regulating inflammatory cytokines levels, and decreasing the protein levels of AIM2, NOD1, RIP2, and phosphorylated p65. Conclusion: AIM2 participates in HDM-induced epithelial dysfunctions and OVA-induced allergic asthma progression. AIM2 could be a promising target for pediatric allergic asthma treatment regimens, which warrants further in vivo investigations.
Collapse
Affiliation(s)
- Shengzhi Yang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Ru Guo
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Xianmei Meng
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Yunhong Zhang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| |
Collapse
|
3
|
Chang X, Wang B, Zhao Y, Deng B, Liu P, Wang Y. The role of IFI16 in regulating PANoptosis and implication in heart diseases. Cell Death Discov 2024; 10:204. [PMID: 38693141 PMCID: PMC11063201 DOI: 10.1038/s41420-024-01978-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Interferon Gamma Inducible Protein 16 (IFI16) belongs to the HIN-200 protein family and is pivotal in immunological responses. Serving as a DNA sensor, IFI16 identifies viral and aberrant DNA, triggering immune and inflammatory responses. It is implicated in diverse cellular death mechanisms, such as pyroptosis, apoptosis, and necroptosis. Notably, these processes are integral to the emergent concept of PANoptosis, which encompasses cellular demise and inflammatory pathways. Current research implies a significant regulatory role for IFI16 in PANoptosis, particularly regarding cardiac pathologies. This review delves into the complex interplay between IFI16 and PANoptosis in heart diseases, including atherosclerosis, myocardial infarction, heart failure, and diabetic cardiomyopathy. It synthesizes evidence of IFI16's impact on PANoptosis, with the intention of providing novel insights for therapeutic strategies targeting heart diseases.
Collapse
Affiliation(s)
- Xindi Chang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bei Wang
- Department of Emergency, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Yingli Zhao
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bing Deng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| |
Collapse
|
4
|
Chiarini A, Armato U, Gui L, Dal Prà I. "Other Than NLRP3" Inflammasomes: Multiple Roles in Brain Disease. Neuroscientist 2024; 30:23-48. [PMID: 35815856 DOI: 10.1177/10738584221106114] [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] [Indexed: 11/15/2022]
Abstract
Human neuroinflammatory and neurodegenerative diseases, whose prevalence keeps rising, are still unsolved pathobiological/therapeutical problems. Among others, recent etiology hypotheses stressed as their main driver a chronic neuroinflammation, which is mediated by innate immunity-related protein oligomers: the inflammasomes. A panoply of exogenous and/or endogenous harmful agents activates inflammasomes' assembly, signaling, and IL-1β/IL-18 production and neural cells' pyroptotic death. The underlying concept is that inflammasomes' chronic activation advances neurodegeneration while their short-lasting operation restores tissue homeostasis. Hence, from a therapeutic standpoint, it is crucial to understand inflammasomes' regulatory mechanisms. About this, a deluge of recent studies focused on the NLRP3 inflammasome with suggestions that its pharmacologic block would hinder neurodegeneration. Yet hitherto no evidence proves this view. Moreover, known inflammasomes are numerous, and the mechanisms regulating their expression and function may vary with the involved animal species and strains, as well as organs and cells, and the harmful factors triggered as a result. Therefore, while presently leaving out some little-studied inflammasomes, this review focuses on the "other than NLRP3" inflammasomes that participate in neuroinflammation's complex mechanisms: NLRP1, NLRP2, NLRC4, and AIM2. Although human-specific data about them are relatively scant, we stress that only a holistic view including several human brain inflammasomes and other potential pathogenetic drivers will lead to successful therapies for neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Li Gui
- Department of Neurology, Southwest Hospital, Chongqing, China
| | - Ilaria Dal Prà
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
5
|
Hou J, Zheng Y, Gao C. Regulation of cellular senescence by innate immunity. BIOPHYSICS REPORTS 2023; 9:338-351. [PMID: 38524701 PMCID: PMC10960571 DOI: 10.52601/bpr.2023.230032] [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: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 03/26/2024] Open
Abstract
During the COVID-19 pandemic, the interplay between the processes of immunity and senescence is drawing more and more intensive attention. SARS-CoV-2 infection induces senescence in lung cells, failure to clear infected cells and increased presence of inflammatory factors could lead to a cytokine storm and acute respiratory disease syndrome (ARDS), which together with aging and age-associated disease lead to 70% of COVID-19-related deaths. Studies on how senescence initiates upon viral infection and how to restrict excessive accumulation of senescent cells to avoid harmful inflammation are crucially important. Senescence can induce innate immune signaling, and innate immunity can engage cell senescence. Here, we mainly review the innate immune pathways, such as cGAS-STING, TLRs, NF-κB, and NLRP3 inflammasome, participating in the senescence process. In these pathways, IFN-I and inflammatory factors play key roles. At the end of the review, we propose the strategies by which we can improve the immune function and reduce inflammation based on these findings.
Collapse
Affiliation(s)
- Jinxiu Hou
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
6
|
Garrett TJ, Coatsworth H, Mahmud I, Hamerly T, Stephenson CJ, Ayers JB, Yazd HS, Miller MR, Lednicky JA, Dinglasan RR. Niclosamide as a chemical probe for analyzing SARS-CoV-2 modulation of host cell lipid metabolism. Front Microbiol 2023; 14:1251065. [PMID: 37901834 PMCID: PMC10603251 DOI: 10.3389/fmicb.2023.1251065] [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: 06/30/2023] [Accepted: 09/15/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction SARS-CoV-2 subverts host cell processes to facilitate rapid replication and dissemination, and this leads to pathological inflammation. Methods We used niclosamide (NIC), a poorly soluble anti-helminth drug identified initially for repurposed treatment of COVID-19, which activates the cells' autophagic and lipophagic processes as a chemical probe to determine if it can modulate the host cell's total lipid profile that would otherwise be either amplified or reduced during SARS-CoV-2 infection. Results Through parallel lipidomic and transcriptomic analyses we observed massive reorganization of lipid profiles of SARS-CoV-2 infected Vero E6 cells, especially with triglycerides, which were elevated early during virus replication, but decreased thereafter, as well as plasmalogens, which were elevated at later timepoints during virus replication, but were also elevated under normal cell growth. These findings suggested a complex interplay of lipid profile reorganization involving plasmalogen metabolism. We also observed that NIC treatment of both low and high viral loads does not affect virus entry. Instead, NIC treatment reduced the abundance of plasmalogens, diacylglycerides, and ceramides, which we found elevated during virus infection in the absence of NIC, resulting in a significant reduction in the production of infectious virions. Unexpectedly, at higher viral loads, NIC treatment also resulted in elevated triglyceride levels, and induced significant changes in phospholipid metabolism. Discussion We posit that future screens of approved or new partner drugs should prioritize compounds that effectively counter SARS-CoV-2 subversion of lipid metabolism, thereby reducing virus replication, egress, and the subsequent regulation of key lipid mediators of pathological inflammation.
Collapse
Affiliation(s)
- Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Heather Coatsworth
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Timothy Hamerly
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Caroline J. Stephenson
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Jasmine B. Ayers
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Hoda S. Yazd
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Megan R. Miller
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - John A. Lednicky
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Rhoel R. Dinglasan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
Zhang S, Zheng R, Pan Y, Sun H. Potential Therapeutic Value of the STING Inhibitors. Molecules 2023; 28:3127. [PMID: 37049889 PMCID: PMC10096477 DOI: 10.3390/molecules28073127] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The stimulator of interferon genes (STING) is a critical protein in the activation of the immune system in response to DNA. It can participate the inflammatory response process by modulating the inflammation-preferred translation program through the STING-PKR-like endoplasmic reticulum kinase (PERK)-eIF2α pathway or by inducing the secretion of type I interferons (IFNs) and a variety of proinflammatory factors through the recruitment of TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) or the regulation of the nuclear factor kappa-B (NF-κB) pathway. Based on the structure, location, function, genotype, and regulatory mechanism of STING, this review summarizes the potential value of STING inhibitors in the prevention and treatment of infectious diseases, psoriasis, systemic lupus erythematosus, non-alcoholic fatty liver disease, and other inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Shangran Zhang
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Runan Zheng
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yanhong Pan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| |
Collapse
|
8
|
Choubey D. Cytosolic DNA sensor IFI16 proteins: Potential molecular integrators of interactions among the aging hallmarks. Ageing Res Rev 2022; 82:101765. [PMID: 36270606 DOI: 10.1016/j.arr.2022.101765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/11/2022] [Accepted: 10/16/2022] [Indexed: 01/31/2023]
Abstract
Cellular changes that are linked to aging in humans include genomic instability, telomere attrition, epigenetic alterations, mitochondrial dysfunction, cellular senescence, and altered intercellular communications. The extent of the changes in these aging hallmarks and their interactions with each other are part of the human aging. However, the molecular mechanisms through which the aging hallmarks interact with each other remain unclear. Studies have indicated a potential role for the type I interferon (IFN) and p53-inducible IFI16 proteins in interactions with the aging hallmarks. The IFI16 proteins are members of the PYHIN protein family. Proteins in the family share a DNA-binding domain (the HIN domain) and a protein-protein interaction pyrin domain (PYD). IFI16 proteins are needed for cytosolic DNA-induced activation of the cGAS-STING pathway for type I IFN (IFN-β) expression. The pathway plays an important role in aging-related inflammation (inflammaging). Further, increased levels of the IFI16 proteins potentiate the cell growth inhibitory functions of the p53 and pRb tumor suppressors proteins. Moreover, IFI16 proteins are needed for most aging hallmarks. Therefore, here we discuss how an improved understanding of the role of the IFI16 proteins in integration of the aging hallmarks has potential to improve the human health and lifespan.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental & Public Health Sciences University of Cincinnati, 160 Panzeca Way, P.O. Box 670056, Cincinnati, OH 45267, USA.
| |
Collapse
|
9
|
Justice JL, Cristea IM. Nuclear antiviral innate responses at the intersection of DNA sensing and DNA repair. Trends Microbiol 2022; 30:1056-1071. [PMID: 35641341 PMCID: PMC9560981 DOI: 10.1016/j.tim.2022.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 01/13/2023]
Abstract
The coevolution of vertebrate and mammalian hosts with DNA viruses has driven the ability of host cells to distinguish viral from cellular DNA in the nucleus to induce intrinsic immune responses. Concomitant viral mechanisms have arisen to inhibit DNA sensing. At this virus-host interface, emerging evidence links cytokine responses and cellular homeostasis pathways, particularly the DNA damage response (DDR). Nuclear DNA sensors, such as the interferon (IFN)-γ inducible protein 16 (IFI16), functionally intersect with the DDR regulators ataxia telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK). Here, we discuss accumulating knowledge for the DDR-innate immunity signaling axis. Through the lens of this infection-driven signaling axis, we present host and viral molecular strategies acquired to regulate autoinflammation and antiviral responses.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
10
|
Uresti-Rivera EE, García-Hernández MH. AIM2-inflammasome role in systemic lupus erythematous and rheumatoid arthritis. Autoimmunity 2022; 55:443-454. [PMID: 35880661 DOI: 10.1080/08916934.2022.2103802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The inflammasome AIM2 regulates multiple aspects of innate immune functions and serves as a critical mediator of inflammatory responses. AIM2 inflammasome activation leads to the production of pro-inflammatory cytokines, IL-1β and IL-18 and participates triggering a pyroptosis response needed to counteract excessive cell proliferation. In addition, AIM2 expression and activation is wide regulated since alteration in its activity may derived in pathological consequences. Consequently, deregulated AIM2 activation contributes to the pathogenic processes of various inflammatory diseases. In this review, we will discuss the activation and function of AIM2 inflammasome, as well as its contribution in rheumatoid arthritis and systemic lupus erythematous pathology. Finally, we highlight the participation of the AIM2-inflammasome at the level of joint in rheumatoid arthritis and at kidney in systemic lupus erythematous. The development of therapeutic strategies based on modulation of AIM2-inflammasome activity should have a tissue-specific focus.
Collapse
Affiliation(s)
- E E Uresti-Rivera
- Research Center for Health Sciences and Biomedicine, UASLP, San Luis Potosi, Mexico.,Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, UASLP, San Luis Potosí, Mexico
| | - M H García-Hernández
- Instituto Mexicano del Seguro Social, IMSS, Unidad de Investigación Biomédica, Delegación Zacatecas, Zacatecas, México
| |
Collapse
|
11
|
Khan M, Nur S, Abdulaal W. A study on DNA methylation modifying natural compounds identified EGCG for induction of IFI16 gene expression related to the innate immune response in cancer cells. Oncol Lett 2022; 24:218. [PMID: 35707762 PMCID: PMC9178671 DOI: 10.3892/ol.2022.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/25/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Mohammad Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Suza Nur
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Wesam Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
12
|
Krump NA, Wang R, Liu W, Yang JF, Ma T, You J. Merkel Cell Polyomavirus Infection Induces an Antiviral Innate Immune Response in Human Dermal Fibroblasts. J Virol 2021; 95:e0221120. [PMID: 33883226 PMCID: PMC8437356 DOI: 10.1128/jvi.02211-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Merkel cell polyomavirus (MCPyV) infects most of the human population asymptomatically, but in rare cases it leads to a highly aggressive skin cancer called Merkel cell carcinoma (MCC). MCC incidence is much higher in aging and immunocompromised populations. The epidemiology of MCC suggests that dysbiosis between the host immune response and the MCPyV infectious cycle could contribute to the development of MCPyV-associated MCC. Insufficient restriction of MCPyV by normal cellular processes, for example, could promote the incidental oncogenic MCPyV integration events and/or entry into the original cell of MCC. Progress toward understanding MCPyV biology has been hindered by its narrow cellular tropism. Our discovery that primary human dermal fibroblasts (HDFs) support MCPyV infection has made it possible to closely model cellular responses to different stages of the infectious cycle. The present study reveals that the onset of MCPyV replication and early gene expression induces an inflammatory cytokine and interferon-stimulated gene (ISG) response. The cGAS-STING pathway, in coordination with NF-κB, mediates induction of this innate immune gene expression program. Further, silencing of cGAS or NF-κB pathway factors led to elevated MCPyV replication. We also discovered that the PYHIN protein IFI16 localizes to MCPyV replication centers but does not contribute to the induction of ISGs. Instead, IFI16 upregulates inflammatory cytokines in response to MCPyV infection by an alternative mechanism. The work described herein establishes a foundation for exploring how changes to the skin microenvironment induced by aging or immunodeficiency might alter the fate of MCPyV and its host cell to encourage carcinogenesis. IMPORTANCE MCC has a high rate of mortality and an increasing incidence. Immune-checkpoint therapies have improved the prognosis of patients with metastatic MCC. Still, a significant proportion of the patients fail to respond to immune-checkpoint therapies or have a medical need for iatrogenic immune-suppression. A greater understanding of MCPyV biology could inform targeted therapies for MCPyV-associated MCC. Moreover, cellular events preceding MCC oncogenesis remain largely unknown. The present study aims to explore how MCPyV interfaces with innate immunity during its infectious cycle. We describe how MCPyV replication and/or transcription elicit an innate immune response via cGAS-STING, NF-κB, and IFI16. We also explore the effects of this response on MCPyV replication. Our findings illustrate how healthy cellular conditions may allow low-level infection that evades immune destruction until highly active replication is restricted by host responses. Conversely, pathological conditions could result in unbridled MCPyV replication that licenses MCC tumorigenesis.
Collapse
Affiliation(s)
- Nathan A. Krump
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ranran Wang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wei Liu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - June F. Yang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tongcui Ma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jianxin You
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Human Prostate Epithelial Cells Activate the AIM2 Inflammasome upon Cellular Senescence: Role of POP3 Protein in Aging-Related Prostatic Inflammation. Life (Basel) 2021; 11:life11040366. [PMID: 33923931 PMCID: PMC8073538 DOI: 10.3390/life11040366] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Increased levels of type I (T1) interferon (IFN)-inducible POP3 protein in myeloid cells inhibit activation of the AIM2 inflammasome and production of IL-1β and IL-18 proinflammatory cytokines. The AIM2 mRNA levels were significantly higher in benign prostate hyperplasia (BPH) than the normal prostate. Further, human normal prostate epithelial cells (PrECs), upon becoming senescent, activated an inflammasome. Because in aging related BPH senescent PrECs accumulate, we investigated the role of POP3 and AIM2 proteins in pre-senescent and senescent PrECs. Here we report that the basal levels of the POP3 mRNA and protein were lower in senescent (versus young or old) PrECs that exhibited activation of the T1 IFN response. Further, treatment of PrECs and a BPH cell line (BPH-1) that expresses the androgen receptor (AR) with the male sex hormone dihydrotestosterone (DHT) increased the basal levels of POP3 mRNA and protein, but not AIM2, and inhibited activation of the AIM2 inflammasome. Of interest, a stable knockdown of POP3 protein expression in the BPH-1 cell line increased cytosolic DNA-induced activation of AIM2 inflammasome. These observations suggest a potential role of POP3 protein in aging-related prostatic inflammation.
Collapse
|
14
|
Zhang P, Lu B, Zhu R, Yang D, Liu W, Wang Q, Ji N, Chen Q, Ding Y, Liang X, Wang Q. Hyperglycemia accelerates inflammaging in the gingival epithelium through inflammasomes activation. J Periodontal Res 2021; 56:667-678. [PMID: 33650689 DOI: 10.1111/jre.12863] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Diabetes accelerates inflammaging in various tissue with an increase in senescent cell burden and senescence-associated secretory phenotype (SASP) secretion, which is a significant cause of tissue dysfunction and contributes to the diabetic complications. Recently, inflammasomes are thought to contribute to inflammaging. Here, utilizing diabetic models in vivo and in vitro, we investigated the potential association between hyperglycemia-induced inflammaging and gingival tissue dysfunction and the mechanism underlying inflammasome-associated inflammaging. MATERIALS AND METHODS Gingival epithelium and serum were collected from control and diabetic patients and mice. The expression of p16, p21, and inflammasomes in the gingival epithelium, SASP factors in serum, and the molecular factors associated with gingival epithelial barrier function were assessed. Human oral keratinocyte (HOK) was stimulated with normal and high glucose, and pre-treated with Z-YVAD-FMK (Caspase-1 inhibitor) prior to evaluating cellular senescence, SASP secretion, and inflammasome activation. RESULTS In vivo, hyperglycemia significantly elevated the local burden of senescent cells in the gingival epithelium and SASP factors in the serum and simultaneously reduced the expression levels of Claudin-1, E-cadherin, and Connexin 43 in the gingival epithelium. Interestingly, the inflammasomes were activated in the gingival epithelium. In vitro, high glucose-induced the inflammaging in HOK, and blocking inflammasome activation through inhibiting Caspase-1 and glucose-induced inflammaging. CONCLUSIONS Hyperglycemia accelerated inflammaging in the gingival epithelium through inflammasomes activation, which is potentially affiliated with a decline in the gingival epithelial barrier function in diabetes. Inflammasomes-related inflammaging may be the crucial mechanism underlying diabetic periodontitis and represents significant opportunities for advancing prevention and treatment options.
Collapse
Affiliation(s)
- Peng Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boyao Lu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dawei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weiqing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xing Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Hong SM, Lee J, Jang SG, Lee J, Cho ML, Kwok SK, Park SH. Type I Interferon Increases Inflammasomes Associated Pyroptosis in the Salivary Glands of Patients with Primary Sjögren's Syndrome. Immune Netw 2020; 20:e39. [PMID: 33163247 PMCID: PMC7609163 DOI: 10.4110/in.2020.20.e39] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Sjögren's syndrome (SS) is a chronic and systemic autoimmune disease characterized by lymphocytic infiltration in the exocrine glands. In SS, type I IFN has a pathogenic role, and recently, inflammasome activation has been observed in both immune and non-immune cells. However, the relationship between type I IFN and inflammasome-associated pyroptosis in SS has not been studied. We measured IL-18, caspase-1, and IFN-stimulated gene 15 (ISG15) in saliva and serum, and compared whether the expression levels of inflammasome and pyroptosis components, including absent in melanoma 2 (AIM2), NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC), caspase-1, gasdermin D (GSDMD), and gasdermin E (GSDME), in minor salivary gland (MSG) are related to the expression levels of type I IFN signature genes. Expression of type I IFN signature genes was correlated with mRNA levels of caspase-1 and GSDMD in MSG. In confocal analysis, the expression of caspase-1 and GSDMD was higher in salivary gland epithelial cells (SGECs) from SS patients. In the type I IFN-treated human salivary gland epithelial cell line, the expression of caspase-1 and GSDMD was increased, and pyroptosis was accelerated in a caspase-dependent manner upon inflammasome activation. In conclusion, we demonstrate that type I IFN may contribute to inflammasome-associated pyroptosis of the SGECs of SS patients, suggesting another pathogenic role of type I IFN in SS in terms of target tissue -SGECs destruction.
Collapse
Affiliation(s)
- Seung-Min Hong
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jaeseon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Se Gwang Jang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jennifer Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Ki Kwok
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
16
|
Global analysis of inverted repeat sequences in human gene promoters reveals their non-random distribution and association with specific biological pathways. Genomics 2020; 112:2772-2777. [DOI: 10.1016/j.ygeno.2020.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/02/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
|
17
|
Marchesan JT, Girnary MS, Moss K, Monaghan ET, Egnatz GJ, Jiao Y, Zhang S, Beck J, Swanson KV. Role of inflammasomes in the pathogenesis of periodontal disease and therapeutics. Periodontol 2000 2020; 82:93-114. [PMID: 31850638 PMCID: PMC6927484 DOI: 10.1111/prd.12269] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammasomes are a group of multimolecular intracellular complexes assembled around several innate immune proteins. Recognition of a diverse range of microbial, stress and damage signals by inflammasomes results in direct activation of caspase‐1, which subsequently induces the only known form of secretion of active interleukin‐1β and interleukin‐18. Although the importance of interleukin‐1β in the periodontium is not questioned, the impact of inflammasomes in periodontal disease and its potential for therapeutics in periodontology is still in its very early stages. Increasing evidence in preclinical models and human data strongly implicate the involvement of inflammasomes in a number of inflammatory, autoinflammatory and autoimmune disorders. Here we review: (a) the currently known inflammasome functions, (b) clinical/preclinical data supporting inflammasome involvement in the context of periodontal and comorbid diseases and (c) potential therapies targeting inflammasomes. To clarify further the inflammasome involvement in periodontitis, we present analyses of data from a large clinical study (n = 5809) that measured the gingival crevicular fluid‐interleukin‐1β and grouped the participants based on current periodontal disease classifications. We review data on 4910 European‐Americans that correlate 16 polymorphisms in the interleukin‐1B region with high gingival crevicular fluid‐interleukin‐1β levels. We show that inflammasome components are increased in diseased periodontal tissues and that the caspase‐1 inhibitor, VX‐765, inhibits ~50% of alveolar bone loss in experimental periodontitis. The literature review further supports that although patients clinically present with the same phenotype, the disease that develops probably has different underlying biological pathways. The current data indicate that inflammasomes have a role in periodontal disease pathogenesis. Understanding the contribution of different inflammasomes to disease development and distinct patient susceptibility will probably translate into improved, personalized therapies.
Collapse
Affiliation(s)
- Julie T Marchesan
- Department of Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mustafa Saadat Girnary
- Department of Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kevin Moss
- Department of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eugenia Timofeev Monaghan
- Department of Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Grant Joseph Egnatz
- Department of Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yizu Jiao
- Department of Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Shaoping Zhang
- Periodontics Department, College of Dentistry, University of Iowa, Iowa City, Iowa, USA
| | - Jim Beck
- Department of Dental Ecology, Adams School of Dentistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Karen V Swanson
- Department of Medicine, Infectious Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
18
|
Gonzalez-Perez AC, Stempel M, Chan B, Brinkmann MM. One Step Ahead: Herpesviruses Light the Way to Understanding Interferon-Stimulated Genes (ISGs). Front Microbiol 2020; 11:124. [PMID: 32117146 PMCID: PMC7018705 DOI: 10.3389/fmicb.2020.00124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
The host immune system is engaged in a constant battle with microorganisms, with the immediate detection of pathogenic invasion and subsequent signalling acting as crucial deterrents against the establishment of a successful infection. For this purpose, cells are equipped with a variety of sensors called pattern recognition receptors (PRR), which rapidly detect intruders leading to the expression of antiviral type I interferons (IFN). Type I IFN are crucial cytokines which exert their biological effects through the induction of hundreds of IFN-stimulated genes (ISGs). The expression profile of these ISGs varies depending on the virus. For a small subset of ISGs, their anti- or even proviral effects have been revealed, however, the vast majority are uncharacterised. The spotlight is now on herpesviruses, with their large coding capacity and long co-evolution with their hosts, as a key to understanding the impact of ISGs during viral infection. Studies are emerging which have identified multiple herpesviral antagonists specifically targeting ISGs, hinting at the significant role these proteins must play in host defence against viral infection, with the promise of more to come. In this review, we will discuss the current knowledge of the complex interplay between ISGs and human herpesviruses: the antiviral role of selected ISGs during herpesviral infections, how herpesviruses antagonise these ISGs and, in some cases, even exploit them to benefit viral infection.
Collapse
Affiliation(s)
| | - Markus Stempel
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Baca Chan
- Viral Genomics Group, Institute for Respiratory Health, The University of Western Australia, Perth, WA, Australia
| | - Melanie M. Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
19
|
Lee HC, Chathuranga K, Lee JS. Intracellular sensing of viral genomes and viral evasion. Exp Mol Med 2019; 51:1-13. [PMID: 31827068 PMCID: PMC6906418 DOI: 10.1038/s12276-019-0299-y] [Citation(s) in RCA: 380] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
During viral infection, virus-derived cytosolic nucleic acids are recognized by host intracellular specific sensors. The efficacy of this recognition system is crucial for triggering innate host defenses, which then stimulate more specific adaptive immune responses against the virus. Recent studies show that signal transduction pathways activated by sensing proteins are positively or negatively regulated by many modulators to maintain host immune homeostasis. However, viruses have evolved several strategies to counteract/evade host immune reactions. These systems involve viral proteins that interact with host sensor proteins and prevent them from detecting the viral genome or from initiating immune signaling. In this review, we discuss key regulators of cytosolic sensor proteins and viral proteins based on experimental evidence.
Collapse
Affiliation(s)
- Hyun-Cheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
- Central Research Institute, Komipharm International Co., Ltd, Shiheung, 15094, Korea
| | - Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea.
| |
Collapse
|
20
|
Méndez-Frausto G, Medina-Rosales MN, Uresti-Rivera EE, Baranda-Cándido L, Zapata-Zúñiga M, Bastián Y, González Amaro R, Enciso-Moreno JA, García-Hernández MH. Expression and activity of AIM2-inflammasome in rheumatoid arthritis patients. Immunobiology 2019; 225:151880. [PMID: 31836304 DOI: 10.1016/j.imbio.2019.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 11/26/2019] [Indexed: 01/12/2023]
Abstract
INTRODUCTION AIM2 inflammasome activation leads to the release of IL-β, which plays an important role in rheumatoid arthritis pathogenesis. In this work, we evaluated AIM2 expression and activity in RA patients and healthy controls. METHODS AIM2 and RANKL expression were evaluated by flow cytometry. Inflammasome activity was determined in monocyte cultures stimulated with synthetic DNA by measuring IL-1β levels in supernatants using an ELISA assay. The caspase-1 expression in monocytes was measured by western blot, the POP3 expression was analysed by qPCR, and serum levels of IFN-γ were evaluated using ELISA assay. RESULTS We observed a diminution of CD14+AIM2+ cells in RA patients, associated with disease activity and evolution. Likewise, the levels of IL-1β were increased in monocyte cultures un-stimulated and stimulated with LPS from RA patients with DAS28 ≥ 4. The Caspase-1 activity and RANKL + monocytes in RA patients were slightly increased. Finally, augmented POP3 expression and diminished IFN-γ serum levels were detected in RA patients. CONCLUSION Our results showed that the monocytes from RA patients were prone to release IL-1β in the absence of the AIM2 inflammasome signal. The down-regulation of AIM2 to a systemic level in RA patients might be a consequence of augmented POP3 expression and might imply the survival of pro-inflammatory cells contributing to the inflammation process.
Collapse
Affiliation(s)
- Gwendolyne Méndez-Frausto
- Unidad de Investigación Biomédica. Delegación Zacatecas. Instituto Mexicano del Seguro Social, IMSS, C.P. 98000, Mexico
| | - Marina Nayeli Medina-Rosales
- Unidad de Investigación Biomédica. Delegación Zacatecas. Instituto Mexicano del Seguro Social, IMSS, C.P. 98000, Mexico
| | - Edith Elena Uresti-Rivera
- Centro de Investigación en Ciencias de la Salud y Biomedicina. CICSaB Universidad Autónoma de San Luis Potosí, UASLP, C.P. 78000, Mexico
| | - Lourdes Baranda-Cándido
- Centro de Investigación en Ciencias de la Salud y Biomedicina. CICSaB Universidad Autónoma de San Luis Potosí, UASLP, C.P. 78000, Mexico; Unidad Regional de Reumatología y Osteoporosis Hospital Central Dr. Ignacio Morones Prieto. San Luis Potosí, SLP, C.P. 78290, Mexico
| | - Martín Zapata-Zúñiga
- Facultad de Medicina y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Hospital Rural No. 51 IMSS Bienestar, Villanueva, Zacatecas, C.P. 99559, Mexico
| | - Yadira Bastián
- Unidad de Investigación Biomédica. Delegación Zacatecas. Instituto Mexicano del Seguro Social, IMSS, C.P. 98000, Mexico; Cátedras CONACYT- Unidad de Investigación Biomédica de Zacatecas-IMSS, Zacatecas, C.P. 98000, Mexico
| | - Roberto González Amaro
- Centro de Investigación en Ciencias de la Salud y Biomedicina. CICSaB Universidad Autónoma de San Luis Potosí, UASLP, C.P. 78000, Mexico
| | - José Antonio Enciso-Moreno
- Unidad de Investigación Biomédica. Delegación Zacatecas. Instituto Mexicano del Seguro Social, IMSS, C.P. 98000, Mexico
| | | |
Collapse
|
21
|
Choubey D. Type I interferon (IFN)-inducible Absent in Melanoma 2 proteins in neuroinflammation: implications for Alzheimer's disease. J Neuroinflammation 2019; 16:236. [PMID: 31771614 PMCID: PMC6880379 DOI: 10.1186/s12974-019-1639-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/11/2019] [Indexed: 01/09/2023] Open
Abstract
Cumulative evidence indicates that activation of innate immune responses in the central nervous system (CNS) induces the expression of type 1 interferons (T1 IFNs), a family of cytokines. The T1 IFNs (IFN-α/β), through activation of the JAK/STAT-signaling in microglia, astrocytes, and neurons, induce the expression of IFN-inducible proteins, which mediate the pro- and anti-inflammatory functions of IFNs. Accordingly, T1 IFN-inducible Absent in Melanoma 2 proteins (murine Aim2 and human AIM2) negatively regulate the expression of TI IFNs and, upon sensing higher levels of cytosolic DNA, assemble the Aim2/AIM2 inflammasome, resulting in activation of caspase-1, pyroptosis, and the secretion of pro-inflammatory cytokines (e.g., IL-1β and IL-18). Of interest, studies have indicated a role for the Aim2/AIM2 proteins in neuroinflammation and neurodegenerative diseases, including Alzheimer's disease (AD). The ability of Aim2/AIM2 proteins to exert pro- and anti-inflammatory effects in CNS may depend upon age, sex hormones, cell-types, and the expression of species-specific negative regulators of the Aim2/AIM2 inflammasome. Therefore, we discuss the role of Aim2/AIM2 proteins in the development of AD. An improved understanding of the role of Absent in Melanoma 2 proteins in AD could identify new approaches to treat patients.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P. O. Box 670056, Cincinnati, OH, 45267, USA.
| |
Collapse
|
22
|
Davis SE, Khatua AK, Popik W. Nucleosomal dsDNA Stimulates APOL1 Expression in Human Cultured Podocytes by Activating the cGAS/IFI16-STING Signaling Pathway. Sci Rep 2019; 9:15485. [PMID: 31664093 PMCID: PMC6820523 DOI: 10.1038/s41598-019-51998-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022] Open
Abstract
APOL1 alleles G1 and G2 are associated with faster progression to lupus nephritis (LN)-associated end-stage renal disease (LN-ESRD) in African Americans. Increased levels of type I interferons (IFNs) and nucleosome-associated double-stranded DNA (dsDNA) fragments (nsDNA) are the hallmark of this disease. Here, we identify cyclic GMP-AMP synthase (cGAS) and interferon-inducible protein 16 (IFI16) as the major DNA sensors in human immortalized podocytes. We also show that nsDNA triggers the expression of APOL1 and IFNβ via IRF3 activation through the cGAS/IFI16-STING pathway. We demonstrate that maximal APOL1 expression also requires the activation of type I IFN receptor (IFNAR) and STAT1 signaling triggered by IFNβ produced in response to nsDNA, or by exogenous IFNβ. Finally, we show that STAT1 activation is sufficient to upregulate IFI16, subsequently boosting APOL1 expression through a positive feedback mechanism. Collectively, we find that nsDNA-induced APOL1 expression is mediated by both IFNβ-independent and dependent signaling pathways triggered by activation of the cGAS/IFI16-STING pathway. We propose that simultaneous inhibition of STING and the IFNAR-STAT1 pathway may attenuate IFI16 expression, reduce IFI16-cGAS cross-talk, and prevent excessive APOL1 expression in human podocytes in response to nsDNA.
Collapse
Affiliation(s)
- Shamara E Davis
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA
| | - Atanu K Khatua
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA
| | - Waldemar Popik
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA.
- Department of Internal Medicine, 1005 D. B. Todd Blvd, Nashville, TN, 37208, USA.
| |
Collapse
|
23
|
Lan YY, Heather JM, Eisenhaure T, Garris CS, Lieb D, Raychowdhury R, Hacohen N. Extranuclear DNA accumulates in aged cells and contributes to senescence and inflammation. Aging Cell 2019; 18:e12901. [PMID: 30706626 PMCID: PMC6413746 DOI: 10.1111/acel.12901] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Systemic inflammation is central to aging‐related conditions. However, the intrinsic factors that induce inflammation are not well understood. We previously identified a cell‐autonomous pathway through which damaged nuclear DNA is trafficked to the cytosol where it activates innate cytosolic DNA sensors that trigger inflammation. These results led us to hypothesize that DNA released after cumulative damage contributes to persistent inflammation in aging cells through a similar mechanism. Consistent with this notion, we found that older cells harbored higher levels of extranuclear DNA compared to younger cells. Extranuclear DNA was exported by a leptomycin B‐sensitive process, degraded through the autophagosome–lysosomal pathway and triggered innate immune responses through the DNA‐sensing cGAS‐STING pathway. Patient cells from the aging diseases ataxia and progeria also displayed extranuclear DNA accumulation, increased pIRF3 and pTBK1, and STING‐dependent p16 expression. Removing extranuclear DNA in old cells using DNASE2A reduced innate immune responses and senescence‐associated (SA) β‐gal enzyme activity. Cells and tissues of Dnase2a−/− mice with defective DNA degradation exhibited slower growth, higher activity of β‐gal, or increased expression of HP‐1β and p16 proteins, while Dnase2a−/−;Sting−/− cells and tissues were rescued from these phenotypes, supporting a role for extranuclear DNA in senescence. We hypothesize a direct role for excess DNA in aging‐related inflammation and in replicative senescence, and propose DNA degradation as a therapeutic approach to remove intrinsic DNA and revert inflammation associated with aging.
Collapse
Affiliation(s)
- Yuk Yuen Lan
- Center for Cancer Research; Massachusetts General Hospital; Charlestown Massachusetts
- Broad Institute; Cambridge Massachusetts
- Department of Medicine; Harvard Medical School; Boston Massachusetts
| | - James M. Heather
- Center for Cancer Research; Massachusetts General Hospital; Charlestown Massachusetts
| | | | - Christopher Stuart Garris
- Center for Systems Biology; Massachusetts General Hospital; Boston Massachusetts
- Graduate Program in Immunology; Harvard Medical School; Boston Massachusetts
| | - David Lieb
- Broad Institute; Cambridge Massachusetts
| | | | - Nir Hacohen
- Center for Cancer Research; Massachusetts General Hospital; Charlestown Massachusetts
- Broad Institute; Cambridge Massachusetts
- Department of Medicine; Harvard Medical School; Boston Massachusetts
| |
Collapse
|
24
|
Choubey D, Panchanathan R. Interferon (IFN)-inducible Absent in Melanoma 2 proteins in the negative regulation of the type I IFN response: Implications for lupus nephritis. Cytokine 2019; 132:154682. [PMID: 30904426 DOI: 10.1016/j.cyto.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that exhibits a strong female bias (female-to-male ratio 9:1) in patients. Further, 40-60% SLE patients develop lupus nephritis (LN), which significantly increases the mortality rates. The failure of current therapies to adequately treat LN in patients reflects an incomplete understanding of the disease pathogenesis. Notably, a chronic increase in serum interferon-α (IFN-α) activity is a heritable risk factor to develop SLE. Accordingly, blood cells from most SLE patients with an active disease exhibit an increase in the expression of the type I IFN (IFN-α/β)-stimulated genes (ISGs, also referred to as "IFN-signature"), a type I IFN response. Further, LN patients during renal flares also exhibit an "IFN-signature" in renal biopsies. Therefore, an improved understanding of the regulation of type I IFNs expression is needed. Basal levels of the IFN-β through "priming" of IFN-α producing cells augment the expression of the IFN-α genes. Of interest, recent studies have indicated a role for the type I IFN-inducible Absent in Melanoma 2 proteins (the murine Aim2 and human AIM2) in the negative regulation of the type I IFN response through inflammasome-dependent and independent mechanisms. Further, an increase in the expression of Aim2 and AIM2 proteins in kidney and renal macrophages associated with the development of nephritis. Therefore, we discuss the role of Aim2/AIM2 proteins in the regulation of type I IFNs and LN. An improved understanding of the mechanisms by which the Absent in Melanoma 2 proteins suppress the type I IFN response and modulate nephritis is key to identify novel therapeutic targets to treat a group of LN patients.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States; Research Service, ML-151, Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States.
| | - Ravichandran Panchanathan
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States; Research Service, ML-151, Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States
| |
Collapse
|
25
|
Dong M, Yang L, Qu M, Hu X, Duan H, Zhang X, Shi W, Zhou Q. Autocrine IL-1β mediates the promotion of corneal neovascularization by senescent fibroblasts. Am J Physiol Cell Physiol 2018; 315:C734-C743. [PMID: 30156862 DOI: 10.1152/ajpcell.00205.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our previous study has confirmed that senescent fibroblasts promote corneal neovascularization (CNV) partially via the enhanced secretion of matrix metalloproteases (MMPs). However, the regulation of MMP expression in senescent fibroblasts remained unclear. In this study, we identified that the expression and secretion levels of interleukin-1β (IL-1β) were significantly upregulated in senescent human corneal fibroblasts than that in normal fibroblasts. Moreover, compared with vehicle-pretreated senescent fibroblasts, IL-1β pretreatment enhanced the expression of angiogenic factors but reduced the expression of angiostatic factors in senescent fibroblasts. When cocultured with human umbilical vein endothelial cells, IL-1β-pretreated senescent fibroblasts more strongly promoted their proliferation, migration, and tube-formation capacities than the vehicle-controlled senescent fibroblasts. In addition, either interleukin-1 receptor antagonist or anti-IL-1β neutralization completely inhibited the promotion of senescent fibroblasts in vascular tube formation in vitro and CNV in vivo. Therefore, we concluded that autocrine IL-1β mediated the promotion of senescent fibroblasts on corneal neovascularization.
Collapse
Affiliation(s)
- Muchen Dong
- Shandong Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences , Jinan , China
| | - Lingling Yang
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Mingli Qu
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Xiaoli Hu
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Haoyun Duan
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Xiaoping Zhang
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Weiyun Shi
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| | - Qingjun Zhou
- Shandong Provincial Key Lab of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences , Qingdao, Shandong , China
| |
Collapse
|
26
|
Yang W, Wang H, Ju H, Dou C. A study on the correlation between STAT‑1 and mutant p53 expression in glioma. Mol Med Rep 2018; 17:7807-7812. [PMID: 29620180 DOI: 10.3892/mmr.2018.8796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 01/05/2017] [Indexed: 11/06/2022] Open
Abstract
Glioma is the most common primary brain tumor in adults and the second most common malignant tumor in children. Aberrant expression of signal transducer and activator of transcription 1 (STAT‑1) and p53 are known to affect the occurrence and progression of malignant tumors. The aim of the present study was to investigate the expression of STAT‑1 and mutant p53 gene, as well as their correlation, in patients with glioma. The present study included 50 patients who underwent glioma resection at the First Affiliated Hospital of Inner Mongolia Medical University between December 2007 and December 2011, and 10 patients with acute cerebral contusion who underwent intracerebral hematoma removal at the same hospital between January 2013 and January 2014. The expression of STAT‑1 and mutant p53 protein in patients with different grades of glioma was assessed by immunohistochemistry. Spearman's correlation coefficient was employed to examine the correlation between STAT‑1 and the grade of glioma, and mutant p53 expression. The results demonstrated that the mean expression of STAT‑1 in glioma was significantly lower compared with normal brain tissue (P<0.05). However, there was no significant difference in the STAT‑1 positive expression rate between the two groups (χ2=1.38, P>0.05). The expression score (P<0.05) and positive expression rate (χ2=31.27, P<0.05) of mutant p53 in glioma was significantly higher compared with those in normal brain tissue. Statistical analysis revealed a negative correlation between STAT‑1 expression and the grade of glioma (r=‑0.767, P<0.05). In addition, mutant p53 expression was negatively correlated with STAT‑1 expression in glioma (r=‑0.876, P<0.05). The observed negative correlation between STAT‑1 and the pathological grade of glioma suggested an association between STAT‑1 and the occurrence and development of glioma, thus revealing the potential of STAT‑1 as a diagnostic biomarker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Wenbo Yang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010055, P.R. China
| | - Hongwei Wang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010055, P.R. China
| | - Haitao Ju
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010055, P.R. China
| | - Changwu Dou
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010055, P.R. China
| |
Collapse
|
27
|
Nosratabadi R, Alavian SM, Zare-Bidaki M, Shahrokhi VM, Arababadi MK. Innate immunity related pathogen recognition receptors and chronic hepatitis B infection. Mol Immunol 2017; 90:64-73. [PMID: 28704708 DOI: 10.1016/j.molimm.2017.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/07/2017] [Accepted: 07/01/2017] [Indexed: 01/30/2023]
Abstract
Innate immunity consists of several kinds of pathogen recognition receptors (PRRs), which participate in the recognition of pathogens and consequently activation of innate immune system against pathogens. Recently, several investigations reported that PRRs may also play key roles in the induction/stimulation of immune system related complications in microbial infections. Hepatitis B virus (HBV), as the main cause of viral hepatitis in human, can induce several clinical forms of hepatitis B and also might be associated with hepatic complications such as cirrhosis and hepatocellular carcinoma (HCC). Based on the important roles of PRRs in the eradication of microbial infections including viral infections and their related complications, it appears that the molecules may be a main part of immune responses against viral infections including HBV and participate in the HBV related complications. Thus, this review article has brought together information regarding the roles of PRRs in immunity against HBV and its complications.
Collapse
Affiliation(s)
- Reza Nosratabadi
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Dept. of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Seyed Moayed Alavian
- Baqiyatallah Research Center for Gastroenterology and Liver Disease, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Zare-Bidaki
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Dept. of Microbiology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Vahid Mohammadi Shahrokhi
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Dept. of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Kazemi Arababadi
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Dept. of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
28
|
Birch J, Passos JF. Targeting the SASP to combat ageing: Mitochondria as possible intracellular allies? Bioessays 2017; 39. [DOI: 10.1002/bies.201600235] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jodie Birch
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing; Newcastle University; Newcastle Upon Tyne UK
| | - João F. Passos
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
29
|
Choubey D, Panchanathan R. Absent in Melanoma 2 proteins in SLE. Clin Immunol 2017; 176:42-48. [PMID: 28062222 DOI: 10.1016/j.clim.2016.12.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/29/2016] [Accepted: 12/31/2016] [Indexed: 12/13/2022]
Abstract
Type I interferons (IFN-α/β)-inducible PYRIN and HIN domain-containing protein family includes Absent in Melanoma 2 (murine Aim2 and human AIM2), murine p202, and human PYRIN-only protein 3 (POP3). The generation of Aim2-deficient mice indicated that the Aim2 protein is essential for inflammasome activation, resulting in the secretion of interleukin-1β (IL-1β) and IL-18 and cell death by pyroptosis. Further, Aim2-deficiency also increased constitutive expression of the IFN-β and expression of the p202 protein. Notably, an increased expression of p202 protein in female mice associated with the development of systemic lupus erythematosus (SLE). SLE in patients is characterized by a constitutive increase in serum levels of IFN-α and an increase in the expression IFN-stimulated genes. Recent studies indicate that p202 and POP3 proteins inhibit activation of the Aim2/AIM2 inflammasome and promote IFN-β expression. Therefore, we discuss the role of Aim2/AIM2 proteins in the suppression of type I IFNs production and lupus susceptibility.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P. O. Box-670056, Cincinnati, OH 45267, United States; Research Service, ML-151, Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States.
| | - Ravichandran Panchanathan
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P. O. Box-670056, Cincinnati, OH 45267, United States; Research Service, ML-151, Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States
| |
Collapse
|
30
|
Choubey D. Absent in melanoma 2 proteins in the development of cancer. Cell Mol Life Sci 2016; 73:4383-4395. [PMID: 27328971 PMCID: PMC11108365 DOI: 10.1007/s00018-016-2296-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/04/2016] [Accepted: 06/16/2016] [Indexed: 12/19/2022]
Abstract
Recent studies utilizing chemical-induced colitis-associated and sporadic colon cancer in mouse models indicated a protective role for absent in melanoma 2 (Aim2) in colon epithelial cells. Accordingly, mutations in the human AIM2 gene have been found in colorectal cancer (CRC), and reduced expression of AIM2 in CRC is associated with its progression. Furthermore, the overexpression of AIM2 protein in human cancer cell lines inhibits cell proliferation. Interferon-inducible Aim2 and AIM2 are members of the PYHIN (PYRIN and HIN domain-containing) protein family and share ~57 % amino acid identity. The family also includes murine p202, human PYRIN-only protein 3, and IFI16, which negatively regulate Aim2/AIM2 functions. Because the CRC incidence and mortality rates are higher among men compared with women and the expression of Aim2/AIM2 proteins and their regulators is dependent upon age, gender, and sex hormones, we discuss the potential roles of Aim2/AIM2 in the development of cancer. An improved understanding of the biological functions of the AIM2 in the development of CRC will likely identify new therapeutic approaches to treat patients.
Collapse
Affiliation(s)
- Divaker Choubey
- Research Service, Cincinnati VA Medical Center, 3200 Vine Street, ML-151, Cincinnati, OH, 45220, USA.
- Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P. O. Box-670056, Cincinnati, OH, 45267, USA.
| |
Collapse
|
31
|
Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep 2016; 43:1213-1220. [DOI: 10.1007/s11033-016-4065-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022]
|
32
|
Choubey D, Panchanathan R. IFI16, an amplifier of DNA-damage response: Role in cellular senescence and aging-associated inflammatory diseases. Ageing Res Rev 2016; 28:27-36. [PMID: 27063514 DOI: 10.1016/j.arr.2016.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/30/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022]
Abstract
DNA-damage induces a DNA-damage response (DDR) in mammalian cells. The response, depending upon the cell-type and the extent of DNA-damage, ultimately results in cell death or cellular senescence. DDR-induced signaling in cells activates the ATM-p53 and ATM-IKKα/β-interferon (IFN)-β signaling pathways, thus leading to an induction of the p53 and IFN-inducible IFI16 gene. Further, upon DNA-damage, DNA accumulates in the cytoplasm, thereby inducing the IFI16 protein and STING-dependent IFN-β production and activation of the IFI16 inflammasome, resulting in the production of proinflammatory cytokines (e.g., IL-1β and IL-18). Increased expression of IFI16 protein in a variety of cell-types promotes cellular senescence. However, reduced expression of IFI16 in cells promotes cell proliferation. Because expression of the IFI16 gene is induced by activation of DNA-damage response in cells and increased levels of IFI16 protein in cells potentiate the p53-mediated transcriptional activation of genes and p53 and pRb-mediated cell cycle arrest, we discuss how an improved understanding of the role of IFI16 protein in cellular senescence and associated inflammatory secretory phenotype is likely to identify the molecular mechanisms that contribute to the development of aging-associated human inflammatory diseases and a failure to cancer therapy.
Collapse
Affiliation(s)
- Divaker Choubey
- Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States; Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States.
| | - Ravichandran Panchanathan
- Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States; Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States
| |
Collapse
|
33
|
Sharma N, Jha S. NLR-regulated pathways in cancer: opportunities and obstacles for therapeutic interventions. Cell Mol Life Sci 2016; 73:1741-64. [PMID: 26708292 PMCID: PMC11108278 DOI: 10.1007/s00018-015-2123-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 02/08/2023]
Abstract
NLRs (nucleotide-binding domain, leucine-rich repeat containing receptors) are pattern recognition receptors associated with immunity and inflammation in response to endogenous and exogenous pathogen and damage associated molecular patterns (PAMPs and DAMPs respectively). Dysregulated NLR function is associated with several diseases including cancers, metabolic diseases, autoimmune disorders and autoinflammatory syndromes. In the last decade, distinct cell and organ specific roles for NLRs have been identified however; their roles in cancer initiation, development and progression remain controversial. This review summarizes the emerging role of NLRs in cancer and their possible future as targets for cancer therapeutics.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Biology, Indian Institute of Technology Jodhpur, Old Residency Road, Ratanada, Jodhpur, Rajasthan, 342011, India
| | - Sushmita Jha
- Department of Biology, Indian Institute of Technology Jodhpur, Old Residency Road, Ratanada, Jodhpur, Rajasthan, 342011, India.
| |
Collapse
|
34
|
Reduced levels of cytosolic DNA sensor AIM2 are associated with impaired cytokine responses in healthy elderly. Exp Gerontol 2016; 78:39-46. [PMID: 26944367 DOI: 10.1016/j.exger.2016.02.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/02/2016] [Accepted: 02/26/2016] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Human aging is associated with remodeling of the immune system. While most studies on immunosenescence have focused on adaptive immunity, the effects of aging on innate immunity are not well understood. Here, we investigated whether aging affects cytokine responses to a wide range of well-defined pattern recognition receptor (PRR) ligands, such as ligands for Toll-like receptors (TLRs), C-type lectin receptors (CLRs), NOD-like receptors (NLRs), retinoic-acid-inducible gene-I like receptors (RLRs) and the cytosolic DNA sensor absent in melanoma 2 (AIM2). METHOD Blood was collected from 16 young (20-39 years) and 18 elderly (60-84 years) healthy participants. Pro-inflammatory cytokine (TNF-α, IL-1β, IL-6, and IL-8) production in a whole blood assay (WBA) after stimulation with TLR ligands (Pam3csk4, poly(I:C), LPS, CpG), CLR ligand (β-glucan), NLR ligand (MDP), RLR ligands (5'ppp-dsDNA and poly(I:C)/lyovec) and the AIM2 ligand (poly(dA:dT) was assessed by ELISA. TLR2 and TLR4 expression by leukocytes and monocytes was determined by flow-cytometry. Expression of AIM2 by peripheral blood mononuclear cells (PBMC) was assessed by qRT-PCR and Western blot. RESULT Cytokine responses to Pam3csk4, poly(I:C) and CpG, β-glucan, MDP, 5'ppp-dsDNA and poly(I:C)/lyovec were comparable between young and old participants. We observed a higher IL-8 response following stimulation of elderly blood samples with the TLR4 ligand LPS, which was associated with higher proportions of TLR4 expressing monocytes. Interestingly, stimulation of whole blood cells with the AIM2 ligand poly(dA:dT) resulted in significantly lower cytokine responses in old participants. Moreover, these lower cytokine responses were associated with lower AIM2 protein expression and activation in PBMC of old participants. CONCLUSION Our findings reveal an age-dependent reduction of AIM2 expression and activation which may explain reduced cytokine responses to the cytosolic DNA mimic poly(dA:dT) in healthy elderly individuals. Reduced AIM2-mediated sensing with age may contribute to increased vulnerability to bacterial or viral infections in the elderly.
Collapse
|
35
|
Ebersole JL, Kirakodu S, Novak MJ, Exposto CR, Stromberg AJ, Shen S, Orraca L, Gonzalez-Martinez J, Gonzalez OA. Effects of aging in the expression of NOD-like receptors and inflammasome-related genes in oral mucosa. Mol Oral Microbiol 2016; 31:18-32. [PMID: 26197995 PMCID: PMC4712099 DOI: 10.1111/omi.12121] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2015] [Indexed: 01/28/2023]
Abstract
The molecular changes underlying the higher risk of chronic inflammatory disorders during aging remain incompletely understood. Molecular variations in the innate immune response related to recognition and interaction with microbes at mucosal surfaces could be involved in aging-related inflammation. We developed an ontology analysis of 20 nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) and seven inflammasome-related genes (IRGs) in healthy and inflamed/periodontitis oral mucosal tissues from young, adolescent, adult, and aged non-human primates (Macaca mulatta) using the GeneChip(®) Rhesus Macaque Genome array. Validation of some of the significant changes was done by quantitative reverse transcription-polymerase chain reaction. The expression of NLRB/NAIP, NLRP12, and AIM2 increased with aging in healthy mucosa whereas NLRC2/NOD2 expression decreased. Although higher expression levels of some NLRs were generally observed with periodontitis in adult mucosal tissues (e.g. NLRB/NAIP, NLRP5, and NLRX1), various receptors (e.g. NLRC2/NOD2 and NLRP2) and the inflammasome adaptor protein ASC, exhibited a significant reduction in expression in aged periodontitis tissues. Accordingly, the expression of NLR-activated innate immune genes, such as HBD3 and IFNB1, was impaired in aged but not adult periodontitis tissues. Both adult and aged tissues showed significant increase in interleukin-1β expression. These findings suggest that the expression of a subset of NLRs appears to change with aging in healthy oral mucosa, and that aging-related oral mucosal inflammation could involve an impaired regulation of the inflammatory and antimicrobial response associated with downregulation of specific NLRs and IRGs.
Collapse
Affiliation(s)
- Jeffrey L. Ebersole
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - M. John Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Cristina R. Exposto
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Arnold J. Stromberg
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Shu Shen
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Luis Orraca
- School of Dental Medicine, University of Puerto Rico, San Juan, PR
| | | | - Octavio A. Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
36
|
Strong Upregulation of AIM2 and IFI16 Inflammasomes in the Mucosa of Patients with Active Inflammatory Bowel Disease. Inflamm Bowel Dis 2015; 21:2673-82. [PMID: 26313692 DOI: 10.1097/mib.0000000000000535] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by a chronic inflammation of the gut, partly driven by defects in the innate immune system. Considering the central role of inflammasome signaling in innate immunity, we studied inflammasome components in IBD mucosa. METHODS Expression of genes encoding inflammasome sensor subunits was investigated in colonic mucosal biopsies from 2 cohorts of patients with IBD and controls. RESULTS A significant upregulation (>2-fold change in expression, false discovery rate <0.05) of the PYHIN inflammasomes AIM2 and IFI16 in active IBD versus controls was found. Also IFI16 was significantly increased in inactive IBD versus controls. Moreover, responders to anti-tumor necrosis factor therapy showed decreased expression of these inflammasomes although IFI16 remained significantly increased in responders showing endoscopic healing versus controls. AIM2 was mainly expressed in epithelial cells, whereas IFI16 was expressed in both lymphocytes and epithelial cells. Functional activation of predominant AIM2/IFI16-mediated inflammasomes in active IBD colon was shown by the presence of the downstream effectors CASP1 and HMGB-1 in inflamed mucosa. CONCLUSIONS Our results highlight the importance of PYHIN inflammasome signaling in IBD and also link anti-tumor necrosis factor responsiveness to inflammasome signaling. Together, this points to the potential value of the inflammasome pathway as a new therapeutic target for IBD treatment.
Collapse
|
37
|
Wu X, Hakimi M, Wortmann M, Zhang J, Böckler D, Dihlmann S. Gene expression of inflammasome components in peripheral blood mononuclear cells (PBMC) of vascular patients increases with age. IMMUNITY & AGEING 2015; 12:15. [PMID: 26448778 PMCID: PMC4596365 DOI: 10.1186/s12979-015-0043-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/23/2015] [Indexed: 11/17/2022]
Abstract
Background Chronic low-grade inflammation is considered a driver of many age-related disorders, including vascular diseases (inflammaging). Inhibition of autophagic capacity with ageing was postulated to generate a pro-inflammatory condition via activation of inflammasomes, a group of Interleukin-1 activating intracellular multi-protein complexes. We thus investigated gene expression of inflammasome components in PBMC of 77 vascular patients (age 22–82) in association with age. Findings Linear regression of real-time qRT-PCR data revealed a significant positive association of gene expression of each of the inflammasome components with age (Pearson correlation coefficients: AIM2: r = 0.245; P = 0.032; NLRP3: r = 0.367; P = 0.001; ASC (PYCARD): r = 0.252; P = 0.027; CASP1: r = 0.296; P = 0.009; CASP5: r = 0.453; P = 0.00003; IL1B: r = 0.247; P = 0.030). No difference in gene expression of AIM2, NLRP3, ASC CASP1, and CASP5 was detected between PBMC of patients with advanced atherosclerosis and other vascular patients, whereas IL1B expression was increased in PBMC of the latter group (P = 0.0005). Conclusion The findings reinforce the systemic pro-inflammatory phenotype reported in elderly by demonstrating an increased phase-1 activation of inflammasomes in PBMC of vascular patients. Electronic supplementary material The online version of this article (doi:10.1186/s12979-015-0043-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany ; Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Maani Hakimi
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany ; Vaskuläre Biomaterialbank Heidelberg, VBBH, Heidelberg, Germany
| | - Markus Wortmann
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Jian Zhang
- Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Susanne Dihlmann
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| |
Collapse
|
38
|
Piera-Velazquez S, Jimenez SA. Role of cellular senescence and NOX4-mediated oxidative stress in systemic sclerosis pathogenesis. Curr Rheumatol Rep 2015; 17:473. [PMID: 25475596 DOI: 10.1007/s11926-014-0473-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Systemic sclerosis (SSc) is a systemic autoimmune disease characterized by progressive fibrosis of skin and numerous internal organs and a severe fibroproliferative vasculopathy resulting frequently in severe disability and high mortality. Although the etiology of SSc is unknown and the detailed mechanisms responsible for the fibrotic process have not been fully elucidated, one important observation from a large US population study was the demonstration of a late onset of SSc with a peak incidence between 45 and 54 years of age in African-American females and between 65 and 74 years of age in white females. Although it is not appropriate to consider SSc as a disease of aging, the possibility that senescence changes in the cellular elements involved in its pathogenesis may play a role has not been thoroughly examined. The process of cellular senescence is extremely complex, and the mechanisms, molecular events, and signaling pathways involved have not been fully elucidated; however, there is strong evidence to support the concept that oxidative stress caused by the excessive generation of reactive oxygen species may be one important mechanism involved. On the other hand, numerous studies have implicated oxidative stress in SSc pathogenesis, thus, suggesting a plausible mechanism in which excessive oxidative stress induces cellular senescence and that the molecular events associated with this complex process play an important role in the fibrotic and fibroproliferative vasculopathy characteristic of SSc. Here, recent studies examining the role of cellular senescence and of oxidative stress in SSc pathogenesis will be reviewed.
Collapse
Affiliation(s)
- Sonsoles Piera-Velazquez
- Scleroderma Center, Thomas Jefferson University, 233 South 10th Street, Suite 509 BLSB, Philadelphia, PA, 19107, USA
| | | |
Collapse
|
39
|
The DNA damage response and immune signaling alliance: Is it good or bad? Nature decides when and where. Pharmacol Ther 2015; 154:36-56. [PMID: 26145166 DOI: 10.1016/j.pharmthera.2015.06.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/10/2015] [Indexed: 12/15/2022]
Abstract
The characteristic feature of healthy living organisms is the preservation of homeostasis. Compelling evidence highlight that the DNA damage response and repair (DDR/R) and immune response (ImmR) signaling networks work together favoring the harmonized function of (multi)cellular organisms. DNA and RNA viruses activate the DDR/R machinery in the host cells both directly and indirectly. Activation of DDR/R in turn favors the immunogenicity of the incipient cell. Hence, stimulation of DDR/R by exogenous or endogenous insults triggers innate and adaptive ImmR. The immunogenic properties of ionizing radiation, a prototypic DDR/R inducer, serve as suitable examples of how DDR/R stimulation alerts host immunity. Thus, critical cellular danger signals stimulate defense at the systemic level and vice versa. Disruption of DDR/R-ImmR cross talk compromises (multi)cellular integrity, leading to cell-cycle-related and immune defects. The emerging DDR/R-ImmR concept opens up a new avenue of therapeutic options, recalling the Hippocrates quote "everything in excess is opposed by nature."
Collapse
|
40
|
Martin-Gayo E, Buzon MJ, Ouyang Z, Hickman T, Cronin J, Pimenova D, Walker BD, Lichterfeld M, Yu XG. Potent Cell-Intrinsic Immune Responses in Dendritic Cells Facilitate HIV-1-Specific T Cell Immunity in HIV-1 Elite Controllers. PLoS Pathog 2015; 11:e1004930. [PMID: 26067651 PMCID: PMC4466270 DOI: 10.1371/journal.ppat.1004930] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/01/2015] [Indexed: 12/29/2022] Open
Abstract
The majority of HIV-1 elite controllers (EC) restrict HIV-1 replication through highly functional HIV-1-specific T cell responses, but mechanisms supporting the evolution of effective HIV-1-specific T cell immunity in these patients remain undefined. Cytosolic immune recognition of HIV-1 in conventional dendritic cells (cDC) can facilitate priming and expansion of HIV-1-specific T cells; however, HIV-1 seems to be able to avoid intracellular immune recognition in cDCs in most infected individuals. Here, we show that exposure of cDCs from EC to HIV-1 leads to a rapid and sustained production of type I interferons and upregulation of several interferon-stimulated effector genes. Emergence of these cell-intrinsic immune responses was associated with a reduced induction of SAMHD1 and LEDGF/p75, and an accumulation of viral reverse transcripts, but inhibited by pharmacological blockade of viral reverse transcription or siRNA-mediated silencing of the cytosolic DNA sensor cGAS. Importantly, improved cell-intrinsic immune recognition of HIV-1 in cDCs from elite controllers translated into stronger abilities to stimulate and expand HIV-1-specific CD8 T cell responses. These data suggest an important role of cell-intrinsic type I interferon secretion in dendritic cells for the induction of effective HIV-1-specific CD8 T cells, and may be helpful for eliciting functional T cell immunity against HIV-1 for preventative or therapeutic clinical purposes.
Collapse
Affiliation(s)
- Enrique Martin-Gayo
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Maria Jose Buzon
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Zhengyu Ouyang
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Taylor Hickman
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Jacqueline Cronin
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Dina Pimenova
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
41
|
Chaum E, Winborn CS, Bhattacharya S. Genomic regulation of senescence and innate immunity signaling in the retinal pigment epithelium. Mamm Genome 2015; 26:210-21. [PMID: 25963977 DOI: 10.1007/s00335-015-9568-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/02/2015] [Indexed: 01/04/2023]
Abstract
The tumor suppressor p53 is a major regulator of genes important for cell cycle arrest, senescence, apoptosis, and innate immunity, and has recently been implicated in retinal aging. In this study we sought to identify the genetic networks that regulate p53 function in the retina using quantitative trait locus (QTL) analysis. First we examined age-associated changes in the activation and expression levels of p53; known p53 target proteins and markers of innate immune system activation in primary retinal pigment epithelial (RPE) cells that were harvested from young and aged human donors. We observed increased expression of p53, activated caspase-1, CDKN1A, CDKN2A (p16INK4a), TLR4, and IFNα in aged primary RPE cell lines. We used the Hamilton Eye Institute (HEI) retinal dataset ( www.genenetwork.org ) to identify genomic loci that modulate expression of genes in the p53 pathway in recombinant inbred BXD mouse strains using a QTL systems biology-based approach. We identified a significant trans-QTL on chromosome 1 (region 172-177 Mb) that regulates the expression of Cdkn1a. Many of the genes in this QTL locus are involved in innate immune responses, including Fc receptors, interferon-inducible family genes, and formin 2. Importantly, we found an age-related increase in FCGR3A and FMN2 and a decrease in IFI16 levels in RPE cultures. There is a complex multigenic innate immunity locus that controls expression of genes in the p53 pathway in the RPE, which may play an important role in modulating age-related changes in the retina.
Collapse
Affiliation(s)
- Edward Chaum
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA,
| | | | | |
Collapse
|
42
|
Yu Q, Katlinskaya YV, Carbone CJ, Zhao B, Katlinski KV, Zheng H, Guha M, Li N, Chen Q, Yang T, Lengner CJ, Greenberg RA, Johnson FB, Fuchs SY. DNA-damage-induced type I interferon promotes senescence and inhibits stem cell function. Cell Rep 2015; 11:785-797. [PMID: 25921537 PMCID: PMC4426031 DOI: 10.1016/j.celrep.2015.03.069] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/18/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
Expression of type I interferons (IFNs) can be induced by DNA-damaging agents, but the mechanisms and significance of this regulation are not completely understood. We found that the transcription factor IRF3, activated in an ATM-IKKα/β-dependent manner, stimulates cell-autonomous IFN-β expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-β and stimulate IFN signaling in vitro and in vivo. In turn, IFN acts to amplify DNA-damage responses, activate the p53 pathway, promote senescence, and inhibit stem cell function in response to telomere shortening. Inactivation of the IFN pathway abrogates the development of diverse progeric phenotypes and extends the lifespan of Terc knockout mice. These data identify DNA-damage-response-induced IFN signaling as a critical mechanism that links accumulating DNA damage with senescence and premature aging.
Collapse
Affiliation(s)
- Qiujing Yu
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Yuliya V. Katlinskaya
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Christopher J. Carbone
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Bin Zhao
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Kanstantsin V. Katlinski
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Hui Zheng
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Manti Guha
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Ning Li
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Ting Yang
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Christopher J. Lengner
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Roger A. Greenberg
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - F. Brad Johnson
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| | - Serge Y. Fuchs
- Department of Animal Biology, School of Veterinary Medicine, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, 380 S. University Ave, Philadelphia, PA 19104, USA
| |
Collapse
|
43
|
Microbial DNA recognition by cGAS-STING and other sensors in dendritic cells in inflammatory bowel diseases. Inflamm Bowel Dis 2015; 21:901-11. [PMID: 25581829 DOI: 10.1097/mib.0000000000000299] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recognition of microbial nucleic acid initiates host immune defenses against pathogens. Impaired recognition of nucleic acid is involved in the pathogenesis of inflammatory bowel diseases. In contrast to the relatively well-established mechanism of microbial RNA sensing and associated signaling cascades, very little is known on how microbial DNA activates intracellular DNA sensors and controls the function of antigen-presenting cells (especially dendritic cells) to shape mucosal immune responses in intestine. In this review, we will introduce mucosal dendritic cell population, describe various putative DNA sensors, emphasize on newly identified cGAS-cGAMP-STING complex, and discuss how the detection of foreign DNA by mucosal dendritic cells activates innate and adaptive immune responses in intestine. Finally, we will identify certain inflammatory bowel disease-susceptibility genes that associate with impaired microbial DNA recognition in human.
Collapse
|
44
|
Ortiz-Fernández L, García-Lozano JR, Montes-Cano MA, Conde-Jaldón M, Ortego-Centeno N, García-Hernández FJ, Espinosa G, Graña-Gil G, Sánchez-Bursón J, Blanco R, Barnosi-Marín AC, Solans R, Fanlo P, Rodríguez-Carballeira M, Camps T, Castañeda S, Núñez-Roldán A, Martín J, González-Escribano MF. Variants of the IFI16 gene affecting the levels of expression of mRNA are associated with susceptibility to Behçet disease. J Rheumatol 2015; 42:695-701. [PMID: 25641891 DOI: 10.3899/jrheum.140949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Behçet disease (BD) is a multifactorial disease in which infectious agents have been proposed as triggers in genetically predisposed individuals. The aim of our study was to investigate the role of innate immunity receptors, specifically the nucleic acid sensors, in susceptibility to BD. METHODS Seventy-four tag single nucleotide polymorphisms (tSNP) selected in 9 candidate genes (DDX58, IFIH1, TLR3, TLR7, TLR8, AIM2, IFI16, ZBP1, and TLR9) were genotyped in 371 patients and 854 controls. Assays of mRNA expression and allele-specific transcript quantification (ASTQ) were performed in 110 and 50 controls, respectively. RESULTS Patients and controls were genotyped and 2 tSNP (rs6940 in IFI16 and rs855873 in AIM2) were associated with BD. To confirm this association, these tSNP were genotyped in 850 additional controls, and the total cohort was randomly divided into 2 cohorts. The association of these 2 tSNP with the disease remained in both cohorts. One haplotype (rs6940T-rs855873G) was identified as a risk factor (OR 1.41, 95% CI 1.06-1.86, p = 0.015), and another (rs6940A-rs855873A) as a protective factor (OR 0.65, 95% CI 0.47-0.90, p = 0.009). Samples with the risk haplotype had lower IFI16 expression levels than samples with the protective (0.99 ± 0.29 vs 1.23 ± 0.50, p = 0.022). Consistently, in the ASTQ assays performed with the nonsynonymous rs6940 SNP, the risk allele had lower IFI16 expression levels than the protective (p = 0.027). CONCLUSION Our findings suggest association of IFI16, a cytosolic sensor of dsDNA and mediator of the AIM2 inflammasome-dependent pathway, in susceptibility to BD. Differences genetically determined in the levels of this molecule could be the cause of this association.
Collapse
Affiliation(s)
- Lourdes Ortiz-Fernández
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - José-Raúl García-Lozano
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Marco-Antonio Montes-Cano
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Marta Conde-Jaldón
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Norberto Ortego-Centeno
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Francisco-José García-Hernández
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Gerard Espinosa
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Genaro Graña-Gil
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Juan Sánchez-Bursón
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Ricardo Blanco
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Ana-Celia Barnosi-Marín
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Roser Solans
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Patricia Fanlo
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Mónica Rodríguez-Carballeira
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Teresa Camps
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Santos Castañeda
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Antonio Núñez-Roldán
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - Javier Martín
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| | - María-Francisca González-Escribano
- From the Servicio de Inmunología, IBiS, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Sevilla; Servicio de Medicina Interna, Hospital Clínico San Cecilio, Granada; Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla; Servicio de Enfermedades Autoinmunes, Hospital Clinic, Barcelona; Servicio de Reumatología, Complejo Hospitalario Universitario, La Coruña; Servicio de Reumatología, Hospital Universitario de Valme, Sevilla; Servicio de Reumatología, Hospital Marqués de Valdecilla, Santander; Servicio de Medicina Interna, Hospital de Torrecárdenas, Almería; Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona; Servicio de Medicina Interna, Hospital Virgen del Camino, Pamplona; Servicio de Medicina Interna, Hospital Universitari Mútua, Terrassa; Servicio de Medicina Interna, Hospital Universitario Carlos Haya, Málaga; Servicio de Reumatología, Hospital de la Princesa, Madrid; Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.L. Ortíz-Fernández, BSc; J.R. García-Lozano, MD, PhD; M.A. Montes-Cano, PhD; M. Conde-Jaldón, BSc; A. Núñez-Roldán, MD, PhD; M.F. González-Escribano, PhD; Servicio de Inmunología, Hospital Universitario Virgen del Rocío; N. Ortego-Centeno, MD, Servicio de Medicina Interna, Hospital Clínico San Cecilio; F.J. García-Hernández, MD, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío; G. Espinosa, MD, Servicio de Enfermedades Autoinmunes, Hospital Clinic; G. Graña-Gil, MD, Servicio de Reumatología, Complejo Hospitalario Universitario; J. Sánchez-Bursón, MD, Servicio de Reumatología, Hospital Universitario de Valme; R. Blanco, MD, Servicio de Reumatología, Hospital Marqués de Valdecilla; A-C. Barnosi-Marín, MD, Servicio de Medicina Interna, Hospital de Torrecárdenas; R. Solans, MD, Servicio de Medicina Interna, Hospital Vall d'Hebron; P. Fanlo, MD, Servicio de Medi
| |
Collapse
|
45
|
Abstract
Alphaherpesviruses include human and animal pathogens, such as herpes simplex virus type 1, which establish life-long latent infections with episodes of recurrence. The immunocompetence of the infected host is an important determinant for the outcome of infections with alphaherpesviruses. Recognition of pathogen-associated molecular patterns by pattern recognition receptors is an essential, early step in the innate immune response to pathogens. In recent years, it has been discovered that herpesvirus DNA is a strong inducer of the innate immune system. The viral genome can be recognized in endosomes by TLR9, as well as intracellularly by a variety of DNA sensors, the best documented being cGAS, RNA Pol III, IFI16, and AIM2. These DNA sensors use converging signaling pathways to activate transcription factors, such as IRF3 and NF-κB, which induce the expression of type I interferons and other inflammatory cytokines and activate the inflammasome. This review summarizes the recent literature on the innate sensing of alphaherpesvirus DNA, the mechanisms of activation of the different sensors, their mechanisms of signal transduction, their physiological role in defense against herpesvirus infection, and how alphaherpesviruses seek to evade these responses to allow establishment and maintenance of infection.
Collapse
Affiliation(s)
- Stefanie Luecke
- Graduate School of Life Sciences, Universiteit Utrecht, Utrecht, The Netherlands
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Aarhus Research Center for Innate Immunology, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
46
|
Kigerl KA, de Rivero Vaccari JP, Dietrich WD, Popovich PG, Keane RW. Pattern recognition receptors and central nervous system repair. Exp Neurol 2014; 258:5-16. [PMID: 25017883 DOI: 10.1016/j.expneurol.2014.01.001] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/31/2013] [Accepted: 01/02/2014] [Indexed: 12/12/2022]
Abstract
Pattern recognition receptors (PRRs) are part of the innate immune response and were originally discovered for their role in recognizing pathogens by ligating specific pathogen associated molecular patterns (PAMPs) expressed by microbes. Now the role of PRRs in sterile inflammation is also appreciated, responding to endogenous stimuli referred to as "damage associated molecular patterns" (DAMPs) instead of PAMPs. The main families of PRRs include Toll-like receptors (TLRs), Nod-like receptors (NLRs), RIG-like receptors (RLRs), AIM2-like receptors (ALRs), and C-type lectin receptors. Broad expression of these PRRs in the CNS and the release of DAMPs in and around sites of injury suggest an important role for these receptor families in mediating post-injury inflammation. Considerable data now show that PRRs are among the first responders to CNS injury and activation of these receptors on microglia, neurons, and astrocytes triggers an innate immune response in the brain and spinal cord. Here we discuss how the various PRR families are activated and can influence injury and repair processes following CNS injury.
Collapse
Affiliation(s)
- Kristina A Kigerl
- Department of Neuroscience - Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, USA
| | | | - W Dalton Dietrich
- Department of Neurological Surgery - The Miami Project to Cure Paralysis, USA
| | - Phillip G Popovich
- Department of Neuroscience - Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, USA.
| | - Robert W Keane
- Department of Physiology & Biophysics - University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
47
|
Wang J, Alexander J, Wiebe M, Jones C. Bovine herpesvirus 1 productive infection stimulates inflammasome formation and caspase 1 activity. Virus Res 2014; 185:72-6. [PMID: 24657787 PMCID: PMC6240421 DOI: 10.1016/j.virusres.2014.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/05/2014] [Accepted: 03/05/2014] [Indexed: 10/25/2022]
Abstract
Bovine herpesvirus 1 (BoHV-1), a significant viral pathogen of cattle, causes inflammation in affected tissue during acute infection. Consequently, we tested whether productively infected bovine cells stimulate inflammasome formation. Expression of two components required for inflammasome formation, the DNA sensor IFI16 (gamma-interferon-inducible protein 16) and NLRP3 (NOD-like receptor family, pyrin domain containing 3), were induced in bovine kidney cells by eight hours after infection. IFI16 was detected in punctate granules localized to the cytoplasm and nucleus. During productive infection, more than ten times more cells were caspase 1 positive, which is activated following inflammasome formation. Two caspase 1 inhibitors had no effect on productive infection. Conversely, another caspase 1 inhibitor, glyburide, significantly inhibited virus infection suggesting it had off-target effects on related enzymes or interfered with infection via non-enzymatic mechanisms. Collectively, these studies demonstrated that BoHV-1 infection stimulated inflammasome formation, which we predict is important for clinical symptoms in cattle.
Collapse
Affiliation(s)
- Jianlin Wang
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Changcheng Road 700, Chengyang District, Qingdao 266109, PR China
| | - Jeff Alexander
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln Morisson Life Science Center, RM234, Lincoln, NE 68583-0900, United States
| | - Matthew Wiebe
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln Morisson Life Science Center, RM234, Lincoln, NE 68583-0900, United States
| | - Clinton Jones
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln Morisson Life Science Center, RM234, Lincoln, NE 68583-0900, United States.
| |
Collapse
|
48
|
Unterholzner L. The interferon response to intracellular DNA: why so many receptors? Immunobiology 2013; 218:1312-21. [PMID: 23962476 DOI: 10.1016/j.imbio.2013.07.007] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/11/2013] [Accepted: 07/17/2013] [Indexed: 12/22/2022]
Abstract
The detection of intracellular DNA has emerged to be a key event in the innate immune response to viruses and intracellular bacteria, and during conditions of sterile inflammation and autoimmunity. One of the consequences of the detection of DNA as a 'stranger' and a 'danger' signal is the production of type I interferons and pro-inflammatory cytokines. Much work has been dedicated to the elucidation of the signalling cascades that activate this DNA-induced gene expression programme. However, while many proteins have been proposed to act as sensors for intracellular DNA in recent years, none has been met with universal acceptance, and a theory linking all the recent observations is, as yet, lacking. This review presents the evidence for the various interferon-inducing DNA receptors proposed to date, and examines the hypotheses that might explain why so many different receptors appear to be involved in the innate immune recognition of intracellular DNA.
Collapse
Affiliation(s)
- Leonie Unterholzner
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, DD1 5EH, UK.
| |
Collapse
|
49
|
SORBS2 and TLR3 induce premature senescence in primary human fibroblasts and keratinocytes. BMC Cancer 2013; 13:507. [PMID: 24165198 PMCID: PMC3819711 DOI: 10.1186/1471-2407-13-507] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 09/19/2013] [Indexed: 01/05/2023] Open
Abstract
Background Genetic aberrations are required for the progression of HPV-induced cervical precancers. A prerequisite for clonal expansion of cancer cells is unlimited proliferative capacity. In a cell culture model for cervical carcinogenesis loss of genes located on chromosome 4q35→qter and chromosome 10p14-p15 were found to be associated with escape from senescence. Moreover, by LOH and I-FISH analyses a higher frequency of allele loss of these regions was also observed in cervical carcinomas as compared to CIN3. The aim of this study was to identify candidate senescence-related genes located on chromosome 4q35→qter and chromosome 10p14-p15 which may contribute to clonal expansion at the transition of CIN3 to cancer. Methods Microarray expression analyses were used to identify candidate genes down-regulated in cervical carcinomas as compared to CIN3. In order to relate these genes with the process of senescence their respective cDNAs were overexpressed in HPV16-immortalized keratinocytes as well as in primary human fibroblasts and keratinocytes using lentivirus mediated gene transduction. Results Overall fifteen genes located on chromosome 4q35→qter and chromosome 10p14-p15 were identified. Ten of these genes could be validated in biopsies by RT-PCR. Of interest is the novel finding that SORBS2 and TLR3 can induce senescence in primary human fibroblasts and keratinocytes but not in HPV-immortalized cell lines. Intriguingly, the endogenous expression of both genes increases during finite passaging of primary keratinocytes in vitro. Conclusions The relevance of the genes SORBS2 and TLR3 in the process of cellular senescence warrants further investigation. In ongoing experiments we are investigating whether this increase in gene expression is also characteristic of replicative senescence.
Collapse
|
50
|
Abstract
The oligonucleotide/oligosaccharide binding (OB) fold is employed by proteins to bind nucleic acids during replication, transcription, and translation. Recently, a variation of the OB fold consisting of a tandem pair of OB folds named the HIN (hematopoietic expression, interferon-inducible nature, and nuclear localization) domain was shown to play essential roles in the regulation of innate immune responses originating from binding of nucleic acids in the cytoplasm or the nucleus of the cell. Although the two OB folds of the HIN domain are linked via a long linker region, conserved hydrophobic contacts between the two OB folds hold them together firmly, resulting in a single compact domain. This overall topology of the HIN domain seems to be highly conserved, and proteins containing the HIN domain have been grouped in the PYHIN family. Structures of the recently solved HIN domains reveal that these domains exhibit either absent in melanoma2 (Aim2) HIN-like or p202 HINa-like modes of DNA binding. These two modes of DNA binding seem to result in different responses and as a consequence confer distinct roles on the proteins. This review summarizes our current understanding of the structure and function of the HIN domains in context with the innate immune responses.
Collapse
|