1
|
Holland M, Rutkowski R, C. Levin T. Evolutionary Dynamics of Proinflammatory Caspases in Primates and Rodents. Mol Biol Evol 2024; 41:msae220. [PMID: 39431598 PMCID: PMC11630849 DOI: 10.1093/molbev/msae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/05/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Caspase-1 and related proteases are key players in inflammation and innate immunity. Here, we characterize the evolutionary history of caspase-1 and its close relatives across 19 primates and 21 rodents, focusing on differences that may cause discrepancies between humans and animal studies. While caspase-1 has been retained in all these taxa, other members of the caspase-1 subfamily (caspase-4, caspase-5, caspase-11, and caspase-12 and CARD16, 17, and 18) each have unique evolutionary trajectories. Caspase-4 is found across simian primates, whereas we identified multiple pseudogenization and gene loss events in caspase-5, caspase-11, and the CARDs. Because caspase-4 and caspase-11 are both key players in the noncanonical inflammasome pathway, we expected that these proteins would be likely to evolve rapidly. Instead, we found that these two proteins are largely conserved, whereas caspase-4's close paralog, caspase-5, showed significant indications of positive selection, as did primate caspase-1. Caspase-12 is a nonfunctional pseudogene in humans. We find this extends across most primates, although many rodents and some primates retain an intact, and likely functional, caspase-12. In mouse laboratory lines, we found that 50% of common strains carry nonsynonymous variants that may impact the functions of caspase-11 and caspase-12 and therefore recommend specific strains to be used (and avoided). Finally, unlike rodents, primate caspases have undergone repeated rounds of gene conversion, duplication, and loss leading to a highly dynamic proinflammatory caspase repertoire. Thus, we uncovered many differences in the evolution of primate and rodent proinflammatory caspases and discuss the potential implications of this history for caspase gene functions.
Collapse
Affiliation(s)
- Mische Holland
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachel Rutkowski
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tera C. Levin
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Xuan R, Hu T, Cai L, Zhao B, Han E, Xia Z. CARD16 restores tumorigenesis and restraints apoptosis in glioma cells Via FOXO1/TRAIL axis. Cell Death Dis 2024; 15:804. [PMID: 39516471 PMCID: PMC11549220 DOI: 10.1038/s41419-024-07196-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
A hallmark of glioma cells, particularly glioblastoma multiforme (GBM) cells, is their resistance to apoptosis. Accumulating evidences has demonstrated that CARD16, a caspase recruitment domain (CARD) only protein, enhances both anti-apoptotic and tumorigenic properties. Nevertheless, there is a limited understanding of the expression and functional role of CARD16 in glioma. This study seeks to investigate, through in silico analysis and clinical specimens, the role of CARD16 as a potential tumor promoter in glioma. Functional assays and molecular studies revealed that CARD16 promotes tumorigenesis and suppresses apoptosis in glioma cells. Moreover, knockdown of CARD16 enhances the expression of the FOXO1/TRAIL axis in GBM cells. Additionally, FOXO1 downregulation in CARD16 knockdown GBM cells restores proliferation and reduces apoptosis. Further investigation demonstrated that elevated P21 expression inhibits CDK2-mediated FOXO1 phosphorylation and ubiquitination in CARD16-knockdown GBM cells. Collectively, these findings suggest that CARD16 is a tumor-promoting molecular in glioma via downregulating FOXO1/TRAIL axis, and suppressing TRAIL-induced apoptosis. The CARD16 gene presents significant potential for prognostic prediction and advances in innovative apoptotic therapeutics.
Collapse
Affiliation(s)
- Ruoheng Xuan
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tianyu Hu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lingshan Cai
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Beichuan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Erqiao Han
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhibo Xia
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Xu C, Jing W, Liu C, Yuan B, Zhang X, Liu L, Zhang F, Chen P, Liu Q, Wang H, Du X. Cytoplasmic DNA and AIM2 inflammasome in RA: where they come from and where they go? Front Immunol 2024; 15:1343325. [PMID: 39450183 PMCID: PMC11499118 DOI: 10.3389/fimmu.2024.1343325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease of undetermined etiology characterized by symmetric synovitis with predominantly destructive and multiple joint inflammation. Cytoplasmic DNA sensors that recognize protein molecules that are not themselves or abnormal dsDNA fragments play an integral role in the generation and perpetuation of autoimmune diseases by activating different signaling pathways and triggering innate immune signaling pathways and host defenses. Among them, melanoma deficiency factor 2 (AIM2) recognizes damaged DNA and double-stranded DNA and binds to them to further assemble inflammasome, initiating the innate immune response and participating in the pathophysiological process of rheumatoid arthritis. In this article, we review the research progress on the source of cytoplasmic DNA, the mechanism of assembly and activation of AIM2 inflammasome, and the related roles of other cytoplasmic DNA sensors in rheumatoid arthritis.
Collapse
Affiliation(s)
- Conghui Xu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Zheng's Acupuncture, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Fengfan Zhang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Qiang Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
4
|
Fetter T, de Graaf DM, Claus I, Wenzel J. Aberrant inflammasome activation as a driving force of human autoimmune skin disease. Front Immunol 2023; 14:1190388. [PMID: 37325658 PMCID: PMC10266227 DOI: 10.3389/fimmu.2023.1190388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Autoimmune skin diseases are understood as conditions in which the adaptive immune system with autoantigen-specific T cells and autoantibody-producing B cells reacting against self-tissues plays a crucial pathogenic role. However, there is increasing evidence that inflammasomes, which are large multiprotein complexes that were first described 20 years ago, contribute to autoimmune disease progression. The inflammasome and its contribution to the bioactivation of interleukins IL-1β and IL-18 play an essential role in combating foreign pathogens or tissue damage, but may also act as a pathogenic driver of myriad chronic inflammatory diseases when dysfunctionally regulated. Inflammasomes containing the NOD-like receptor family members NLRP1 and NLRP3 as well as the AIM2-like receptor family member AIM2 have been increasingly investigated in inflammatory skin conditions. In addition to autoinflammatory diseases, which are often associated with skin involvement, the aberrant activation of the inflammasome has also been implied in autoimmune diseases that can either affect the skin besides other organs such as systemic lupus erythematosus and systemic sclerosis or are isolated to the skin in humans. The latter include, among others, the T-cell mediated disorders vitiligo, alopecia areata, lichen planus and cutaneous lupus erythematosus as well as the autoantibody-driven blistering skin disease bullous pemphigoid. Some diseases are characterized by both autoinflammatory and autoimmune responses such as the chronic inflammatory skin disease psoriasis. Further insights into inflammasome dysregulation and associated pathways as well as their role in forming adaptive immune responses in human autoimmune skin pathology could potentially offer a new field of therapeutic options in the future.
Collapse
Affiliation(s)
- Tanja Fetter
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | | | - Isabelle Claus
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
5
|
Devi S, Indramohan M, Jäger E, Carriere J, Chu LH, de Almeida L, Greaves DR, Stehlik C, Dorfleutner A. CARD-only proteins regulate in vivo inflammasome responses and ameliorate gout. Cell Rep 2023; 42:112265. [PMID: 36930645 PMCID: PMC10151391 DOI: 10.1016/j.celrep.2023.112265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 01/10/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Inflammatory responses are crucial for controlling infections and initiating tissue repair. However, excessive and uncontrolled inflammation causes inflammatory disease. Processing and release of the pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18 depend on caspase-1 activation within inflammasomes. Assembly of inflammasomes is initiated upon activation of cytosolic pattern recognition receptors (PRRs), followed by sequential polymerization of pyrin domain (PYD)-containing and caspase recruitment domain (CARD)-containing proteins mediated by homotypic PYD and CARD interactions. Small PYD- or CARD-only proteins (POPs and COPs, respectively) evolved in higher primates to target these crucial interactions to limit inflammation. Here, we show the ability of COPs to regulate inflammasome activation by modulating homotypic CARD-CARD interactions in vitro and in vivo. CARD16, CARD17, and CARD18 displace crucial CARD interactions between caspase-1 proteins through competitive binding and ameliorate uric acid crystal-mediated NLRP3 inflammasome activation and inflammatory disease. COPs therefore represent an important family of inflammasome regulators and ameliorate inflammatory disease.
Collapse
Affiliation(s)
- Savita Devi
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mohanalaxmi Indramohan
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Elisabeth Jäger
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica Carriere
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lan H Chu
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Driskill Graduate Program in Life Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lucia de Almeida
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Christian Stehlik
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; The Kao Autoimmunity Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Andrea Dorfleutner
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; The Kao Autoimmunity Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
6
|
Bencze D, Fekete T, Pfliegler W, Szöőr Á, Csoma E, Szántó A, Tarr T, Bácsi A, Kemény L, Veréb Z, Pázmándi K. Interactions between the NLRP3-Dependent IL-1β and the Type I Interferon Pathways in Human Plasmacytoid Dendritic Cells. Int J Mol Sci 2022; 23:12154. [PMID: 36293012 PMCID: PMC9602791 DOI: 10.3390/ijms232012154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Generally, a reciprocal antagonistic interaction exists between the antiviral type I interferon (IFN) and the antibacterial nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3)-dependent IL-1β pathways that can significantly shape immune responses. Plasmacytoid dendritic cells (pDCs), as professional type I IFN-producing cells, are the major coordinators of antiviral immunity; however, their NLRP3-dependent IL-1β secretory pathway is poorly studied. Our aim was to determine the functional activity of the IL-1β pathway and its possible interaction with the type I IFN pathway in pDCs. We found that potent nuclear factor-kappa B (NF-κB) inducers promote higher levels of pro-IL-1β during priming compared to those activation signals, which mainly trigger interferon regulatory factor (IRF)-mediated type I IFN production. The generation of cleaved IL-1β requires certain secondary signals in pDCs and IFN-α or type I IFN-inducing viruses inhibit IL-1β production of pDCs, presumably by promoting the expression of various NLRP3 pathway inhibitors. In line with that, we detected significantly lower IL-1β production in pDCs of psoriasis patients with elevated IFN-α levels. Collectively, our results show that the NLRP3-dependent IL-1β secretory pathway is inducible in pDCs; however, it may only prevail under inflammatory conditions, in which the type I IFN pathway is not dominant.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Walter Pfliegler
- Department of Molecular Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eszter Csoma
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Antónia Szántó
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
7
|
Javaheri A, Diab A, Zhao L, Qian C, Cohen JB, Zamani P, Kumar A, Wang Z, Ebert C, Maranville J, Kvikstad E, Basso M, van Empel V, Richards AM, Doughty R, Rietzschell E, Kammerhoff K, Gogain J, Schafer P, Seiffert DA, Gordon DA, Ramirez-Valle F, Mann DL, Cappola TP, Chirinos JA. Proteomic Analysis of Effects of Spironolactone in Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2022; 15:e009693. [PMID: 36126144 PMCID: PMC9504263 DOI: 10.1161/circheartfailure.121.009693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The TOPCAT trial (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial) suggested clinical benefits of spironolactone treatment among patients with heart failure with preserved ejection fraction enrolled in the Americas. However, a comprehensive assessment of biologic pathways impacted by spironolactone therapy in heart failure with preserved ejection fraction has not been performed. METHODS We conducted aptamer-based proteomic analysis utilizing 5284 modified aptamers to 4928 unique proteins on plasma samples from TOPCAT participants from the Americas (n=164 subjects with paired samples at baseline and 1 year) to identify proteins and pathways impacted by spironolactone therapy in heart failure with preserved ejection fraction. Mean percentage change from baseline was calculated for each protein. Additionally, we conducted pathway analysis of proteins altered by spironolactone. RESULTS Spironolactone therapy was associated with proteome-wide significant changes in 7 proteins. Among these, CARD18 (caspase recruitment domain-containing protein 18), PKD2 (polycystin 2), and PSG2 (pregnancy-specific glycoprotein 2) were upregulated, whereas HGF (hepatic growth factor), PLTP (phospholipid transfer protein), IGF2R (insulin growth factor 2 receptor), and SWP70 (switch-associated protein 70) were downregulated. CARD18, a caspase-1 inhibitor, was the most upregulated protein by spironolactone (-0.5% with placebo versus +66.5% with spironolactone, P<0.0001). The top canonical pathways that were significantly associated with spironolactone were apelin signaling, stellate cell activation, glycoprotein 6 signaling, atherosclerosis signaling, liver X receptor activation, and farnesoid X receptor activation. Among the top pathways, collagens were a consistent theme that increased in patients receiving placebo but decreased in patients randomized to spironolactone. CONCLUSIONS Proteomic analysis in the TOPCAT trial revealed proteins and pathways altered by spironolactone, including the caspase inhibitor CARD18 and multiple pathways that involved collagens. In addition to effects on fibrosis, our studies suggest potential antiapoptotic effects of spironolactone in heart failure with preserved ejection fraction, a hypothesis that merits further exploration.
Collapse
Affiliation(s)
- Ali Javaheri
- Washington University School of Medicine, St. Louis, MO
| | - Ahmed Diab
- Washington University School of Medicine, St. Louis, MO
| | - Lei Zhao
- Bristol Myers Squibb Company, Lawrenceville, NJ
| | - Chenao Qian
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Jordana B. Cohen
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Payman Zamani
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Anupam Kumar
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | | | | | | | | | | | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Rob Doughty
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Ernst Rietzschell
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | - Thomas P. Cappola
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Julio A. Chirinos
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| |
Collapse
|
8
|
Lara-Reyna S, Caseley EA, Topping J, Rodrigues F, Jimenez Macias J, Lawler SE, McDermott MF. Inflammasome activation: from molecular mechanisms to autoinflammation. Clin Transl Immunology 2022; 11:e1404. [PMID: 35832835 PMCID: PMC9262628 DOI: 10.1002/cti2.1404] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammasomes are assembled by innate immune sensors that cells employ to detect a range of danger signals and respond with pro-inflammatory signalling. Inflammasomes activate inflammatory caspases, which trigger a cascade of molecular events with the potential to compromise cellular integrity and release the IL-1β and IL-18 pro-inflammatory cytokines. Several molecular mechanisms, working in concert, ensure that inflammasome activation is tightly regulated; these include NLRP3 post-translational modifications, ubiquitination and phosphorylation, as well as single-domain proteins that competitively bind to key inflammasome components, such as the CARD-only proteins (COPs) and PYD-only proteins (POPs). These diverse regulatory systems ensure that a suitable level of inflammation is initiated to counteract any cellular insult, while simultaneously preserving tissue architecture. When inflammasomes are aberrantly activated can drive excessive production of pro-inflammatory cytokines and cell death, leading to tissue damage. In several autoinflammatory conditions, inflammasomes are aberrantly activated with subsequent development of clinical features that reflect the degree of underlying tissue and organ damage. Several of the resulting disease complications may be successfully controlled by anti-inflammatory drugs and/or specific cytokine inhibitors, in addition to more recently developed small-molecule inhibitors. In this review, we will explore the molecular processes underlying the activation of several inflammasomes and highlight their role during health and disease. We also describe the detrimental effects of these inflammasome complexes, in some pathological conditions, and review current therapeutic approaches as well as future prospective treatments.
Collapse
Affiliation(s)
- Samuel Lara-Reyna
- Institute of Microbiology and Infection University of Birmingham Birmingham UK
| | - Emily A Caseley
- School of Biomedical Sciences, Faculty of Biological Sciences University of Leeds Leeds UK
| | - Joanne Topping
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital University of Leeds Leeds UK
| | - François Rodrigues
- AP-HP, Hôpital Tenon, Sorbonne Université, Service de Médecine interne Centre de Référence des Maladies Auto-inflammatoires et des Amyloses d'origine inflammatoire (CEREMAIA) Paris France
| | - Jorge Jimenez Macias
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA.,Brown Cancer Centre, Department of Pathology and Laboratory Medicine Brown University Providence Rhode Island USA
| | - Sean E Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA.,Brown Cancer Centre, Department of Pathology and Laboratory Medicine Brown University Providence Rhode Island USA
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital University of Leeds Leeds UK
| |
Collapse
|
9
|
Anti-Inflammatory and Active Biological Properties of the Plant-Derived Bioactive Compounds Luteolin and Luteolin 7-Glucoside. Nutrients 2022; 14:nu14061155. [PMID: 35334812 PMCID: PMC8949538 DOI: 10.3390/nu14061155] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 12/14/2022] Open
Abstract
Flavonoids are interesting molecules synthetized by plants. They can be found abundantly in seeds and fruits, determining the color, flavor, and other organoleptic characteristics, as well as contributing to important nutritional aspects. Beyond these characteristics, due to their biochemical properties and characteristics, they can be considered bioactive compounds. Several interesting studies have demonstrated their biological activity in different cellular and physiological processes in high-order organisms including humans. The flavonoid molecular structure confers the capability of reacting with and neutralizing reactive oxygen species (ROS), behaving as scavengers in all processes generating this class of molecules, such as UV irradiation, a process widely present in plant physiology. Importantly, the recent scientific literature has demonstrated that flavonoids, in human physiology, are active compounds acting not only as scavengers but also with the important role of counteracting the inflammation process. Among the wide variety of flavonoid molecules, significant results have been shown by investigating the role of the flavones luteolin and luteolin-7-O-glucoside (LUT-7G). For these compounds, experimental results demonstrated an interesting anti-inflammatory action, both in vitro and in vivo, in the interaction with JAK/STAT3, NF-κB, and other pathways described in this review. We also describe the effects in metabolic pathways connected with inflammation, such as cellular glycolysis, diabetes, lipid peroxidation, and effects in cancer cells. Moreover, the inhibition of inflammatory pathway in endothelial tissue, as well as the NLRP3 inflammasome assembly, demonstrates a key role in the progression of such phenomena. Since these micronutrient molecules can be obtained from food, their biochemical properties open new perspectives with respect to the long-term health status of healthy individuals, as well as their use as a coadjutant treatment in specific diseases.
Collapse
|
10
|
Bombaci G, Sarangdhar MA, Andina N, Tardivel A, Yu ECW, Mackie GM, Pugh M, Ozan VB, Banz Y, Spinetti T, Hirzel C, Youd E, Schefold JC, Taylor G, Gazdhar A, Bonadies N, Angelillo-Scherrer A, Schneider P, Maslowski KM, Allam R. LRR-protein RNH1 dampens the inflammasome activation and is associated with COVID-19 severity. Life Sci Alliance 2022; 5:5/6/e202101226. [PMID: 35256513 PMCID: PMC8922048 DOI: 10.26508/lsa.202101226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
RNH1 prevents inflammation by inhibiting inflammasome activation through controlling caspase-1 protein levels. In COVID-19 patients, RNH1 expression levels were negatively associated with disease severity and inflammation, suggesting a role for RNH1 in SARS-CoV-2–mediated inflammation and pathology. Inflammasomes are cytosolic innate immune sensors of pathogen infection and cellular damage that induce caspase-1–mediated inflammation upon activation. Although inflammation is protective, uncontrolled excessive inflammation can cause inflammatory diseases and can be detrimental, such as in coronavirus disease (COVID-19). However, the underlying mechanisms that control inflammasome activation are incompletely understood. Here we report that the leucine-rich repeat (LRR) protein ribonuclease inhibitor (RNH1), which shares homology with LRRs of NLRP (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing) proteins, attenuates inflammasome activation. Deletion of RNH1 in macrophages increases interleukin (IL)-1β production and caspase-1 activation in response to inflammasome stimulation. Mechanistically, RNH1 decreases pro-IL-1β expression and induces proteasome-mediated caspase-1 degradation. Corroborating this, mouse models of monosodium urate (MSU)-induced peritonitis and lipopolysaccharide (LPS)-induced endotoxemia, which are dependent on caspase-1, respectively, show increased neutrophil infiltration and lethality in Rnh1−/− mice compared with wild-type mice. Furthermore, RNH1 protein levels were negatively related with disease severity and inflammation in hospitalized COVID-19 patients. We propose that RNH1 is a new inflammasome regulator with relevance to COVID-19 severity.
Collapse
Affiliation(s)
- Giuseppe Bombaci
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Mayuresh Anant Sarangdhar
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Nicola Andina
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Aubry Tardivel
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Eric Chi-Wang Yu
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Gillian M Mackie
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Matthew Pugh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Vedat Burak Ozan
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thibaud Spinetti
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cedric Hirzel
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Esther Youd
- School of Medicine, Dentistry and Nursing, Forensic Medicine and Science. University of Glasgow, Scotland, UK
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Amiq Gazdhar
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolas Bonadies
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Anne Angelillo-Scherrer
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Kendle M Maslowski
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Ramanjaneyulu Allam
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
The Psoriatic Nonlesional Skin: A Battlefield between Susceptibility and Protective Factors. J Invest Dermatol 2021; 141:2785-2790. [PMID: 34216605 DOI: 10.1016/j.jid.2021.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/31/2022]
Abstract
In the last two decades, large-scale gene-expression studies on psoriatic skin samples revealed that even though nonlesional skin is macroscopically identical to healthy skin, it harbors several molecular differences. Originally, these molecular differences were thought to represent susceptibility factors for plaque formation. However, we review in this paper the several factors of immune regulation and structural alteration that are specific for the nonlesional skin and serve as protective factors by counteracting plaque formation and contributing to the maintenance of the nonlesional phenotype.
Collapse
|
12
|
Péladeau C, Sandhu JK. Aberrant NLRP3 Inflammasome Activation Ignites the Fire of Inflammation in Neuromuscular Diseases. Int J Mol Sci 2021; 22:ijms22116068. [PMID: 34199845 PMCID: PMC8200055 DOI: 10.3390/ijms22116068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are molecular hubs that are assembled and activated by a host in response to various microbial and non-microbial stimuli and play a pivotal role in maintaining tissue homeostasis. The NLRP3 is a highly promiscuous inflammasome that is activated by a wide variety of sterile triggers, including misfolded protein aggregates, and drives chronic inflammation via caspase-1-mediated proteolytic cleavage and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. These cytokines further amplify inflammatory responses by activating various signaling cascades, leading to the recruitment of immune cells and overproduction of proinflammatory cytokines and chemokines, resulting in a vicious cycle of chronic inflammation and tissue damage. Neuromuscular diseases are a heterogeneous group of muscle disorders that involve injury or dysfunction of peripheral nerves, neuromuscular junctions and muscles. A growing body of evidence suggests that dysregulation, impairment or aberrant NLRP3 inflammasome signaling leads to the initiation and exacerbation of pathological processes associated with neuromuscular diseases. In this review, we summarize the available knowledge about the NLRP3 inflammasome in neuromuscular diseases that affect the peripheral nervous system and amyotrophic lateral sclerosis, which affects the central nervous system. In addition, we also examine whether therapeutic targeting of the NLRP3 inflammasome components is a viable approach to alleviating the detrimental phenotype of neuromuscular diseases and improving clinical outcomes.
Collapse
Affiliation(s)
- Christine Péladeau
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
| | - Jagdeep K. Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Correspondence: ; Tel.: +1-613-993-5304
| |
Collapse
|
13
|
Gil-Jaramillo N, Rocha AP, Raiol T, Motta FN, Favali C, Brigido MM, Bastos IMD, Santana JM. The First Contact of Human Dendritic Cells With Trypanosoma cruzi Reveals Response to Virus as an Unexplored Central Pathway. Front Immunol 2021; 12:638020. [PMID: 33897690 PMCID: PMC8062726 DOI: 10.3389/fimmu.2021.638020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Chagas disease is a debilitating and neglected disease caused by the protozoan Trypanosoma cruzi. Soon after infection, interactions among T. cruzi and host innate immunity cells can drive/contribute to disease outcome. Dendritic cells (DCs), present in all tissues, are one of the first immune cells to interact with Trypanosoma cruzi metacyclic trypomastigotes. Elucidating the immunological events triggered immediately after parasite-human DCs encounter may aid in understanding the role of DCs in the establishment of infection and in the course of the disease. Therefore, we performed a transcriptomic analysis of a 12 h interaction between T. cruzi and MoDCs (monocyte-derived DCs) from three human donors. Enrichment analyses of the 468 differentially expressed genes (DEGs) revealed viral infection response as the most regulated pathway. Additionally, exogenous antigen processing and presentation through MHC-I, chemokine signaling, lymphocyte co-stimulation, metallothioneins, and inflammasome activation were found up-regulated. Notable, we were able to identify the increased gene expression of alternative inflammasome sensors such as AIM2, IFI16, and RIG-I for the first time in a T. cruzi infection. Both transcript and protein expression levels suggest proinflammatory cytokine production during early T. cruzi-DCs contact. Our transcriptome data unveil antiviral pathways as an unexplored process during T. cruzi-DC initial interaction, disclosing a new panorama for the study of Chagas disease outcomes.
Collapse
Affiliation(s)
- Natalia Gil-Jaramillo
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Amanda Pereira Rocha
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Tainá Raiol
- Fiocruz Brasília–Gerência Regional de Brasília (GEREB), Fundação Oswaldo Cruz (Fiocruz), Brasília, Brazil
| | - Flávia Nader Motta
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
- Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | - Cecília Favali
- Laboratório de Imunologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Marcelo M. Brigido
- Laboratório de Imunologia Molecular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Izabela M. D. Bastos
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Jaime M. Santana
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
14
|
Tyrkalska SD, Candel S, Mulero V. The neutrophil inflammasome. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103874. [PMID: 32987011 DOI: 10.1016/j.dci.2020.103874] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Since inflammasomes were discovered in the early 21st century, knowledge about their biology has multiplied exponentially. These cytosolic multiprotein complexes alert the immune system about the presence of infection or tissue damage, and regulate the subsequent inflammatory responses. As inflammasome dysregulation is increasingly associated with numerous autoinflammatory disorders, there is an urgent need for further research into the inflammasome's involvement in the pathogenesis of such diseases in order to identify novel therapeutic targets and treatments. The zebrafish has become a widely used animal model to study human diseases in recent years, and has already provided relevant findings in the field of inflammasome biology including the identification of new components and pathways. We provide a detailed analysis of current knowledge on neutrophil inflammasome biology and compare its features with those of the better known macrophage inflammasome, focusing on its contribution to innate immunity and its relevance for human health. Importantly, a large body of evidence points to a link between neutrophil inflammasome dysfunction and many neutrophil-mediated human diseases, but the real contribution of the neutrophil inflammasome to the pathogenesis of these disorders is largely unknown. Although neutrophils have remained in the shadow of macrophages and monocytes in the field of inflammasome research since the discovery of these multiprotein platforms, recent studies strongly suggest that the importance of the neutrophil inflammasome has been underestimated.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain.
| | - Sergio Candel
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain.
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| |
Collapse
|
15
|
Structure, Activation and Regulation of NLRP3 and AIM2 Inflammasomes. Int J Mol Sci 2021; 22:ijms22020872. [PMID: 33467177 PMCID: PMC7830601 DOI: 10.3390/ijms22020872] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
The inflammasome is a three-component (sensor, adaptor, and effector) filamentous signaling platform that shields from multiple pathogenic infections by stimulating the proteolytical maturation of proinflammatory cytokines and pyroptotic cell death. The signaling process initiates with the detection of endogenous and/or external danger signals by specific sensors, followed by the nucleation and polymerization from sensor to downstream adaptor and then to the effector, caspase-1. Aberrant activation of inflammasomes promotes autoinflammatory diseases, cancer, neurodegeneration, and cardiometabolic disorders. Therefore, an equitable level of regulation is required to maintain the equilibrium between inflammasome activation and inhibition. Recent advancement in the structural and mechanistic understanding of inflammasome assembly potentiates the emergence of novel therapeutics against inflammasome-regulated diseases. In this review, we have comprehensively discussed the recent and updated insights into the structure of inflammasome components, their activation, interaction, mechanism of regulation, and finally, the formation of densely packed filamentous inflammasome complex that exists as micron-sized punctum in the cells and mediates the immune responses.
Collapse
|
16
|
Aral K, Milward MR, Cooper PR. Inflammasome dysregulation in human gingival fibroblasts in response to periodontal pathogens. Oral Dis 2020; 28:216-224. [PMID: 33368813 DOI: 10.1111/odi.13760] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Uncontrolled production of Interleukin-1β (IL-1β), a major proinflammatory cytokine, is associated with tissue destruction in periodontal disease. IL-1β production is controlled by inflammasomes which are multiprotein regulatory complexes. The current study aimed to elucidate potential regulatory pathways by monitoring the effects of periodontal pathogens Fusobacterium nucleatum (Fn) and Porphyromonas gingivalis (Pg) on inflammasomes and their regulators in human gingival fibroblasts (HGFs) in vitro. METHODS HGFs were exposed to Fn and Pg alone or in combination for 24 hr at a multiplicity of infection of 100, ±30 min exposure with 5 mM adenosine triphosphate (ATP) incubation. Gene expression of NLRP3 and AIM2, inflammasome regulatory proteins POP1, CARD16 and TRIM16, and inflammasome components ASC and CASPASE 1, and IL-1β, were evaluated by RT-PCR. Pro- and mature IL-1β levels were monitored intracellularly by immunocytochemistry and extracellularly by ELISA. RESULTS Fn + ATP significantly upregulated NLRP3, AIM2, IL-1β, ASC, and CASPASE 1; however, it downregulated POP1 and TRIM16. Pg + ATP downregulated NLRP3, ASC, POP1, but upregulated IL-1β and CARD16. Pg + Fn+ATP significantly upregulated AIM2, IL-1β and CARD16, and downregulated POP1, TRIM16, and CASPASE 1. Pg + ATP exposure significantly increased pro- and mature IL-1β production. CONCLUSION Bacterial exposure with ATP may deregulate IL-1β by dysregulating inflammasomes and their regulators in HGFs.
Collapse
Affiliation(s)
- Kübra Aral
- School of Dentistry, University of Birmingham, Birmingham, UK.,Republic of Turkey Ministry of Health, Uskudar Ahmet Yuksel Ozemre Oral and Dental Health Center, Istanbul, Turkey
| | | | - Paul R Cooper
- School of Dentistry, University of Birmingham, Birmingham, UK.,Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
17
|
Devi S, Stehlik C, Dorfleutner A. An Update on CARD Only Proteins (COPs) and PYD Only Proteins (POPs) as Inflammasome Regulators. Int J Mol Sci 2020; 21:E6901. [PMID: 32962268 PMCID: PMC7555848 DOI: 10.3390/ijms21186901] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammasomes are protein scaffolds required for the activation of caspase-1 and the subsequent release of interleukin (IL)-1β, IL-18, and danger signals, as well as the induction of pyroptotic cell death to restore homeostasis following infection and sterile tissue damage. However, excessive inflammasome activation also causes detrimental inflammatory disease. Therefore, extensive control mechanisms are necessary to prevent improper inflammasome responses and inflammatory disease. Inflammasomes are assembled by sequential nucleated polymerization of Pyrin domain (PYD) and caspase recruitment domain (CARD)-containing inflammasome components. Once polymerization is nucleated, this process proceeds in a self-perpetuating manner and represents a point of no return. Therefore, regulation of this key step is crucial for a controlled inflammasome response. Here, we provide an update on two single domain protein families containing either a PYD or a CARD, the PYD-only proteins (POPs) and CARD-only proteins (COPs), respectively. Their structure allows them to occupy and block access to key protein-protein interaction domains necessary for inflammasome assembly, thereby regulating the threshold of these nucleated polymerization events, and consequently, the inflammatory host response.
Collapse
Affiliation(s)
- Savita Devi
- Department of Pathology and Laboratory Medicine, Cedars Sinai, Los Angeles, CA 90048, USA;
| | - Christian Stehlik
- Department of Pathology and Laboratory Medicine, Cedars Sinai, Los Angeles, CA 90048, USA;
- Department of Biomedical Sciences, and Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai, Los Angeles, CA 90048, USA
| | - Andrea Dorfleutner
- Department of Pathology and Laboratory Medicine, Cedars Sinai, Los Angeles, CA 90048, USA;
- Department of Biomedical Sciences, Cedars Sinai, Los Angeles, CA 90048, USA
| |
Collapse
|
18
|
McKee CM, Coll RC. NLRP3 inflammasome priming: A riddle wrapped in a mystery inside an enigma. J Leukoc Biol 2020; 108:937-952. [PMID: 32745339 DOI: 10.1002/jlb.3mr0720-513r] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
The NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome is an immunological sensor that detects a wide range of microbial- and host-derived signals. Inflammasome activation results in the release of the potent pro-inflammatory cytokines IL-1β and IL-18 and triggers a form of inflammatory cell death known as pyroptosis. Excessive NLRP3 activity is associated with the pathogenesis of a wide range of inflammatory diseases, thus NLRP3 activation mechanisms are an area of intensive research. NLRP3 inflammasome activation is a tightly regulated process that requires both priming and activation signals. In particular, recent research has highlighted the highly complex nature of the priming step, which involves transcriptional and posttranslational mechanisms, and numerous protein binding partners. This review will describe the current understanding of NLRP3 priming and will discuss the potential opportunities for targeting this process therapeutically to treat NLRP3-associated diseases.
Collapse
Affiliation(s)
- Chloe M McKee
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Antrim, UK
| | - Rebecca C Coll
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Antrim, UK
| |
Collapse
|
19
|
Kumari P, Russo AJ, Shivcharan S, Rathinam VA. AIM2 in health and disease: Inflammasome and beyond. Immunol Rev 2020; 297:83-95. [PMID: 32713036 DOI: 10.1111/imr.12903] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022]
Abstract
Nucleic acid sensing is a critical mechanism by which the immune system monitors for pathogen invasion. A set of germline-encoded innate immune receptors detect microbial DNA in various compartments of the cell, such as endosomes, the cytosol, and the nucleus. Sensing of microbial DNA through these receptors stimulates, in most cases, interferon regulatory factor-dependent type I IFN synthesis followed by JAK/STAT-dependent interferon-stimulated gene expression. In contrast, the detection of DNA in the cytosol by AIM2 assembles a macromolecular complex called the inflammasome, which unleashes the proteolytic activity of a cysteine protease caspase-1. Caspase-1 cleaves and activates the pro-inflammatory cytokines such as IL-1β and IL-18 and a pore-forming protein, gasdermin D, which triggers pyroptosis, an inflammatory form of cell death. Research over the past decade has revealed that AIM2 plays essential roles not only in host defense against pathogens but also in inflammatory diseases, autoimmunity, and cancer in inflammasome-dependent and inflammasome-independent manners. This review discusses the latest advancements in our understanding of AIM2 biology and its functions in health and disease.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Sonia Shivcharan
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| |
Collapse
|
20
|
Wang B, Bhattacharya M, Roy S, Tian Y, Yin Q. Immunobiology and structural biology of AIM2 inflammasome. Mol Aspects Med 2020; 76:100869. [PMID: 32660715 DOI: 10.1016/j.mam.2020.100869] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
Absent in melanoma 2 (AIM2) is a cytoplasmic sensor that upon recognizing double-stranded DNA assembles with apoptosis-associated speck-like protein containing a CARD (ASC) and procaspase-1 to form the multi-protein complex AIM2 inflammasome. Double-stranded DNA from bacterial, viral, or host cellular origins triggers AIM2 inflammasome assembly and activation, ultimately resulting in secretion of proinflammatory cytokines and pyroptotic cell death in order to eliminate microbial infection. Many pathogens therefore evade or suppress AIM2 inflammasome to establish infection. On the other hand, AIM2 activation is tightly controlled by multiple cellular factors to prevent autoinflammation. Extensive structural studies have captured the molecular details of multiple steps in AIM2 inflammasome assembly. The structures collectively revealed a nucleated polymerization mechanism that not only pervades each step of AIM2 inflammasome assembly, but also underlies assembly of other inflammasomes and complexes in immune signaling. In this article, we briefly review the identification of AIM2 as a cytoplasmic DNA sensor, summarize the importance of AIM2 inflammasome in infections and diseases, and discuss the molecular mechanisms of AIM2 assembly, activation, and regulation using recent cellular, biochemical, and structural results.
Collapse
Affiliation(s)
- Bing Wang
- Department of Biological Science, Florida State University, Tallahassee, FL, 32301, USA
| | - Madhurima Bhattacharya
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32301, USA
| | - Sayantan Roy
- Department of Biological Science, Florida State University, Tallahassee, FL, 32301, USA
| | - Yuan Tian
- Department of Biological Science, Florida State University, Tallahassee, FL, 32301, USA
| | - Qian Yin
- Department of Biological Science, Florida State University, Tallahassee, FL, 32301, USA; Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32301, USA.
| |
Collapse
|
21
|
Aral K, Milward MR, Gupta D, Cooper PR. Effects of Porphyromonas gingivalis and Fusobacterium nucleatum on inflammasomes and their regulators in H400 cells. Mol Oral Microbiol 2020; 35:158-167. [PMID: 32516848 DOI: 10.1111/omi.12302] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Inflammasomes are multiprotein complexes that regulate immune processes in response to infections and tissue damage. They modulate Interleukin-1beta (IL-1β) expression, a major proinflammatory cytokine. The inflammasome/IL-1β pathway is involved in head and neck squamous cell carcinoma (HNSCC) progression and the periodontal pathogens Fusobacterium nucleatum (Fn) and Porphyromonas gingivalis (Pg) have been reported to cause chronic inflammation in HNSCC. The aim of this study was to characterise the role of these pathogens in regulating inflammasome activity and the IL-1β response in HNSCC in vitro. METHODS An HNSCC cell line (H400) was exposed to Fn and Pg individually or in combination for 24h, ± incubation for 30 min with 5 mM adenosine triphosphate (ATP). Transcript levels of inflammasomes, NLRP3 and AIM2; inflammasome-regulatory proteins, POP1, CARD16 and TRIM16; and inflammasome-component, ASC and caspase 1 and IL-1β, were assayed by RT-PCR. Expression of IL-1β was by immunocytochemistry and ELISA. RESULTS NLRP3 expression was significantly upregulated in response to Pg, Fn + Pg, Pg + ATP and Fn + Pg + ATP. AIM2 was significantly upregulated by Fn, Pg and Fn + Pg + ATP exposure. All conditions significantly upregulated IL-1β gene expression. POP1 expression was significantly downregulated by Pg or Fn exposure but not by Fn + Pg. Intracellular pro- and mature IL-1β were significantly higher following Fn and Pg + ATP exposure. CONCLUSION Pg alone increased IL-1β by upregulating AIM2, NLRP3 and downregulating POP1. Fn promoted IL-1β by increasing AIM2 and downregulating POP1. Pg + ATP with or without Fn upregulated NLRP3, IL-1β by downregulating POP1. Periodontal pathogens may contribute to HNSCC pathogenesis by increasing the IL-1β response due to inflammasome dysregulation.
Collapse
Affiliation(s)
- Kübra Aral
- Research Fellow, School of Dentistry, University of Birmingham, Birmingham, UK.,Republic of Turkey Ministry of Health, Ankara, Turkey
| | | | - Dhanak Gupta
- Research Fellow, School of Dentistry, University of Birmingham, Birmingham, UK
| | - Paul R Cooper
- School of Dentistry, University of Birmingham, Birmingham, UK.,Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
Reichl FX, Högg C, Liu F, Schwarz M, Teupser D, Hickel R, Bloch W, Schweikl H, Thomas P, Summer B. Actovegin® reduces PMA-induced inflammation on human cells. Eur J Appl Physiol 2020; 120:1671-1680. [PMID: 32447451 PMCID: PMC8497287 DOI: 10.1007/s00421-020-04398-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/16/2020] [Indexed: 01/06/2023]
Abstract
Purpose The effect of Actovegin® was investigated on PMA- and LPS-induced human peripheral blood mononuclear cells (PBMCs). Methods PBMCs (1 × 106 cells/ml) from five blood donors (2 f, 3 m; 45–55 years) were grown in medium and exposed to Actovegin® in the presence or absence of PMA or LPS. Supernatants were collected to assess the concentration of cytokines (TNF-α, IL-1beta, IL-6 and IL-10). The reactive oxygen species (ROS) were assessed by a ROS-GloTM H2O2 assay. Results Stimulation of cells by PMA or LPS (without Actovegin®) significantly increased the secretion of IL-1beta, IL-6, IL-10 and TNF-α from PBMCs, compared to controls. Pre-treatment of cells with Actovegin® (1, 5, 25, 125 µg/ml) plus PMA significantly decreased the secretion of IL-1beta from PBMCs, compared to controls (PMA without Actovegin®). In contrast, addition of Actovegin® (1, 5, 25, 125 and 250 µg/ml) plus LPS did not alter the IL-1beta production, compared to controls (LPS without Actovegin®). TNF-α, IL-6 and IL-10 do not contribute to the reduction of inflammatory reactions with Actovegin®. Conclusions Actovegin® can reduce the PMA-induced IL-1beta release and the ROS production from PBMCs. These findings may help to explain the clinically known positive effects of Actovegin® on athletic injuries with inflammatory responses (e.g., muscle injuries, tendinopathies).
Collapse
Affiliation(s)
- Franz-Xaver Reichl
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Goethestr. 70, 80336, Munich, Germany.
| | - Christof Högg
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Goethestr. 70, 80336, Munich, Germany
| | - Fangfang Liu
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Goethestr. 70, 80336, Munich, Germany
| | - Markus Schwarz
- Institute for Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- Institute for Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Reinhard Hickel
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Goethestr. 70, 80336, Munich, Germany
| | - Wilhelm Bloch
- Molecular and Cellular Sport Medicine, German Sport University, Cologne, Germany
| | - Helmut Schweikl
- Department of Conservative Dentistry and Periodontology, University Hospital, Regensburg, Germany
| | - Peter Thomas
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Burkhard Summer
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
23
|
Aral K, Milward MR, Kapila Y, Berdeli A, Cooper PR. Inflammasomes and their regulation in periodontal disease: A review. J Periodontal Res 2020; 55:473-487. [PMID: 31960443 DOI: 10.1111/jre.12733] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/15/2019] [Accepted: 12/28/2019] [Indexed: 12/12/2022]
Abstract
Interleukin-1β (IL-1β), which is secreted by host tissues leading to periodontal tissue inflammation, is a major pro-inflammatory cytokine in the pathogenesis of periodontal disease. The conversion of pro-IL-1β into its biologically active form is controlled by multiprotein complexes named as inflammasomes, which are key regulator of host defense mechanisms and inflammasome involved diseases, including the periodontal diseases. Inflammasomes are regulated by different proteins and processes, including pyrin domain (PYD)-only proteins (POPs), CARD-only proteins (COPs), tripartite motif family proteins (TRIMs), autophagy, and interferons. A review of in vitro, in vivo, and clinical data from these publications revealed that several inflammasomes including (NOD)-like receptor (NLR) pyrin domain-containing 3 (NLRP3) and absent in melanoma 2 (AIM2) have been found to be involved in periodontal disease pathogenesis. To the best of our knowledge, the current article provides the first review of the literature focusing on studies that evaluated both inflammasomes and their regulators in periodontal disease. An upregulation for inflammasomes and a downregulation of inflammasome regulator proteins including POPs, COPs, and TRIMs have been reported in periodontal disease. Although interferons (types I and II) and autophagy have been found to be involved in periodontal disease, their possible role in inflammasome activation has not evaluated yet. Modulating the excessive inflammatory response by the use of inflammasome regulators may have potential in the management of periodontal disease.
Collapse
Affiliation(s)
- Kübra Aral
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK.,Republic of Turkey Ministry of Health, Ankara, Turkey
| | - Michael R Milward
- Periodontology, School of Dentistry, University of Birmingham, Birmingham, UK
| | - Yvonne Kapila
- Orofacial Sciences, The School of Dentistry, University of California San Francisco, San Francisco, CA, USA
| | - Afig Berdeli
- Molecular Genetics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Paul R Cooper
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK.,Department of Oral Sciences, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
24
|
Aral K, Berdeli E, Cooper PR, Milward MR, Kapila Y, Karadede Ünal B, Aral CA, Berdeli A. Differential expression of inflammasome regulatory transcripts in periodontal disease. J Periodontol 2019; 91:606-616. [PMID: 31557327 DOI: 10.1002/jper.19-0222] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/12/2019] [Accepted: 07/30/2019] [Indexed: 11/08/2022]
Abstract
BACKGROUND The inflammasome modulates the release of key proinflammatory cytokines associated with periodontal disease pathogenesis. The aim of this study was to evaluate the expression of proteins that regulate the inflammasome, namely pyrin domain-only proteins (POPs), caspase activation recruitment domain (CARD)-only proteins, and tripartite motif-containing (TRIM) proteins, in periodontal diseases. METHODS A total of 68 participants (34 males and 34 females) were divided into four groups, including periodontal health (H), gingivitis (G), chronic periodontitis (CP), and aggressive periodontitis (AgP) based on clinical parameters. Gingival tissue samples were obtained from all participants for reverse transcription polymerase chain reaction (RT-PCR)-based gene expression analyses of molecules that regulate the inflammasome, including apoptosis-associated speck-like protein (ASC) containing CARD, caspase-1, interleukin-1β (IL-1β), interleukin-18 (IL-18), nucleotide-binding domain, leucine rich family (NLR) pyrin domain containing 3 (NLRP3), NLR family pyrin domain containing 2 (NLRP2), AIM2 (absent in melanoma 2), POP1, POP2, CARD16, CARD18, TRIM16, and TRIM20 by RT-PCR. RESULTS NLRP3 and IL-1β were upregulated in the G, CP, and AgP groups compared with group H (P < 0.05). AIM2 was downregulated in the CP group compared with the H, G, and AgP groups (P < 0.05). TRIM20, TRIM16, and CARD18 were downregulated in the G, CP, and AgP groups compared with the H group (P < 0.05). POP1 and POP2 were downregulated in the CP and AgP, and AgP and G groups, respectively (P < 0.05). CONCLUSION Active periodontal disease may result in downregulation of inflammasome regulators that may increase the activity of NLRP3 and IL-1β in periodontal disease.
Collapse
Affiliation(s)
- Kübra Aral
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK
| | - Eynar Berdeli
- Faculty of Dentistry, Izmir Katip Celebi University, Izmir, Turkey
| | - Paul Roy Cooper
- School of Dentistry, University of Birmingham, Birmingham, UK
| | | | - Yvonne Kapila
- The School of Dentistry, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Afig Berdeli
- Faculty of Medicine, Molecular Medicine Laboratory, Ege University, Izmir, Turkey
| |
Collapse
|
25
|
Abstract
The inflammasome is a multi-molecular platform crucial to the induction of an inflammatory response to cellular danger. Recognition in the cytoplasm of endogenously and exogenously derived ligands initiates conformational change in sensor proteins, such as NLRP3, that permits the subsequent rapid recruitment of adaptor proteins, like ASC, and the resulting assembly of a large-scale inflammatory signalling platform. The assembly process is driven by sensor-sensor interactions as well as sensor-adaptor and adaptor-adaptor interactions. The resulting complex, which can reach diameters of around 1 micron, has a variable composition and stoichiometry. The inflammasome complex functions as a platform for the proximity induced activation of effector caspases, such as caspase-1 and caspase-8. This ultimately leads to the processing of the inflammatory cytokines pro-IL1β and pro-IL18 into their active forms, along with the cleavage of Gasdermin D, a key activator of cell death via pyroptosis.
Collapse
|
26
|
Salvamoser R, Brinkmann K, O'Reilly LA, Whitehead L, Strasser A, Herold MJ. Characterisation of mice lacking the inflammatory caspases-1/11/12 reveals no contribution of caspase-12 to cell death and sepsis. Cell Death Differ 2019; 26:1124-1137. [PMID: 30154447 PMCID: PMC6748106 DOI: 10.1038/s41418-018-0188-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/02/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023] Open
Abstract
Caspases exert critical functions in diverse cell death pathways, including apoptosis and pyroptosis, but some caspases also have roles in the processing of cytokines into their functional forms during inflammation. The roles of many caspases have been unravelled by the generation of knockout mice, but still very little is known about the overlapping functions of caspases as only a few studies report on double or triple caspase knockout mice. For example, the functions of caspase-12 in cell death and inflammation, on its own or overlapping with the functions of caspase-1 and caspase-11, are only poorly understood. Therefore, we generated a novel mutant mouse strain lacking all three inflammatory caspases, caspases-1, -11 and -12. Analysis under steady state conditions showed no obvious differences between caspase-1/11/12-/- and wildtype (WT) mice. Since caspases-1 and -11 are involved in endotoxic shock, we analysed the response of caspase-1/11/12-/- mice to high-dose LPS injection. Interestingly, we could not detect any differences in responses between caspase-1/11/12-/- mice vs. caspase-1/11 double knockout mice. Furthermore, cell lines generated from caspase-1/11/12-/- mice showed no differences in their apoptotic or necroptotic responses to a diverse set of cytotoxic drugs in vitro when compared to WT cells. Importantly, these drugs also included ER stress-inducing agents, such as thapsigargin and tunicamycin, a form of cell death for which a critical pro-apoptotic function of caspase-12 has previously been reported. Additionally, we found no differences between caspase-1/11/12-/- and WT mice in their in vivo responses to the ER stress-inducing agent, tunicamycin. Collectively, these findings reveal that caspase-12 does not have readily recognisable overlapping roles with caspases-1 and -11 in the inflammatory response induced by LPS and in necroptosis and apoptosis induced by diverse cytotoxic agents, including the ones that elicit ER stress.
Collapse
Affiliation(s)
- Ranja Salvamoser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Kerstin Brinkmann
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lorraine A O'Reilly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
27
|
Jamilloux Y, Lagrange B, Di Micco A, Bourdonnay E, Provost A, Tallant R, Henry T, Martinon F. A proximity-dependent biotinylation (BioID) approach flags the p62/sequestosome-1 protein as a caspase-1 substrate. J Biol Chem 2018; 293:12563-12575. [PMID: 29929983 DOI: 10.1074/jbc.ra117.000435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 06/08/2018] [Indexed: 01/24/2023] Open
Abstract
The inflammasome is a major component of the innate immune system, and its main function is to activate caspase-1, a cysteine protease that promotes inflammation by inducing interleukin-1β (IL-1β) maturation and release into the extracellular milieu. To prevent uncontrolled inflammation, this complex is highly regulated. When it is assembled, the inflammasome is insoluble, which has long precluded the analysis of its interactions with other proteins. Here we used the proximity-dependent biotinylation assay (BioID) to identify proteins associated with caspase-1 during inflammasome activation. Using the BioID in a cell-free system in which the inflammasome had been activated, we found that a caspase-1-biotin ligase fusion protein selectively labeled 111 candidates, including the p62/sequestosome-1 protein (p62). Using co-immunoprecipitation experiments, we demonstrated that p62 interacts with caspase-1. This interaction promoted caspase-1-mediated cleavage of p62 at Asp-329. Mechanistic and functional analyses revealed that caspase-1-mediated cleavage of p62 leads to loss of its interaction with the autophagosomal protein microtubule-associated protein 1 light chain 3 β (LC3B). Strikingly, overexpression of a p62 N-terminal fragment generated upon caspase-1 cleavage decreased IL-1β release, whereas overexpression of p62's C-terminal portion enhanced IL-1β release, by regulating pro-IL1β levels. Overall, the overexpression of both fragments together decreased IL-1β release. Taken together, our results indicate that caspase-1-mediated p62 cleavage plays a complex role in balancing caspase-1-induced inflammation.
Collapse
Affiliation(s)
- Yvan Jamilloux
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Brice Lagrange
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Antonia Di Micco
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Emilie Bourdonnay
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Angélina Provost
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Rémy Tallant
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Thomas Henry
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland, .,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| | - Fabio Martinon
- From the Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland, .,the Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, 69007 Lyon, France, and.,the Department of Internal Medicine, Hopital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004 Lyon, France
| |
Collapse
|
28
|
Nanson JD, Rahaman MH, Ve T, Kobe B. Regulation of signaling by cooperative assembly formation in mammalian innate immunity signalosomes by molecular mimics. Semin Cell Dev Biol 2018; 99:96-114. [PMID: 29738879 DOI: 10.1016/j.semcdb.2018.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/18/2018] [Accepted: 05/04/2018] [Indexed: 12/16/2022]
Abstract
Innate immunity pathways constitute the first line of defense against infections and cellular damage. An emerging concept in these pathways is that signaling involves the formation of finite (e.g. rings in NLRs) or open-ended higher-order assemblies (e.g. filamentous assemblies by members of the death-fold family and TIR domains). This signaling by cooperative assembly formation (SCAF) mechanism allows rapid and strongly amplified responses to minute amounts of stimulus. While the characterization of the molecular mechanisms of SCAF has seen rapid progress, little is known about its regulation. One emerging theme involves proteins produced both in host cells and by pathogens that appear to mimic the signaling components. Recently characterized examples involve the capping of the filamentous assemblies formed by caspase-1 CARDs by the CARD-only protein INCA, and those formed by caspase-8 by the DED-containing protein MC159. By contrast, the CARD-only protein ICEBERG and the DED-containing protein cFLIP incorporate into signaling filaments and presumably interfere with proximity based activation of caspases. We review selected examples of SCAF in innate immunity pathways and focus on the current knowledge on signaling component mimics produced by mammalian and pathogen cells and what is known about their mechanisms of action.
Collapse
Affiliation(s)
- Jeffrey D Nanson
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Md Habibur Rahaman
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Thomas Ve
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia; Institute for Glycomics, Griffith University, Southport, QLD, 4222, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
29
|
NLRP3: A promising therapeutic target for autoimmune diseases. Autoimmun Rev 2018; 17:694-702. [PMID: 29729449 DOI: 10.1016/j.autrev.2018.01.020] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 01/26/2018] [Indexed: 12/12/2022]
Abstract
NLRP3, a member of nucleotide-binding domain-(NOD) like receptor family, can be found in large varieties of immune and non-immune cells. Upon activation, the NLRP3, apoptosis-associated speck-like protein (ASC) and pro-caspase-1 would assemble into a multimeric protein, called the NLRP3 inflammasome. Then the inflammasome promotes inflammation (through specific cleavage and production of bioactive IL-1β and IL-18) and pyroptotic cell death. Previous studies have indicated the importance of NLRP3 in regulating innate immunity. Recently, numerous studies have revealed their significance in autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc) and inflammatory bowel disease (IBD). In this review, we will briefly discuss the biological features of NLRP3 and summarize the recent progression of the involvement of NLRP3 in the development and pathogenesis of autoimmune diseases, as well as its clinical implications and therapeutic potential.
Collapse
|
30
|
Xie J, Belosevic M. Characterization and functional assessment of the NLRC3-like molecule of the goldfish (Carassius auratus L.). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 79:1-10. [PMID: 28988993 DOI: 10.1016/j.dci.2017.09.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 06/07/2023]
Abstract
The NLRC3-like (NLRC3L) molecule from the goldfish transcriptome database was identified and characterized. Quantitative gene expression analysis revealed the highest mRNA levels of NLRC3L were in the spleen and intestine, with lower mRNA levels observed in muscle and liver. Goldfish NLRC3L was differentially expressed in goldfish immune cell populations with highest mRNA levels measured in PBLs and macrophages. We generated a recombinant form of the molecule (rgfNLRC3L) and an anti-CT-NLRC3L IgG. Treatment of goldfish primary kidney macrophages in vitro with ATP, LPS and heat-killed Aeromonas salmonicida up-regulated the NLRC3L mRNA and protein. Confocal microscopy and co-immunoprecipitation assays indicated that goldfish rgfNLRC3L interacted with apoptosis-associated spec-like protein (ASC) in eukaryotic cells, indicating that NLRC3L may participate in the regulation of the inflammasome responses. The dual-luciferase reporter assay showed that NLRC3L over-expression did not cause the activation of NF-κB, but that it cooperated with RIP2 to down-regulate NF-κB activation. Our results indicate that the NLRC3L may function as a regulator of NLR pathways in teleosts.
Collapse
Affiliation(s)
- Jiasong Xie
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
31
|
Poudel B, Gurung P. An update on cell intrinsic negative regulators of the NLRP3 inflammasome. J Leukoc Biol 2018; 103:10.1002/JLB.3MIR0917-350R. [PMID: 29377242 PMCID: PMC6202258 DOI: 10.1002/jlb.3mir0917-350r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/09/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammasomes are multimeric protein complexes that promote inflammation (through specific cleavage and production of bioactive IL-1β and IL-18) and pyroptotic cell death. The central role of inflammasomes in combating infection and maintaining homeostasis has been studied extensively. Although inflammasome-mediated inflammation and cell death are vital to limit pathogenic insults and to promote wound healing/tissue regeneration, unchecked/uncontrolled inflammation, and cell death can cause cytokine storm, tissue damage, autoinflammatory and autoimmune diseases, and even death in the afflicted individuals. NLRP3 is one of the major cytosolic sensors that assemble an inflammasome. Given the adverse consequences of uncontrolled inflammasome activation, our immune system has developed tiered mechanisms to inhibit NLRP3 inflammasome activation. In this review, we highlight and discuss recent advances and our current understanding of mechanisms by which NLRP3 inflammasome can be negatively regulated.
Collapse
Affiliation(s)
- Barun Poudel
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
- Immunology Graduate Program, University of Iowa, Iowa City, Iowa, USA
- Center for Immunology and Immune-Based Disease, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
32
|
NOD-like receptor(s) and host immune responses with Pseudomonas aeruginosa infection. Inflamm Res 2018; 67:479-493. [PMID: 29353310 DOI: 10.1007/s00011-018-1132-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Molecular mechanisms underlying the interactions between Pseudomonas aeruginosa, the common opportunistic pathogen in cystic fibrosis individuals, and host induce a number of marked inflammatory responses and associate with complex therapeutic problems due to bacterial resistance to antibiotics in chronic stage of infection. METHODS Pseudomonas aeruginosa is recognized by number of pattern recognition receptors (PRRs); NOD-like receptors (NLRs) are a class of PRRs, which can recognize a variety of endogenous and exogenous ligands, thereby playing a critical role in innate immunity. RESULTS NLR activation initiates forming of a multi-protein complex called inflammasome that induces activation of caspase-1 and resulted in cleavage of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. When the IL-1β is secreted excessively, this causes tissue damage and extensive inflammatory responses that are potentially hazardous for the host. CONCLUSIONS Recent evidence has laid out inflammasome-forming NLR far beyond inflammation. This review summarizes current knowledge regarding the various roles played by different NLRs and associated down-signals, either in recognition of P. aeruginosa or may be associated with such bacterial pathogen infection, which may relate to for the complexity of lung diseases caused by P. aeruginosa.
Collapse
|
33
|
Ravi Kumar S, Paudel S, Ghimire L, Bergeron S, Cai S, Zemans RL, Downey GP, Jeyaseelan S. Emerging Roles of Inflammasomes in Acute Pneumonia. Am J Respir Crit Care Med 2018; 197:160-171. [PMID: 28930487 PMCID: PMC5768907 DOI: 10.1164/rccm.201707-1391pp] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Sangeetha Ravi Kumar
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Sagar Paudel
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Laxman Ghimire
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Scott Bergeron
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Shanshan Cai
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rachel L. Zemans
- Division of Pulmonary, Sleep, and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Aurora, Colorado; and
| | - Gregory P. Downey
- Division of Pulmonary, Sleep, and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Aurora, Colorado; and
| | - Samithamby Jeyaseelan
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
34
|
Indramohan M, Stehlik C, Dorfleutner A. COPs and POPs Patrol Inflammasome Activation. J Mol Biol 2017; 430:153-173. [PMID: 29024695 DOI: 10.1016/j.jmb.2017.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 01/07/2023]
Abstract
Sensing and responding to pathogens and tissue damage is a core mechanism of innate immune host defense, and inflammasomes represent a central cytosolic pattern recognition receptor pathway leading to the generation of the pro-inflammatory cytokines interleukin-1β and interleukin-18 and pyroptotic cell death that causes the subsequent release of danger signals to propagate and perpetuate inflammatory responses. While inflammasome activation is essential for host defense, deregulated inflammasome responses and excessive release of inflammatory cytokines and danger signals are linked to an increasing spectrum of inflammatory diseases. In this review, we will discuss recent developments in elucidating the role of PYRIN domain-only proteins (POPs) and the related CARD-only proteins (COPs) in regulating inflammasome responses and their impact on inflammatory disease.
Collapse
Affiliation(s)
- Mohanalaxmi Indramohan
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Christian Stehlik
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Interdepartmental Immunobiology Center and Skin Disease Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Andrea Dorfleutner
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
35
|
Mahakur S, Saikia B, Minz M, Minz RW, Nada R, Anand S, Sharma A, Jha V, Joshi N, Goel L, Arora A, Joshi K. Allo-specific immune response profiles indicative of acute rejection in kidney allografts using an in vitro lymphocyte culture-based model. Clin Exp Nephrol 2017; 22:465-473. [PMID: 28849286 DOI: 10.1007/s10157-017-1469-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 08/10/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Ability to predict the manner in which a recipient's immune system would respond to a transplanted graft by analyzing cytokine profiles of the "allograft antigen sensitized" recipient lymphocytes in vitro might provide a means to identify patients at risk to adverse clinical endpoints. METHODS Cytokine/chemokine gene expression profiles of peripheral blood mononuclear cells co-cultured with allograft antigen-pulsed macrophages were studied in 49 renal transplant recipients-12 with acute cellular rejection (ACR) with or without antibody-mediated rejection (AMR), 7 with AMR (without ACR), and 30 with stable allografts (SA). An 86-gene inflammatory cytokines and receptors PCR array was used to measure fold changes in gene expression between pulsed and un-pulsed cultures. RESULTS On linear discriminant analysis and multivariate analysis of variance, a gene set comprising C3, CCL3, IL1B, TOLLIP, IL10, CXCL5, ABCF1, CCR3, IL10RB, CXCL1, and IL1R1 differentiated the ACR-AMR from the SA group. Similarly, a gene set comprising IL10, C3, IL37, IL1B, CCL3, CARD18, and TOLLIP differentiated the AMR from the SA group. No significant difference was found between the ACR-AMR vs AMR groups. CONCLUSION Distinct post in vitro stimulation cytokine profiles at the time of transplantation thus correlated with the occurrence of post-transplantation rejection episodes which indicated feasibility of this in vitro model to assess the recipient's anti-graft response at an early stage.
Collapse
Affiliation(s)
- Sobhana Mahakur
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Biman Saikia
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India.
| | - Mukut Minz
- Department of Renal Transplant Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ranjana W Minz
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Ritambhra Nada
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shashi Anand
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Ashish Sharma
- Department of Renal Transplant Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vivekanand Jha
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neha Joshi
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Lekha Goel
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Amit Arora
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kusum Joshi
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
36
|
Qin H, Jin J, Fischer H, Mildner M, Gschwandtner M, Mlitz V, Eckhart L, Tschachler E. The caspase-1 inhibitor CARD18 is specifically expressed during late differentiation of keratinocytes and its expression is lost in lichen planus. J Dermatol Sci 2017; 87:176-182. [PMID: 28506683 DOI: 10.1016/j.jdermsci.2017.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/27/2017] [Indexed: 01/25/2023]
Abstract
BACKGROUND CARD18 contains a caspase recruitment domain (CARD) via which it binds to caspase-1 and thereby inhibits caspase-1-mediated activation of the pro-inflammatory cytokine interleukin (IL)-1β. OBJECTIVES To determine the expression profile and the role of CARD18 during differentiation of keratinocytes and to compare the expression of CARD18 in normal skin and in inflammatory skin diseases. METHODS Human keratinocytes were induced to differentiate in monolayer and in 3D skin equivalent cultures. In some experiments, CARD18-specific siRNAs were used to knock down expression of CARD18. CARD18 mRNA levels were determined by quantitative real-time PCR, and CARD18 protein was detected by Western blot and immunofluorescence analyses. In situ expression was analyzed in skin biopsies obtained from healthy donors and patients with psoriasis and lichen planus. RESULTS CARD18 mRNA was expressed in the epidermis at more than 100-fold higher levels than in any other human tissue. Within the epidermis, CARD18 was specifically expressed in the granular layer. In vitro CARD18 was strongly upregulated at both mRNA and protein levels in keratinocytes undergoing terminal differentiation. In skin equivalent cultures the expression of CARD18 was efficiently suppressed by siRNAs without impairing stratum corneum formation. Epidermal expression of CARD18 was increased after ultraviolet (UV)B irradiation of skin explants. In skin biopsies of patients with psoriasis no consistent regulation of CARD18 expression was observed, however, in lesional epidermis of patients with lichen planus, CARD18 expression was either greatly diminished or entirely absent whereas in non-lesional areas expression was comparable to normal skin. CONCLUSIONS Our results identify CARD18 as a differentiation-associated keratinocyte protein that is altered in abundance by UV stress. Its downregulation in lichen planus indicates a potential role in inflammatory reactions of the epidermis in this disease.
Collapse
Affiliation(s)
- Haihong Qin
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiang Jin
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Heinz Fischer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Gschwandtner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Veronika Mlitz
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Leopold Eckhart
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
Hoss F, Rodriguez-Alcazar JF, Latz E. Assembly and regulation of ASC specks. Cell Mol Life Sci 2017; 74:1211-1229. [PMID: 27761594 PMCID: PMC11107573 DOI: 10.1007/s00018-016-2396-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
Abstract
The inflammasome adapter ASC links activated inflammasome sensors to the effector molecule pro-caspase-1. Recruitment of pro-caspase-1 to ASC promotes the autocatalytic activation of caspase-1, which leads to the release of pro-inflammatory cytokines, such as IL-1β. Upon triggering of inflammasome sensors, ASC assembles into large helical fibrils that interact with each other serving as a supramolecular signaling platform termed the ASC speck. Alternative splicing, post-translational modifications of ASC, as well as interaction with other proteins can perturb ASC function. In several inflammatory diseases, ASC specks can be found in the extracellular space and its presence correlates with poor prognosis. Here, we review the role of ASC in inflammation, and focus on the structural mechanisms that lead to ASC speck formation, the regulation of ASC function during inflammasome assembly, and the importance of ASC specks in disease.
Collapse
Affiliation(s)
- Florian Hoss
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany
| | - Juan F Rodriguez-Alcazar
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
- German Center for Neurodegenerative Diseases, Bonn, Germany.
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
38
|
Pang M, Bai XY, Li Y, Bai JZ, Yuan LR, Ren SA, Hu XY, Zhang XR, Yu BF, Guo R, Wang HL. Label-free LC-MS/MS shotgun proteomics to investigate the anti-inflammatory effect of rCC16. Mol Med Rep 2016; 14:4496-4504. [PMID: 27748820 PMCID: PMC5101986 DOI: 10.3892/mmr.2016.5841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 08/18/2016] [Indexed: 12/30/2022] Open
Abstract
Clara cell protein (CC16) is an anti-inflammatory protein, which is expressed in the airway epithelium. It is involved in the development of airway inflammatory diseases, including chronic obstructive pulmonary disease and asthma. However, the exact molecular mechanism underlying its anti‑inflammatory action remains to be fully elucidated. The aim of the present study was to define the protein profiles of the anti‑inflammatory effect of CC16 in lipopolysaccharide (LPS)‑treated rat tracheal epithelial (RTE) cells using shotgun proteomics. Protein extracts were obtained from control RTE cells, RTE cells treated with LPS and RTE cells treated with LPS and recombinant CC16 (rCC16). Subsequent label‑free quantification and bioinformatics analyses identified 12 proteins that were differentially expressed in the three treatment groups as a cluster of five distinct groups according to their molecular functions. Five of the twelve proteins were revealed to be associated with the cytoskeleton: Matrix metalloproteinase‑9, myosin heavy chain 10, actin‑related protein‑3 homolog, elongation factor 1‑α‑1 (EF‑1‑α‑1), and acidic ribosomal phosphoprotein P0. Five of the twelve proteins were associated with cellular proliferation: DNA‑dependent protein kinase catalytic subunit, EF‑1‑α‑1, tyrosine 3‑monooxygenase, caspase recruitment domain (CARD) protein 12 and adenosylhomocysteinase (SAHH) 3. Three proteins were associated with gene regulation: EF‑1‑α‑1, SAHH 3 and acidic ribosomal phosphoprotein P0. Three proteins were associated with inflammation: Tyrosine 3‑monooxygenase, CARD protein 12 and statin‑related protein. ATPase (H+‑transporting, V1 subunit A, isoform 1) was revealed to be associated with energy metabolism, and uridine diphosphate glycosyltransferase 1 family polypeptide A8 with drug metabolism and detoxification. The identified proteins were further validated using reverse transcription‑quantitative polymerase chain reaction. These protein profiles, and their interacting protein network, may facilitate the elucidation of the molecular mechanisms underlying the anti‑inflammatory effects of CC16.
Collapse
Affiliation(s)
- Min Pang
- Respiratory Department, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xin-Yan Bai
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yan Li
- Fan‑Xing Biological Technology Co., Ltd., Beijing 010000, P.R. China
| | - Ji-Zhong Bai
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Li-Rong Yuan
- Respiratory Department, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Shou-An Ren
- Respiratory Department, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiao-Yun Hu
- Respiratory Department, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xin-Ri Zhang
- Respiratory Department, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bao-Feng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Rui Guo
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Hai-Long Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
39
|
[Interleukin-1, inflammasome and autoinflammatory diseases]. Rev Med Interne 2016; 39:233-239. [PMID: 27639913 DOI: 10.1016/j.revmed.2016.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/29/2016] [Indexed: 01/26/2023]
Abstract
Interleukin-1 is a major cytokine of innate immunity and inflammation. It exerts various systemic effects during the inflammatory response, such as fever induction, thrombopoiesis and granulopoiesis, or leukocyte recruitment. Its involvement has been demonstrated in many inflammatory-mediated diseases, such as diabetes or gout. Moreover, interleukin-1 plays a pivotal role in some autoinflammatory diseases, such as cryopyrinopathies or familial Mediterranean fever. In these diseases, a constitutional defect of the inflammasome, a protein complex responsible for the activation of interleukin-1, explains the hypersecretion of interleukin-1. Other autoinflammatory diseases have a more complex pathophysiology involving deregulation of the interleukin-1 pathway, upstream or downstream of the inflammasome, or through more complex mechanisms. In this review, we are detailing the synthesis, the activation, the signalling, and the regulation of interleukin-1. We then describe the autoinflammatory diseases or related-diseases where the pathological role of interleukin-1 has been demonstrated.
Collapse
|
40
|
Abstract
Inflammasomes are multiprotein signalling platforms that control the inflammatory response and coordinate antimicrobial host defences. They are assembled by pattern-recognition receptors following the detection of pathogenic microorganisms and danger signals in the cytosol of host cells, and they activate inflammatory caspases to produce cytokines and to induce pyroptotic cell death. The clinical importance of inflammasomes reaches beyond infectious disease, as dysregulated inflammasome activity is associated with numerous hereditary and acquired inflammatory disorders. In this Review, we discuss the recent developments in inflammasome research with a focus on the molecular mechanisms that govern inflammasome assembly, signalling and regulation.
Collapse
Affiliation(s)
- Petr Broz
- Focal Area Infection Biology, Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Vishva M Dixit
- Genentech Inc., South San Francisco, California 94080, USA
| |
Collapse
|
41
|
Stewart MK, Cookson BT. Evasion and interference: intracellular pathogens modulate caspase-dependent inflammatory responses. Nat Rev Microbiol 2016; 14:346-59. [PMID: 27174147 DOI: 10.1038/nrmicro.2016.50] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pathogens have evolved to complete the virulence cycle of colonization, replication and dissemination in intimate association with a complex network of extracellular and intracellular surveillance systems that guard tissue spaces. In this Review, we discuss the strategies used by bacteria and viruses to evade or inhibit intracellular detection that is coupled to pro-inflammatory caspase-dependent protective responses. Such strategies include alterations of lipopolysaccharide (LPS) structures, the regulated expression of components of type III secretion systems, and the utilization of proteins that inhibit inflammasome formation, the enzymatic activity of caspases and cytokine signalling. Inflammation is crucial in response to exposure to pathogens, but is potentially damaging and thus tightly regulated. The threshold for the activation of pro-inflammatory caspases is determined by the immediate stimulus in the context of previous signals. Pathogen, genetic and situational factors modulate this threshold, which determines the ability of the host to resist infection while minimizing harm.
Collapse
Affiliation(s)
- Mary K Stewart
- Department of Microbiology, University of Washington, Seattle, Washington 98195, USA
| | - Brad T Cookson
- Department of Microbiology, University of Washington, Seattle, Washington 98195, USA.,Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
42
|
Pourcet B, Gage MC, León TE, Waddington KE, Pello OM, Steffensen KR, Castrillo A, Valledor AF, Pineda-Torra I. The nuclear receptor LXR modulates interleukin-18 levels in macrophages through multiple mechanisms. Sci Rep 2016; 6:25481. [PMID: 27149934 PMCID: PMC4858669 DOI: 10.1038/srep25481] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/19/2016] [Indexed: 12/30/2022] Open
Abstract
IL-18 is a member of the IL-1 family involved in innate immunity and inflammation. Deregulated levels of IL-18 are involved in the pathogenesis of multiple disorders including inflammatory and metabolic diseases, yet relatively little is known regarding its regulation. Liver X receptors or LXRs are key modulators of macrophage cholesterol homeostasis and immune responses. Here we show that LXR ligands negatively regulate LPS-induced mRNA and protein expression of IL-18 in bone marrow-derived macrophages. Consistent with this being an LXR-mediated process, inhibition is abolished in the presence of a specific LXR antagonist and in LXR-deficient macrophages. Additionally, IL-18 processing of its precursor inactive form to its bioactive state is inhibited by LXR through negative regulation of both pro-caspase 1 expression and activation. Finally, LXR ligands further modulate IL-18 levels by inducing the expression of IL-18BP, a potent endogenous inhibitor of IL-18. This regulation occurs via the transcription factor IRF8, thus identifying IL-18BP as a novel LXR and IRF8 target gene. In conclusion, LXR activation inhibits IL-18 production through regulation of its transcription and maturation into an active pro-inflammatory cytokine. This novel regulation of IL-18 by LXR could be applied to modulate the severity of IL-18 driven metabolic and inflammatory disorders.
Collapse
Affiliation(s)
- Benoit Pourcet
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| | - Matthew C Gage
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| | - Theresa E León
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| | - Kirsty E Waddington
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| | - Oscar M Pello
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| | - Knut R Steffensen
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institut, Huddinge, Sweden
| | - Antonio Castrillo
- Instituto de Investigaciones Biomedicas "Alberto Sols" Consejo Superior de Investigaciones Científicas (CSIC) de Madrid, Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Annabel F Valledor
- School of Biology, University of Barcelona, Diagonal 643, Planta 3, 08028 Barcelona, Spain
| | - Inés Pineda-Torra
- Centre for Clinical Pharmacology, Division of Medicine, University College of London, 5 University Street, London, WC1 E6JF, United Kingdom
| |
Collapse
|
43
|
Lu A, Li Y, Schmidt FI, Yin Q, Chen S, Fu TM, Tong AB, Ploegh HL, Mao Y, Wu H. Molecular basis of caspase-1 polymerization and its inhibition by a new capping mechanism. Nat Struct Mol Biol 2016; 23:416-25. [PMID: 27043298 PMCID: PMC4856535 DOI: 10.1038/nsmb.3199] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 03/10/2016] [Indexed: 12/18/2022]
Abstract
Inflammasomes are cytosolic caspase-1-activation complexes that sense intrinsic and extrinsic danger signals, and trigger inflammatory responses and pyroptotic cell death. Homotypic interactions among Pyrin domains and caspase recruitment domains (CARDs) in inflammasome-complex components mediate oligomerization into filamentous assemblies. Several cytosolic proteins consisting of only interaction domains exert inhibitory effects on inflammasome assembly. In this study, we determined the structure of the human caspase-1 CARD domain (caspase-1(CARD)) filament by cryo-electron microscopy and investigated the biophysical properties of two caspase-1-like CARD-only proteins: human inhibitor of CARD (INCA or CARD17) and ICEBERG (CARD18). Our results reveal that INCA caps caspase-1 filaments, thereby exerting potent inhibition with low-nanomolar Ki on caspase-1(CARD) polymerization in vitro and inflammasome activation in cells. Whereas caspase-1(CARD) uses six complementary surfaces of three types for filament assembly, INCA is defective in two of the six interfaces and thus terminates the caspase-1 filament.
Collapse
Affiliation(s)
- Alvin Lu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Yang Li
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Florian I Schmidt
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Qian Yin
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Shuobing Chen
- Center for Quantitative Biology, Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.,Department of Cancer Immunology and Virology, Intel Parallel Computing Center for Structural Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tian-Min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alexander B Tong
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Youdong Mao
- Center for Quantitative Biology, Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.,Department of Cancer Immunology and Virology, Intel Parallel Computing Center for Structural Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Göblös A, Danis J, Vas K, Bata-Csörgő Z, Kemény L, Széll M. Keratinocytes express functional CARD18, a negative regulator of inflammasome activation, and its altered expression in psoriasis may contribute to disease pathogenesis. Mol Immunol 2016; 73:10-8. [PMID: 27023378 DOI: 10.1016/j.molimm.2016.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/10/2016] [Accepted: 03/22/2016] [Indexed: 01/08/2023]
Abstract
Caspase recruitment domain family member 18 (CARD18, Iceberg) is known as a negative regulatory molecule that inhibits inflammatory events by terminating inflammasome activation due to a direct interaction with pro-caspase-1. During the investigation of molecular mechanisms in keratinocytes that contribute to the pathogenesis of psoriasis, we found that CARD18 expression differs in healthy and psoriatic skin; moreover, CARD18 demonstrated altered response under inflammatory conditions in healthy and psoriatic skin. In healthy skin, low basal CARD18 expression was detected, which showed significant elevation in response to inflammatory stimuli (lymphokine treatment or mechanical injury). In contrast, higher basal expression was observed in psoriatic non-involved skin, but no further induction could be detected. We demonstrated that keratinocytes express CARD18 both at mRNA and protein levels and the expression increased in parallel with differentiation. The investigation of cellular inflammatory processes revealed that psoriasis-associated danger signals triggered the expression of inflammasome components (AIM2, Caspase-1) and CARD18 as well as IL-1β production of keratinocytes. Furthermore, gene-specific silencing of CARD18 in cells treated with cytosolic DNA (poly(dA:dT)) resulted in increased IL-1β secretion, suggesting a negative regulatory role for CARD18 in keratinocyte inflammatory signaling. The differential regulation of CARD18 in healthy and psoriatic uninvolved epidermis may contribute to the susceptibility of psoriasis. Furthermore, our in vitro results indicate that CARD18 may contribute to the fine tuning of keratinocyte innate immune processes.
Collapse
Affiliation(s)
- Anikó Göblös
- Department of Dermatology and Allergology, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary.
| | - Judit Danis
- Department of Dermatology and Allergology, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary.
| | - Krisztina Vas
- Department of Dermatology and Allergology, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary.
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary; MTA-SZTE Dermatological Research Group, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary.
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary; MTA-SZTE Dermatological Research Group, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary.
| | - Márta Széll
- MTA-SZTE Dermatological Research Group, University of Szeged, Korányifasor 6, H-6720 Szeged, Hungary; Department of Medical Genetics, University of Szeged, Somogyi u. 4, H-6720 Szeged, Hungary.
| |
Collapse
|
45
|
Matusiak M, Van Opdenbosch N, Lamkanfi M. CARD- and pyrin-only proteins regulating inflammasome activation and immunity. Immunol Rev 2016; 265:217-30. [PMID: 25879296 DOI: 10.1111/imr.12282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Membrane-bound and intracellular immune receptors respond to microbial pathogens by initiating signaling cascades that result in production of inflammatory cytokines and antimicrobial factors. These host responses need to be tightly regulated to prevent tissue damage and other harmful consequences of excessive inflammation. CARD-only proteins (COPs) and Pyrin-only proteins (POPs) are human- and primate-specific dominant negative inhibitors that modulate inflammatory and innate immune responses. In addition, several poxviruses encode POPs that interfere with inflammatory and host defense responses. COPs and POPs modulate inflammatory signaling at several checkpoints by sequestering key components of the inflammasome and NF-κB signaling cascades, thus hampering downstream signal transduction. Here, we review and discuss current understanding of the evolutionary history and molecular mechanisms by which roles of host- and virus-encoded COPs and POPs may regulate inflammatory and immune responses. In addition, we address their (patho)physiological roles and highlight topics for further research.
Collapse
Affiliation(s)
- Magdalena Matusiak
- Department of Medical Protein Research, VIB, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | | | | |
Collapse
|
46
|
Abstract
Inflammasomes are protein complexes that promote the maturation and release of pro-inflammatory cytokines and danger signals as well as pyroptosis in response to infections and cellular stress. Inflammasomes consist of a sensor, an adapter, and the effector caspase-1, which interact through homotypic interactions of caspase recruitment domains (CARDs) or PYRIN domains (PYDs). Hence, decoy proteins encoding only a CARD or PYD, COPs and POPs, respectively, are assumed to inhibit inflammasome assembly. Sensors encoding a PYD belong to the families of NOD-like receptors containing a PYD (NLRPs) or AIM2-like receptors (ALRs), which interact with the PYD- and CARD-containing adapter ASC through homotypic PYD interactions. Subsequently, ASC undergoes PYD-dependent oligomerization, which promotes CARD-mediated interactions between ASC and caspase-1, resulting in caspase-1 activation. POPs are suggested to interfere with the interaction between NLRPs/ALRs and ASC to prevent nucleation of ASC and therefore prevent an oligomeric platform for caspase-1 activation. Similarly, COPs are suggested to bind to the CARD of caspase-1 to prevent its recruitment to the oligomeric ASC platform and its activation. Alternatively, the adapter ASC may regulate inflammasome activity by expressing different isoforms, which are either capable or incapable of assembling an oligomeric ASC platform. The molecular mechanism of inflammasome assembly has only recently been elucidated, but the effects of most COPs and POPs on inflammasome assembly have not been investigated. Here, we discuss our model of COP- and POP-mediated inflammasome regulation.
Collapse
Affiliation(s)
- Andrea Dorfleutner
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | |
Collapse
|
47
|
Pedraza-Alva G, Pérez-Martínez L, Valdez-Hernández L, Meza-Sosa KF, Ando-Kuri M. Negative regulation of the inflammasome: keeping inflammation under control. Immunol Rev 2016; 265:231-57. [PMID: 25879297 DOI: 10.1111/imr.12294] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In addition to its roles in controlling infection and tissue repair, inflammation plays a critical role in diverse and distinct chronic diseases, such as cancer, metabolic syndrome, and neurodegenerative disorders, underscoring the harmful effect of an uncontrolled inflammatory response. Regardless of the nature of the stimulus, initiation of the inflammatory response is mediated by assembly of a multimolecular protein complex called the inflammasome, which is responsible for the production of inflammatory cytokines, such as interleukin-1β (IL-1β) and IL-18. The different stimuli and mechanisms that control inflammasome activation are fairly well understood, but the mechanisms underlying the control of undesired inflammasome activation and its inactivation remain largely unknown. Here, we review recent advances in our understanding of the molecular mechanisms that negatively regulate inflammasome activation to prevent unwanted activation in the resting state, as well as those involved in terminating the inflammatory response after a specific insult to maintain homeostasis.
Collapse
Affiliation(s)
- Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | | | | | | |
Collapse
|
48
|
Lind KF, Østerud B, Hansen E, Jørgensen TØ, Andersen JH. The immunomodulatory effects of barettin and involvement of the kinases CAMK1α and RIPK2. Immunopharmacol Immunotoxicol 2015; 37:458-64. [DOI: 10.3109/08923973.2015.1082584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
49
|
Kao WP, Yang CY, Su TW, Wang YT, Lo YC, Lin SC. The versatile roles of CARDs in regulating apoptosis, inflammation, and NF-κB signaling. Apoptosis 2015; 20:174-95. [PMID: 25420757 DOI: 10.1007/s10495-014-1062-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CARD subfamily is the second largest subfamily in the DD superfamily that plays important roles in regulating various signaling pathways, including but not limited to NF-kB activation signaling, apoptosis signaling and inflammatory signaling. The CARD subfamily contains 33 human CARD-containing proteins, regulating the assembly of many signaling complexes, including apoptosome, inflammsome, nodosome, the CBM complex, PIDDosome, the TRAF2 complex, and the MAVS signalosome, by homotypic CARD-CARD interactions. The mechanism of how CARDs find the right binding partner to form a specific complex remains unclear. This review uses different classification schemes to update the classification of CARD-containing proteins. Combining the classification based on domain structures, functions, associated signaling complexes, and roles would help better understand the structural and function diversity of CARD-containing proteins. This review also summarizes recent structural studies on CARDs. Especially, the CARD-containing complexes can be divided into the homodimeric, heterodimeric, oligomeric, filamentous CARD complexes and the CARD-ubiquitin complex. This review will give an overview of the versatile roles of CARDs in regulating signaling transduction, as well as the therapeutic drugs targeting CARD-containing proteins.
Collapse
Affiliation(s)
- Wen-Pin Kao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
50
|
Cheon SY, Chung KS, Jeon E, Nugroho A, Park HJ, An HJ. Anti-inflammatory Activity of Saxifragin via Inhibition of NF-κB Involves Caspase-1 Activation. JOURNAL OF NATURAL PRODUCTS 2015; 78:1579-1585. [PMID: 26171782 DOI: 10.1021/acs.jnatprod.5b00145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Saxifragin, the 5-glucoside of the flavonoid quercetin, is found in plants and insects. It has been reported that saxifragin has peroxynitrite-scavenging effects. However, the mechanism of anti-inflammatory effects of saxifragin has not yet been clearly identified. In this study, we investigated the anti-inflammatory effects of saxifragin in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and animal models of inflammation. We found that saxifragin suppressed the production of nitric oxide (NO) and prostaglandin E2 (PGE2) in LPS-activated RAW 264.7 macrophages by suppressing the level of protein and mRNA expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), respectively. Furthermore, saxifragin inhibited mRNA expression of pro-inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β. We studied the inhibitory effects of saxifragin on the nuclear translocation of nuclear factor (NF)-κB, activation of caspase-1, and phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Furthermore, pretreatment with saxifragin increased the survival rate of mice with LPS-induced septic death. Collectively, these findings suggest that saxifragin exerts anti-inflammatory activity by inhibiting NF-κB, caspase-1, and mitogen-activated protein kinase (MAPK) activation.
Collapse
Affiliation(s)
- Se-Yun Cheon
- †Department of Pharmacology, College of Oriental Medicine, Sangji University, Gangwon-do 220-702, Republic of Korea
| | - Kyung-Sook Chung
- †Department of Pharmacology, College of Oriental Medicine, Sangji University, Gangwon-do 220-702, Republic of Korea
| | - Eunjin Jeon
- †Department of Pharmacology, College of Oriental Medicine, Sangji University, Gangwon-do 220-702, Republic of Korea
| | - Agung Nugroho
- ‡Department of Agroindustrial Technology, Lambung Mangkurat University, Banjarbaru, Indonesia
| | - Hee-Jun Park
- §Department of Pharmaceutical Engineering, College of Health Science, Sangji University, Wonju, Republic of Korea
| | - Hyo-Jin An
- †Department of Pharmacology, College of Oriental Medicine, Sangji University, Gangwon-do 220-702, Republic of Korea
| |
Collapse
|