51
|
de Gaetano M. Development of synthetic lipoxin-A4 mimetics (sLXms): New avenues in the treatment of cardio-metabolic diseases. Semin Immunol 2023; 65:101699. [PMID: 36428172 DOI: 10.1016/j.smim.2022.101699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
Resolution of inflammation is a complex, dynamic process consisting of several distinct processes, including inhibition of endothelial activation and leukocyte trafficking; promotion of inflammatory cell apoptosis and subsequent non-phlogistic scavenging and degradation; augmentation of pathogen phagocytosis; modulation of stromal cell phenotype coupled to the promotion of tissue regeneration and repair. Among these tightly regulated processes, the clearance and degradation of apoptotic cells without eliciting an inflammatory response is a crucial allostatic mechanism vital to developmental processes, host defence, and the effective resolution of inflammation. These efferocytic and subsequent effero-metabolism processes can be carried out by professional and non-professional phagocytes. Defective removal or inadequate processing of apoptotic cells leads to persistent unresolved inflammation, which may promote insidious pathologies including scarring, fibrosis, and eventual organ failure. In this manuscript, the well-established role of endothelial activation and leukocyte extravasation, as classical vascular targets of the 'inflammation pharmacology', will be briefly reviewed. The main focus of this work is to bring attention to a less explored aspect of the 'resolution pharmacology', aimed at tackling defective efferocytosis and inefficient effero-metabolism, as key targeted mechanisms to prevent or pre-empt vascular complications in cardio-metabolic diseases. Despite the use of gold standard lipid-lowering drugs or glucose-lowering drugs, none of them are able to tackle the so called residual inflammatory risk and/or the metabolic memory. In this review, the development of synthetic mimetics of endogenous mediators of inflammation is highlighted. Such molecules finely tune key components across the whole inflammatory process, amongst various other novel therapeutic paradigms that have emerged over the past decade, including anti-inflammatory therapy. More specifically, FPR2-agonists in general, and Lipoxin analogues in particular, greatly enhance the reprogramming and cross-talk between classical and non-classical innate immune cells, thus inducing both termination of the pro-inflammatory state as well as promoting the subsequent resolving phase, which represent pivotal mechanisms in inflammatory cardio-metabolic diseases.
Collapse
Affiliation(s)
- Monica de Gaetano
- Diabetes Complications Research Centre, Conway Institute & School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
52
|
Wei S, Feng M, Zhang S. Molecular Characteristics of Cell Pyroptosis and Its Inhibitors: A Review of Activation, Regulation, and Inhibitors. Int J Mol Sci 2022; 23:ijms232416115. [PMID: 36555757 PMCID: PMC9783510 DOI: 10.3390/ijms232416115] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Pyroptosis is an active and ordered form of programmed cell death. The signaling pathways of pyroptosis are mainly divided into canonical pathways mediated by caspase-1 and noncanonical pathways mediated by caspase-11. Cell pyroptosis is characterized by the activation of inflammatory caspases (mainly caspase-1, 4, 5, 11) and cleavage of various members of the Gasdermin family to form membrane perforation components, leading to cell membrane rupture, inflammatory mediators release, and cell death. Moderate pyroptosis is an innate immune response that fights against infection and plays an important role in the occurrence and development of the normal function of the immune system. However, excessive pyroptosis occurs and leads to immune disorders in many pathological conditions. Based on canonical pathways, research on pyroptosis regulation has demonstrated several pyroptotic inhibitors, including small-molecule drugs, natural products, and formulations of traditional Chinese medicines. In this paper, we review the characteristics and molecular mechanisms of pyroptosis, summarize inhibitors of pyroptosis, and propound that herbal medicines should be a focus on the research and development for pyroptosis blockers.
Collapse
Affiliation(s)
| | | | - Shidong Zhang
- Correspondence: ; Tel.: +86-931-211-5256; Fax: +86-931-211-5191
| |
Collapse
|
53
|
Yu X, Ni T, Munson G, Zhang P, Gilbert RJC. Cryo-EM structures of perforin-2 in isolation and assembled on a membrane suggest a mechanism for pore formation. EMBO J 2022; 41:e111857. [PMID: 36245269 PMCID: PMC9713709 DOI: 10.15252/embj.2022111857] [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: 06/13/2022] [Revised: 07/29/2022] [Accepted: 09/09/2022] [Indexed: 01/15/2023] Open
Abstract
Perforin-2 (PFN2, MPEG1) is a key pore-forming protein in mammalian innate immunity restricting intracellular bacteria proliferation. It forms a membrane-bound pre-pore complex that converts to a pore-forming structure upon acidification; but its mechanism of conformational transition has been debated. Here we used cryo-electron microscopy, tomography and subtomogram averaging to determine structures of PFN2 in pre-pore and pore conformations in isolation and bound to liposomes. In isolation and upon acidification, the pre-assembled complete pre-pore rings convert to pores in both flat ring and twisted conformations. On membranes, in situ assembled PFN2 pre-pores display various degrees of completeness; whereas PFN2 pores are mainly incomplete arc structures that follow the same subunit packing arrangements as found in isolation. Both assemblies on membranes use their P2 β-hairpin for binding to the lipid membrane surface. Overall, these structural snapshots suggest a molecular mechanism for PFN2 pre-pore to pore transition on a targeted membrane, potentially using the twisted pore as an intermediate or alternative state to the flat conformation, with the capacity to cause bilayer distortion during membrane insertion.
Collapse
Affiliation(s)
- Xiulian Yu
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Calleva Research Centre for Evolution and Human Sciences, Magdalen CollegeUniversity of OxfordOxfordUK
| | - Tao Ni
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Present address:
School of Biomedical Sciences, LKS Faculty of MedicineThe University of Hong KongPokfulamHong Kong SARChina
| | - George Munson
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Peijun Zhang
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotUK
- Chinese Academy of Medical Sciences Oxford InstituteUniversity of OxfordOxfordUK
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Calleva Research Centre for Evolution and Human Sciences, Magdalen CollegeUniversity of OxfordOxfordUK
| |
Collapse
|
54
|
Schaefer SL, Hummer G. Sublytic gasdermin-D pores captured in atomistic molecular simulations. eLife 2022; 11:e81432. [PMID: 36374182 PMCID: PMC9699695 DOI: 10.7554/elife.81432] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Gasdermin-D (GSDMD) is the ultimate effector of pyroptosis, a form of programmed cell death associated with pathogen invasion and inflammation. After proteolytic cleavage by caspases, the GSDMD N-terminal domain (GSDMDNT) assembles on the inner leaflet of the plasma membrane and induces the formation of membrane pores. We use atomistic molecular dynamics simulations to study GSDMDNT monomers, oligomers, and rings in an asymmetric plasma membrane mimetic. We identify distinct interaction motifs of GSDMDNT with phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) and phosphatidylserine (PS) headgroups and describe their conformational dependence. Oligomers are stabilized by shared lipid binding sites between neighboring monomers acting akin to double-sided tape. We show that already small GSDMDNT oligomers support stable, water-filled, and ion-conducting membrane pores bounded by curled beta-sheets. In large-scale simulations, we resolve the process of pore formation from GSDMDNT arcs and lipid efflux from partial rings. We find that high-order GSDMDNT oligomers can crack under the line tension of 86 pN created by an open membrane edge to form the slit pores or closed GSDMDNT rings seen in atomic force microscopy experiments. Our simulations provide a detailed view of key steps in GSDMDNT-induced plasma membrane pore formation, including sublytic pores that explain nonselective ion flux during early pyroptosis.
Collapse
Affiliation(s)
- Stefan L Schaefer
- Department of Theoretical Biophysics, Max Planck Institute of BiophysicsFrankfurt am MainGermany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of BiophysicsFrankfurt am MainGermany
- Institute of Biophysics, Goethe University FrankfurtFrankfurt am MainGermany
| |
Collapse
|
55
|
Yang F, Bettadapura SN, Smeltzer MS, Zhu H, Wang S. Pyroptosis and pyroptosis-inducing cancer drugs. Acta Pharmacol Sin 2022; 43:2462-2473. [PMID: 35288674 PMCID: PMC9525650 DOI: 10.1038/s41401-022-00887-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/07/2022] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis, an inflammatory form of lytic cell death, is a type of cell death mediated by the gasdermin (GSDM) protein family. Upon recognizing exogenous or endogenous signals, cells undergo inflammasome assembly, GSDM cleavage, the release of proinflammatory cytokines and other cellular contents, eventually leading to inflammatory cell death. In this review, we discuss the roles of the GSDM family for anti-cancer functions and various antitumor drugs that could activate the pyroptosis pathways.
Collapse
Affiliation(s)
- Fan Yang
- Healthville LLC, Little Rock, AR, 72204, USA
| | - Sahana N Bettadapura
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR, 72204, USA
| | - Mark S Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | - Shanzhi Wang
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, 72204, USA.
| |
Collapse
|
56
|
Russell CM, Schaefer KG, Dixson A, Gray ALH, Pyron RJ, Alves DS, Moore N, Conley EA, Schuck RJ, White TA, Do TD, King GM, Barrera FN. The Candida albicans virulence factor candidalysin polymerizes in solution to form membrane pores and damage epithelial cells. eLife 2022; 11:e75490. [PMID: 36173096 PMCID: PMC9522247 DOI: 10.7554/elife.75490] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 08/15/2022] [Indexed: 11/28/2022] Open
Abstract
Candida albicans causes severe invasive candidiasis. C. albicans infection requires the virulence factor candidalysin (CL) which damages target cell membranes. However, the mechanism that CL uses to permeabilize membranes is unclear. We reveal that CL forms membrane pores using a unique mechanism. Unexpectedly, CL readily assembled into polymers in solution. We propose that the basic structural unit in polymer formation is a CL oligomer, which is sequentially added into a string configuration that can close into a loop. CL loops appear to spontaneously insert into the membrane to become pores. A CL mutation (G4W) inhibited the formation of polymers in solution and prevented pore formation in synthetic lipid systems. Epithelial cell studies showed that G4W CL failed to activate the danger response pathway, a hallmark of the pathogenic effect of CL. These results indicate that CL polymerization in solution is a necessary step for the damage of cellular membranes. Analysis of CL pores by atomic force microscopy revealed co-existence of simple depressions and more complex pores, which are likely formed by CL assembled in an alternate oligomer orientation. We propose that this structural rearrangement represents a maturation mechanism that stabilizes pore formation to achieve more robust cellular damage. To summarize, CL uses a previously unknown mechanism to damage membranes, whereby pre-assembly of CL loops in solution leads to formation of membrane pores. Our investigation not only unravels a new paradigm for the formation of membrane pores, but additionally identifies CL polymerization as a novel therapeutic target to treat candidiasis.
Collapse
Affiliation(s)
- Charles M Russell
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Katherine G Schaefer
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
| | - Andrew Dixson
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Amber LH Gray
- Department of Chemistry, University of TennesseeKnoxvilleUnited States
| | - Robert J Pyron
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Daiane S Alves
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Nicholas Moore
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Elizabeth A Conley
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
| | - Ryan J Schuck
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Tommi A White
- Department of Biochemistry, University of MissouriColumbiaUnited States
- Electron Microscopy Core, University of MissouriColumbiaUnited States
| | - Thanh D Do
- Department of Chemistry, University of TennesseeKnoxvilleUnited States
| | - Gavin M King
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| |
Collapse
|
57
|
Qi S, Wang Q, Zhang J, Liu Q, Li C. Pyroptosis and Its Role in the Modulation of Cancer Progression and Antitumor Immunity. Int J Mol Sci 2022; 23:ijms231810494. [PMID: 36142404 PMCID: PMC9501080 DOI: 10.3390/ijms231810494] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Pyroptosis is a type of programmed cell death (PCD) accompanied by an inflammatory reaction and the rupture of a membrane. Pyroptosis is divided into a canonical pathway triggered by caspase-1, and a non-canonical pathway independent of caspase-1. More and more pyroptosis-related participants, pathways, and regulatory mechanisms have been exploited in recent years. Pyroptosis plays crucial roles in the initiation, progression, and metastasis of cancer and it affects the immunotherapeutic outcome by influencing immune cell infiltration as well. Extensive studies are required to elucidate the molecular mechanisms between pyroptosis and cancer. In this review, we introduce the discovery history of pyroptosis, delineate the signaling pathways of pyroptosis, and then make comparisons between pyroptosis and other types of PCD. Finally, we provide an overview of pyroptosis in different cancer types. With the progression in the field of pyroptosis, new therapeutic targets and strategies can be explored to combat cancer.
Collapse
Affiliation(s)
- Sihan Qi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qian Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
- Correspondence:
| |
Collapse
|
58
|
Zhang H, Xu X, Xu R, Ye T. Drug repurposing of ivermectin abrogates neutrophil extracellular traps and prevents melanoma metastasis. Front Oncol 2022; 12:989167. [PMID: 36132145 PMCID: PMC9484526 DOI: 10.3389/fonc.2022.989167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Neutrophil extracellular traps (NETs) have recently been identified to play a crucial role in cancer metastasis. However, the therapeutic target in NETs of melanoma cancer metastasis is still unknown. In this work, we screened a collection of 231 small molecule compounds. We identified ivermectin (IVM), a widely used antiparasitic drug, significantly inhibits neutrophil extracellular traps (NETs) formation after cathepsin B (CTSB) treatment. In vivo, IVM treatment showed no effects of melanoma tumor growth, while the orthotopic melanoma to lung metastasis was significantly suppressed by IVM. Serum level of myeloperoxidase-DNA and neutrophil elastase-DNA were suppressed after IVM treatment. Tumor infiltrated myeloid-derived suppressor cells (MDSCs) were significantly suppressed while tumor infiltrated CD8+T cells in lung was increased after IVM treatment in mouse melanoma model. Mechanistically, IVM targeted a pyroptotic driving factor gasdermin D (GSDMD), and exhibited a Kd of 267.96 nM by microscale thermophoresis (MST) assay. Furthermore, the direct interaction of IVM and GSDMD significantly suppressed GSDMD oligomerization, which are essential for GSDMD-dependent NETs formation. In vitro, treatment with CTSB in bone marrow neutrophils significantly promotes NETs formation, and the release of extracellular DNA was significantly suppressed by IVM pretreatment. Collectively, our results reveal that with the regulation role of IVM in neutrophils and NETs, IVM may potentially be used as a viable therapeutic approach for the treatment of melanoma cancer metastasis.
Collapse
Affiliation(s)
- Hongjun Zhang
- Department of Ophthalmology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - XiaoZhu Xu
- Department of Quality Arbitration, Shanghai Institute of Biological Products, Shanghai, China
| | - Rui Xu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Rui Xu, ; Tao Ye,
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
- *Correspondence: Rui Xu, ; Tao Ye,
| |
Collapse
|
59
|
Korn V, Pluhackova K. Not sorcery after all: Roles of multiple charged residues in membrane insertion of gasdermin-A3. Front Cell Dev Biol 2022; 10:958957. [PMID: 36120563 PMCID: PMC9479151 DOI: 10.3389/fcell.2022.958957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Gasdermins execute programmatory cell death, known as pyroptosis, by forming medium-sized membrane pores. Recently, the molecular structure of those pores as well as the diversity in their shape and size have been revealed by cryoTEM and atomic force microscopy, respectively. Even though a growth of smaller to larger oligomers and reshaping from slits to rings could be documented, the initiation of the gasdermin pore formation remains a mystery. In one hypothesis, gasdermin monomers insert into membranes before associating into oligomeric pores. In the other hypothesis, gasdermin oligomers preassemble on the membrane surface prior to membrane insertion. Here, by studying the behavior of monomeric membrane-inserted gasdermin-A3 (GSDMA3), we unveil that a monomeric gasdermin prefers the membrane-adsorbed over the membrane-inserted state. Our results thus support the hypothesis of oligomers preassembling on the membrane surface before membrane penetration. At the same time, our simulations of small membrane-inserted arcs of GSDMA3 suggest that the inserting oligomer can be small and does not have to comprise a full ring of approximately 26-30 subunits. Moreover, our simulations have revealed an astonishingly large impact of salt-bridge formation and protein surroundings on the transmembrane passage of charged residues, reducing the energetic cost by up to 53% as compared to their free forms. The here observed free energy barrier of mere 5.6 kcal/mol for the membrane insertion of monomeric GSDMA3 explains the surprising ability of gasdermins to spontaneously self-insert into cellular membranes.
Collapse
Affiliation(s)
| | - Kristyna Pluhackova
- Stuttgart Center for Simulation Science, Cluster of Excellence EXC 2075, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
60
|
Harris J, Borg NA. The multifaceted roles of NLRP3-modulating proteins in virus infection. Front Immunol 2022; 13:987453. [PMID: 36110852 PMCID: PMC9468583 DOI: 10.3389/fimmu.2022.987453] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune response to viruses is critical for the correct establishment of protective adaptive immunity. Amongst the many pathways involved, the NLRP3 [nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3)] inflammasome has received considerable attention, particularly in the context of immunity and pathogenesis during infection with influenza A (IAV) and SARS-CoV-2, the causative agent of COVID-19. Activation of the NLRP3 inflammasome results in the secretion of the proinflammatory cytokines IL-1β and IL-18, commonly coupled with pyroptotic cell death. While this mechanism is protective and key to host defense, aberrant NLRP3 inflammasome activation causes a hyperinflammatory response and excessive release of cytokines, both locally and systemically. Here, we discuss key molecules in the NLRP3 pathway that have also been shown to have significant roles in innate and adaptive immunity to viruses, including DEAD box helicase X-linked (DDX3X), vimentin and macrophage migration inhibitory factor (MIF). We also discuss the clinical opportunities to suppress NLRP3-mediated inflammation and reduce disease severity.
Collapse
Affiliation(s)
- James Harris
- Cell Biology Assays Team, Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Natalie A. Borg
- Immunity and Immune Evasion Laboratory, Chronic Infectious and Inflammatory Diseases Research, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
61
|
IL-18 Signaling Is Essential for Causing Streptococcal Toxic Shock-like Syndrome (STSLS). Life (Basel) 2022; 12:life12091324. [PMID: 36143361 PMCID: PMC9503922 DOI: 10.3390/life12091324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/09/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis (S. suis) is an emerging zoonotic pathogen that can cause multiple diseases, including streptococcal toxic shock-like syndrome (STSLS). The S. suis SC-19 strain could cause NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome hyperactivation, then induce a cytokine storm and STSLS. Although IL-18 is the downstream effector of NLRP3 signaling, the role of IL-18 signaling on STSLS remains to be elucidated. Thus, il18r1 gene knockout mice were constructed and challenged with the SC-19 strain. Alleviated clinical signs and tissue damages, as well as improved survival were observed in il18r−/− mice compared with the WT mice post-SC-19 challenge. Meanwhile, an obvious decrease in the inflammatory cytokine levels in blood was observed in the il18r-/- mice infected with SC-19. Therefore, IL-18, the downstream effector of NLRP3 inflammasome activation, was responsible for the cytokine storm and STSLS development caused by S. suis, suggesting that IL-18/IL-18Rα signaling could serve as a new target for STSLS.
Collapse
|
62
|
Jiao F, Dehez F, Ni T, Yu X, Dittman JS, Gilbert R, Chipot C, Scheuring S. Perforin-2 clockwise hand-over-hand pre-pore to pore transition mechanism. Nat Commun 2022; 13:5039. [PMID: 36028507 PMCID: PMC9418332 DOI: 10.1038/s41467-022-32757-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Perforin-2 (PFN2, MPEG1) is a pore-forming protein that acts as a first line of defense in the mammalian immune system, rapidly killing engulfed microbes within the phagolysosome in macrophages. PFN2 self-assembles into hexadecameric pre-pore rings that transition upon acidification into pores damaging target cell membranes. Here, using high-speed atomic force microscopy (HS-AFM) imaging and line-scanning and molecular dynamics simulation, we elucidate PFN2 pre-pore to pore transition pathways and dynamics. Upon acidification, the pre-pore rings (pre-pore-I) display frequent, 1.8 s-1, ring-opening dynamics that eventually, 0.2 s-1, initiate transition into an intermediate, short-lived, ~75 ms, pre-pore-II state, inducing a clockwise pre-pore-I to pre-pore-II propagation. Concomitantly, the first pre-pore-II subunit, undergoes a major conformational change to the pore state that propagates also clockwise at a rate ~15 s-1. Thus, the pre-pore to pore transition is a clockwise hand-over-hand mechanism that is accomplished within ~1.3 s. Our findings suggest a clockwise mechanism of membrane insertion that with variations may be general for the MACPF/CDC superfamily.
Collapse
Affiliation(s)
- Fang Jiao
- Department of Anesthesiology, Weill Cornell Medicine, New York City, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA.
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China.
| | - François Dehez
- Laboratoire International Associé, Centre National de la Recherche Scientifique et University of Illinois at Urbana-Champaign, Unité Mixte de Recherche no 7019, Université de Lorraine, Vandœuvre-lès-Nancy cedex, France
| | - Tao Ni
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Xiulian Yu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Calleva Research Centre for Evolution and Human Sciences, Magdalen College, University of Oxford, Oxford, UK
| | - Jeremy S Dittman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Robert Gilbert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Calleva Research Centre for Evolution and Human Sciences, Magdalen College, University of Oxford, Oxford, UK
| | - Christophe Chipot
- Laboratoire International Associé, Centre National de la Recherche Scientifique et University of Illinois at Urbana-Champaign, Unité Mixte de Recherche no 7019, Université de Lorraine, Vandœuvre-lès-Nancy cedex, France
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Simon Scheuring
- Department of Anesthesiology, Weill Cornell Medicine, New York City, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
63
|
Weindel CG, Martinez EL, Zhao X, Mabry CJ, Bell SL, Vail KJ, Coleman AK, VanPortfliet JJ, Zhao B, Wagner AR, Azam S, Scott HM, Li P, West AP, Karpac J, Patrick KL, Watson RO. Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell 2022; 185:3214-3231.e23. [PMID: 35907404 PMCID: PMC9531054 DOI: 10.1016/j.cell.2022.06.038] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/05/2022] [Accepted: 06/18/2022] [Indexed: 10/16/2022]
Abstract
Although mutations in mitochondrial-associated genes are linked to inflammation and susceptibility to infection, their mechanistic contributions to immune outcomes remain ill-defined. We discovered that the disease-associated gain-of-function allele Lrrk2G2019S (leucine-rich repeat kinase 2) perturbs mitochondrial homeostasis and reprograms cell death pathways in macrophages. When the inflammasome is activated in Lrrk2G2019S macrophages, elevated mitochondrial ROS (mtROS) directs association of the pore-forming protein gasdermin D (GSDMD) to mitochondrial membranes. Mitochondrial GSDMD pore formation then releases mtROS, promoting a switch to RIPK1/RIPK3/MLKL-dependent necroptosis. Consistent with enhanced necroptosis, infection of Lrrk2G2019S mice with Mycobacterium tuberculosis elicits hyperinflammation and severe immunopathology. Our findings suggest a pivotal role for GSDMD as an executer of multiple cell death pathways and demonstrate that mitochondrial dysfunction can direct immune outcomes via cell death modality switching. This work provides insights into how LRRK2 mutations manifest or exacerbate human diseases and identifies GSDMD-dependent necroptosis as a potential target to limit Lrrk2G2019S-mediated immunopathology.
Collapse
Affiliation(s)
- Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Eduardo L Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Xiao Zhao
- Department of Molecular and Cellular Medicine, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Cory J Mabry
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA; Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Krystal J Vail
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Aja K Coleman
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Jordyn J VanPortfliet
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Baoyu Zhao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Allison R Wagner
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Sikandar Azam
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Haley M Scott
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA.
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
64
|
Yu Z, Xiao Z, Guan L, Bao P, Yu Y, Liang Y, Li M, Huang Z, Chen X, Chen R, Su Y, Ge J. Translocation of gasdermin D induced mitochondrial injury and mitophagy mediated quality control in lipopolysaccharide related cardiomyocyte injury. Clin Transl Med 2022; 12:e1002. [PMID: 36030524 PMCID: PMC9420421 DOI: 10.1002/ctm2.1002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUNDS Inflammation underlies the mechanism of different kinds of heart disease. Cytoplasmic membrane localized N-terminal fragment of gasdermin-D (GSDMD-N) could induce inflammatory injury to cardiomyocyte. However, effects and dynamic changes of GSDMD during the process of lipopolysaccharide (LPS) related inflammatory stress induced cardiomyocyte injury are barely elucidated to date. In this study, LPS related cardiomyocyte injury was investigated based on potential interaction of GSDMD-N induced mitochondrial injury and mitophagy mediated mitochondria quality control. METHODS HL-1 cardiomyocytes were treated with LPS and Nigericin to induce inflammatory stress. The dual-fluorescence-labelled GSDMD expressed HL-1 cardiomyocytes were constructed to study the translocation of GSDMD. The mitochondrial membrane potential (MMP) was measured by JC-1 staining. Mitophagy and autophagic flux were recorded by transmission electron microscopy and fluorescent image. RESULTS GSDMD-N showed a time-dependent pattern of translocation from mitochondria to cytoplasmic membrane under LPS and Nigericin induced inflammatory stress in HL-1 cardiomyocytes. GSDMD-N preferred to localize to mitochondria to permeablize its membrane and dissipate the MMP. This effect couldn't be reversed by cyclosporine-A (mPTP inhibitor), indicating GSDMD-N pores as alternative mechanism underlying MMP regulation, in addition to mitochondrial permeability transition pore (mPTP). Moreover, the combination between GSDMD-N and autophagy related Microtubule Associated Protein 1 Light Chain 3 Beta (LC3B) was verified by co-immunoprecipitation. Besides, mitophagy alleviating GSDMD-N induced mitochondrial injury was proved by pre-treatment of autophagy antagonist or agonist in GSDMD-knock out or GSDMD-overexpression cells. A time-dependent pattern of GSDMD translocation and mitochondrial GSDMD targeted mitophagy were verified. CONCLUSION Herein, our study confirmed a crosstalk between GSDMD-N induced mitochondrial injury and mitophagy mediated mitochondria quality control during LPS related inflammation induced cardiomyocyte injury, which potentially facilitating the development of therapeutic target to myocardial inflammatory disease. Our findings support pharmaceutical intervention on enhancing autophagy or inhibiting GSDMD as potential target for inflammatory heart disease treatment.
Collapse
Affiliation(s)
- Ziqing Yu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zilong Xiao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Graduate School, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lichun Guan
- Department of Cardiovascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, P. R. China
| | - Pei Bao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Graduate School, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yong Yu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yixiu Liang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Minghui Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhenzhen Huang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Xueying Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Ruizhen Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yangang Su
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
65
|
Liang F, Qin W, Zeng Y, Wang D. Modulation of Autoimmune and Autoinflammatory Diseases by Gasdermins. Front Immunol 2022; 13:841729. [PMID: 35720396 PMCID: PMC9199384 DOI: 10.3389/fimmu.2022.841729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/05/2022] [Indexed: 12/29/2022] Open
Abstract
Autoimmune diseases and autoinflammatory diseases are two types of the immune system disorders. Pyroptosis, a highly inflammatory cell death, plays an important role in diseases of immune system. The gasdermins belong to a pore-forming protein gene family which are mainly expressed in immune cells, gastrointestinal tract, and skin. Gasdermins are regarded as an executor of pyroptosis and have been shown to possess various cellular functions and pathological effects such as pro-inflammatory, immune activation, mediation of tumor, etc. Except for infectious diseases, the vital role of gasdermins in autoimmune diseases, autoinflammatory diseases, and immune-related neoplastic diseases has been proved recently. Therefore, gasdermins have been served as a potential therapeutic target for immune disordered diseases. The review summarizes the basic molecular structure and biological function of gasdermins, mainly discusses their role in autoimmune and autoinflammatory diseases, and highlights the recent research on gasdermin family inhibitors so as to provide potential therapeutic prospects.
Collapse
Affiliation(s)
- Fang Liang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weixiao Qin
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yilan Zeng
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dan Wang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
66
|
Zhang S, Liang Y, Yao J, Li DF, Wang LS. Role of Pyroptosis in Inflammatory Bowel Disease (IBD): From Gasdermins to DAMPs. Front Pharmacol 2022; 13:833588. [PMID: 35677444 PMCID: PMC9168461 DOI: 10.3389/fphar.2022.833588] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/22/2022] [Indexed: 12/11/2022] Open
Abstract
Pyroptosis is a pro-inflammatory cell death executed by gasdermin family proteins that involve the formation of pores on cells, recognition of danger signals, and release of pro-inflammatory cytokines IL-1β and IL-18. Pyroptosis modulates mucosal innate immunity and enteropathogenic bacterial infection. Similarly, the gasdermin family has been reported to be involved in the defense of the intestinal epithelium against bacterial infection and in the regulation of intestinal inflammation. Pyroptosis initiates damage signals that activate multiple pathways to cause inflammation, which may be a potential cause of chronic intestinal inflammation. In this review, we discuss the impact of pyroptosis on inflammatory bowel disease (IBD), with a focus on the executive proteins of pyroptosis (GSDMB, GADMD, and GSDME) and IBD-related endogenous damage-associated molecular patterns (DAMPs) produced by pyroptosis.
Collapse
Affiliation(s)
- Shuxia Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen People Hospital, Shenzhen, China
| | | | - Jun Yao
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - De-Feng Li
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Li-Sheng Wang
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| |
Collapse
|
67
|
Gan C, Cai Q, Tang C, Gao J. Inflammasomes and Pyroptosis of Liver Cells in Liver Fibrosis. Front Immunol 2022; 13:896473. [PMID: 35707547 PMCID: PMC9189314 DOI: 10.3389/fimmu.2022.896473] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammasomes are multiprotein complexes that can sense danger signals and activate caspase-1 to mediate pro-inflammatory cytokines release and pyroptotic cell death. There are two main canonical and non-canonical signaling pathways that trigger inflammasome activation. Inflammasomes are expressed and assembled in parenchymal and nonparenchymal cells in response to liver injury in the liver. Additionally, the hepatocytes, biliary epithelial cells (cholangiocytes), hepatic stellate cells (HSCs), hepatic macrophages, and liver sinusoidal endothelial cells (LSECs) contribute to liver fibrosis via different mechanisms. However, the underlying mechanism of the inflammasome and pyroptosis in these liver cells in liver fibrosis remains elusive. This review summarizes the activation and function of inflammasome complexes and then discusses the association between inflammasomes, pyroptosis, and liver fibrosis. Unlike other similar reviewers, we will focus on the effect of inflammasome activation and pyroptosis in the various liver cells during the development of liver fibrosis. We will also highlight the latest progress of pharmacological intervention in inflammasome-mediated liver fibrosis.
Collapse
Affiliation(s)
- Can Gan
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyu Cai
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jinhang Gao, ; ; Chengwei Tang,
| | - Jinhang Gao
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jinhang Gao, ; ; Chengwei Tang,
| |
Collapse
|
68
|
Liu J, Chen J, Xu B, Lin L, Liu S, Ma X, Liu J. 3,4,5-O-tricaffeoylquinic acid with anti-radiation activity suppresses LPS-induced NLRP3 inflammasome activation via autophagy in THP-1 macrophages. Mol Immunol 2022; 147:187-198. [PMID: 35633614 DOI: 10.1016/j.molimm.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/09/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Damage to normal tissues caused by excessive ionizing radiation (IR) exposure is the major side effect of radiotherapy. Several recent studies have shown that IR-induced damage to tissues leads to a systemic immune response and NLRP3 inflammasome activation in immune cells. 3,4,5-O-tricaffeoylquinic acid (tCQA), extracted from the natural plant Azolla imbricata, relieves inflammation and has radioprotective function. Here, we aimed to investigate the inhibitory effect and molecular mechanism of tCQA on IR-induced NLRP3 inflammasome activation. First, the results of ELISA and qPCR assays showed that tCQA has anti-inflammatory effects in THP-1 cell line and healthy human peripheral blood mononuclear cells. Western blotting and ELISA suggested tCQA could inhibit NF-κB/MAPK signaling pathway, NLRP3 expression and the secretion of IL-1β in lipopolysaccharide (LPS)-stimulated THP-1 macrophages. Then, flow cytometry, LDH assay and western blotting demonstrated that tCQA could inhibit LPS- and nigericin-induced Caspase-1 activation and gasdermin D cleavage, thereby suppressing inflammatory cell death. Furthermore, we found that the autophagy inhibitor chloroquine, not the proteasome inhibitor MG132, could counteract the promoting effect of tCQA on NLRP3 degradation and the inhibitory effect on cell death. Western blotting and autophagosome staining results suggested tCQA could significantly enhance LPS-induced autophagic flux in macrophages and ATG5/ATG7 knockdown reverses the inhibitory effect of tCQA on NLRP3 expression and Caspase-1 activation, indicating that tCQA induces NLRP3 degradation via autophagy. Finally, THP-1 macrophages and BALB/c mice were irradiated with 137Cs γ-rays and tCQA could inhibit IR-induced NLRP3 inflammasome activation both in vitro and in vivo. To conclude, tCQA controls inflammation and NLRP3 inflammasome activation in vitro via NF-κB/MAPK signaling pathway and autophagy, meanwhile inhibits IR-induced NLRP3 inflammasome activation in vivo. Overall, our study provides an experimental and theoretical basis for the application of tCQA as a radioprotectant in clinical radiotherapy.
Collapse
Affiliation(s)
- Jiajun Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jingyun Chen
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Baixue Xu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Long Lin
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shaoqun Liu
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hosipital & AHS, Fudan University, Shanghai, 201199, PR China; Department of Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, PR China.
| | - Xiaoying Ma
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
69
|
Mari SA, Pluhackova K, Pipercevic J, Leipner M, Hiller S, Engel A, Müller DJ. Gasdermin-A3 pore formation propagates along variable pathways. Nat Commun 2022; 13:2609. [PMID: 35545613 PMCID: PMC9095878 DOI: 10.1038/s41467-022-30232-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 04/22/2022] [Indexed: 12/31/2022] Open
Abstract
Gasdermins are main effectors of pyroptosis, an inflammatory form of cell death. Released by proteolysis, the N-terminal gasdermin domain assembles large oligomers to punch lytic pores into the cell membrane. While the endpoint of this reaction, the fully formed pore, has been well characterized, the assembly and pore-forming mechanisms remain largely unknown. To resolve these mechanisms, we characterize mouse gasdermin-A3 by high-resolution time-lapse atomic force microscopy. We find that gasdermin-A3 oligomers assemble on the membrane surface where they remain attached and mobile. Once inserted into the membrane gasdermin-A3 grows variable oligomeric stoichiometries and shapes, each able to open transmembrane pores. Molecular dynamics simulations resolve how the membrane-inserted amphiphilic β-hairpins and the structurally adapting hydrophilic head domains stabilize variable oligomeric conformations and open the pore. The results show that without a vertical collapse gasdermin pore formation propagates along a set of multiple parallel but connected reaction pathways to ensure a robust cellular response.
Collapse
Affiliation(s)
- Stefania A Mari
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland
| | - Kristyna Pluhackova
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland.
| | | | - Matthew Leipner
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland
| | | | - Andreas Engel
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland.
| |
Collapse
|
70
|
Sala R, Rioja-Blanco E, Serna N, Sánchez-García L, Álamo P, Alba-Castellón L, Casanova I, López-Pousa A, Unzueta U, Céspedes MV, Vázquez E, Villaverde A, Mangues R. GSDMD-dependent pyroptotic induction by a multivalent CXCR4-targeted nanotoxin blocks colorectal cancer metastases. Drug Deliv 2022; 29:1384-1397. [PMID: 35532120 PMCID: PMC9090371 DOI: 10.1080/10717544.2022.2069302] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer (CRC) remains the third cause of cancer-related mortality in Western countries, metastases are the main cause of death. CRC treatment remains limited by systemic toxicity and chemotherapy resistance. Therefore, nanoparticle-mediated delivery of cytotoxic agents selectively to cancer cells represents an efficient strategy to increase the therapeutic index and overcome drug resistance. We have developed the T22-PE24-H6 therapeutic protein-only nanoparticle that incorporates the exotoxin A from Pseudomonas aeruginosa to selectively target CRC cells because of its multivalent ligand display that triggers a high selectivity interaction with the CXCR4 receptor overexpressed on the surface of CRC stem cells. We here observed a CXCR4-dependent cytotoxic effect for T22-PE24-H6, which was not mediated by apoptosis, but instead capable of inducing a time-dependent and sequential activation of pyroptotic markers in CRC cells in vitro. Next, we demonstrated that repeated doses of T22-PE24-H6 inhibit tumor growth in a subcutaneous CXCR4+ CRC model, also through pyroptotic activation. Most importantly, this nanoparticle also blocked the development of lymphatic and hematogenous metastases, in a highly aggressive CXCR4+ SW1417 orthotopic CRC model, in the absence of systemic toxicity. This targeted drug delivery approach supports for the first time the clinical relevance of inducing GSDMD-dependent pyroptosis, a cell death mechanism alternative to apoptosis, in CRC models, leading to the selective elimination of CXCR4+ cancer stem cells, which are associated with resistance, metastases and anti-apoptotic upregulation.
Collapse
Affiliation(s)
- Rita Sala
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | - Elisa Rioja-Blanco
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | - Naroa Serna
- CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Institut de Biotecnologia I de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departament de Genètica I de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Sánchez-García
- CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Institut de Biotecnologia I de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departament de Genètica I de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Patricia Álamo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | - Lorena Alba-Castellón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | - Antonio López-Pousa
- CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Department of Medical Oncology, Hospital de la Santa Creu I Sant Pau, Barcelon, Spain
| | - Ugutz Unzueta
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| | | | - Esther Vázquez
- CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Institut de Biotecnologia I de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departament de Genètica I de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Antonio Villaverde
- CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Institut de Biotecnologia I de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departament de Genètica I de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Madrid, Spain.,Josep Carreras Research Institute, Barcelona, Spain
| |
Collapse
|
71
|
Tan C, Reilly B, Jha A, Murao A, Lee Y, Brenner M, Aziz M, Wang P. Active Release of eCIRP via Gasdermin D Channels to Induce Inflammation in Sepsis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2184-2195. [PMID: 35418465 PMCID: PMC9050887 DOI: 10.4049/jimmunol.2101004] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022]
Abstract
Extracellular cold-inducible RNA binding protein (eCIRP) is an inflammatory mediator that causes inflammation and tissue injury in sepsis. Gasdermin D (GSDMD) is a protein that, when cleaved, forms pores in the cell membrane, releasing intracellular contents into the extracellular milieu to exacerbate inflammation. We hypothesize that eCIRP is released actively from viable macrophages via GSDMD pores. We found that LPS induced eCIRP secretion from macrophages into the extracellular space. LPS significantly increased the expression of caspase-11 and cleavage of the GSDMD, as evidenced by increased N-terminal GSDMD expression in RAW 264.7 cells and mouse primary peritoneal macrophages. GSDMD inhibitor disulfiram decreased eCIRP release in vitro. Treatment with glycine to prevent pyroptosis-induced cell lysis did not significantly decrease eCIRP release from LPS-treated macrophages, indicating that eCIRP was actively released and was independent of pyroptosis. Downregulation of GSDMD gene expression by siRNA transfection suppressed eCIRP release in vitro after LPS stimulation. Moreover, GSDMD-/- peritoneal macrophages and mice had decreased levels of eCIRP in the culture supernatants and in blood treated with LPS in vitro and in vivo, respectively. GSDMD inhibitor disulfiram inhibited serum levels of eCIRP in endotoxemia and cecal ligation and puncture-induced sepsis. We conclude that eCIRP release from living macrophages is mediated through GSDMD pores, suggesting that targeting GSDMD could be a novel and potential therapeutic approach to inhibit eCIRP-mediated inflammation in sepsis.
Collapse
Affiliation(s)
- Chuyi Tan
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Bridgette Reilly
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Alok Jha
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY.,Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY; and
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; .,Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY; and.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; .,Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY; and.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| |
Collapse
|
72
|
McManus RM. The Role of Immunity in Alzheimer's Disease. Adv Biol (Weinh) 2022; 6:e2101166. [PMID: 35254006 DOI: 10.1002/adbi.202101166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/03/2022] [Indexed: 01/27/2023]
Abstract
With the increase in the aging population, age-related conditions such as dementia and Alzheimer's disease will become ever more prevalent in society. As there is no cure for dementia and extremely limited therapeutic options, researchers are examining the mechanisms that contribute to the progression of cognitive decline in hopes of developing better therapies and even an effective, long-lasting treatment for this devastating condition. This review will provide an updated perspective on the role of immunity in triggering the changes that lead to the development of dementia. It will detail the latest findings on Aβ- and tau-induced microglial activation, including the role of the inflammasome. The contribution of the adaptive immune system, specifically T cells, will be discussed. Finally, whether the innate and adaptive immune system can be modulated to protect against dementia will be examined, along with an assessment of the prospective candidates for these that are currently in clinical trials.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| |
Collapse
|
73
|
Bertheloot D, Wanderley CW, Schneider AH, Schiffelers LD, Wuerth JD, Tödtmann JM, Maasewerd S, Hawwari I, Duthie F, Rohland C, Ribeiro LS, Jenster LM, Rosero N, Tesfamariam YM, Cunha FQ, Schmidt FI, Franklin BS. Nanobodies dismantle post-pyroptotic ASC specks and counteract inflammation in vivo. EMBO Mol Med 2022; 14:e15415. [PMID: 35438238 PMCID: PMC9174887 DOI: 10.15252/emmm.202115415] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammasomes sense intracellular clues of infection, damage, or metabolic imbalances. Activated inflammasome sensors polymerize the adaptor ASC into micron‐sized “specks” to maximize caspase‐1 activation and the maturation of IL‐1 cytokines. Caspase‐1 also drives pyroptosis, a lytic cell death characterized by leakage of intracellular content to the extracellular space. ASC specks are released among cytosolic content, and accumulate in tissues of patients with chronic inflammation. However, if extracellular ASC specks contribute to disease, or are merely inert remnants of cell death remains unknown. Here, we show that camelid‐derived nanobodies against ASC (VHHASC) target and disassemble post‐pyroptotic inflammasomes, neutralizing their prionoid, and inflammatory functions. Notably, pyroptosis‐driven membrane perforation and exposure of ASC specks to the extracellular environment allowed VHHASC to target inflammasomes while preserving pre‐pyroptotic IL‐1β release, essential to host defense. Systemically administrated mouse‐specific VHHASC attenuated inflammation and clinical gout, and antigen‐induced arthritis disease. Hence, VHHASC neutralized post‐pyroptotic inflammasomes revealing a previously unappreciated role for these complexes in disease. VHHASC are the first biologicals that disassemble pre‐formed inflammasomes while preserving their functions in host defense.
Collapse
Affiliation(s)
- Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Carlos Ws Wanderley
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.,Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Ayda H Schneider
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.,Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Lisa Dj Schiffelers
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jennifer D Wuerth
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jan Mp Tödtmann
- Core Facility Nanobodies, Medical Faculty, University of Bonn, Bonn, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Fraser Duthie
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Cornelia Rohland
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lucas S Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yonas M Tesfamariam
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.,Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.,Core Facility Nanobodies, Medical Faculty, University of Bonn, Bonn, Germany
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
74
|
Demarco B, Danielli S, Fischer FA, Bezbradica JS. How Pyroptosis Contributes to Inflammation and Fibroblast-Macrophage Cross-Talk in Rheumatoid Arthritis. Cells 2022; 11:1307. [PMID: 35455985 PMCID: PMC9028325 DOI: 10.3390/cells11081307] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
About thirty years ago, a new form of pro-inflammatory lytic cell death was observed and termed pyroptosis. Only in 2015, gasdermins were defined as molecules that create pores at the plasma membrane and drive pyroptosis. Today, we know that gasdermin-mediated death is an important antimicrobial defence mechanism in bacteria, yeast and mammals as it destroys the intracellular niche for pathogen replication. However, excessive and uncontrolled cell death also contributes to immunopathology in several chronic inflammatory diseases, including arthritis. In this review, we discuss recent findings where pyroptosis contributes to tissue damage and inflammation with a main focus on injury-induced and autoimmune arthritis. We also review novel functions and regulatory mechanisms of the pyroptotic executors gasdermins. Finally, we discuss possible models of how pyroptosis may contribute to the cross-talk between fibroblast and macrophages, and also how this cross-talk may regulate inflammation by modulating inflammasome activation and pyroptosis induction.
Collapse
Affiliation(s)
- Benjamin Demarco
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; (S.D.); (F.A.F.)
| | | | | | - Jelena S. Bezbradica
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; (S.D.); (F.A.F.)
| |
Collapse
|
75
|
Regulation of the release of damage-associated molecular patterns from necroptotic cells. Biochem J 2022; 479:677-685. [PMID: 35293986 DOI: 10.1042/bcj20210604] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are molecules within living cells that are released when cell membranes are ruptured. Although DAMPs have physiological functions inside the cell, once DAMPs are released extracellularly, they elicit various biological responses, including inflammation, proliferation, tissue damage, and tissue repair, in a context-dependent manner. In past decades, it was assumed that the release of DAMPs was induced by a membrane rupture, caused by passive ATP depletion, or by chemical or mechanical damage to the membrane. However, that concept has been challenged by recent advancements in understanding the regulation of cell death. Necroptosis is a form of regulated cell death, where cells show necrotic morphology. Necroptosis is triggered by death receptors, toll-like receptors, and some viral infections. The membrane rupture is executed by the mixed lineage-like kinase domain-like pseudokinase (MLKL), which forms oligomers that translocate to the plasma membrane during necroptosis. Although the causal relationship between MLKL function and membrane rupture has been extensively investigated, the detailed molecular mechanisms by which oligomerized MLKL induces membrane rupture are not fully understood. This review summarizes recent advances in understanding how MLKL regulates DAMP release and new technologies for visualizing DAMP release at single-cell resolution.
Collapse
|
76
|
Gu L, Sun M, Li R, Zhang X, Tao Y, Yuan Y, Luo X, Xie Z. Didymin Suppresses Microglia Pyroptosis and Neuroinflammation Through the Asc/Caspase-1/GSDMD Pathway Following Experimental Intracerebral Hemorrhage. Front Immunol 2022; 13:810582. [PMID: 35154128 PMCID: PMC8828494 DOI: 10.3389/fimmu.2022.810582] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation has been proven to exert an important effect on brain injury after intracerebral hemorrhage (ICH). Previous studies reported that Didymin possessed anti-inflammatory properties after acute hepatic injury, hyperglycemia-induced endothelial dysfunction, and death. However, the role of Didymin in microglial pyroptosis and neuroinflammation after ICH is unclear. The current study aimed to investigate the effect of Didymin on neuroinflammation mediated by microglial pyroptosis in mouse models of ICH and shed some light on the underlying mechanisms. In this study, we observed that Didymin treatment remarkably improved neurobehavioral performance and decreased BBB disruption and brain water content. Microglial activation and neutrophil infiltration in the peri-hematoma tissue after ICH were strikingly mitigated by Didymin as well. At the molecular level, administration of Didymin significantly unregulated the expression of Rkip and downregulated the expression of pyroptotic molecules and inflammatory cytokines such as Nlrp3 inflammasome, GSDMD, caspase-1, and mature IL-1β, TNF-α, and MPO after ICH. Besides, Didymin treatment decreased the number of Caspase-1-positive microglia and GSDMD-positive microglia after ICH. Inversely, Locostatin, an Rkip-specific inhibitor, significantly abolished the anti-pyroptosis and anti-neuroinflammation effects of Didymin. Moreover, Rkip binding with Asc could interrupt the activation and assembly of the inflammasome. Mechanistically, inhibition of Caspase-1 by VX-765 attenuated brain injury and suppressed microglial pyroptosis and neuroinflammation by downregulation of GSDMD, mature IL-1β, TNF-α, and MPO based on Locostatin-treated ICH. Taken together, Didymin alleviated microglial pyroptosis and neuroinflammation, at least in part through the Asc/Caspase-1/GSDMD pathway via upregulating Rkip expression after ICH. Therefore, Didymin may be a potential agent to attenuate neuroinflammation via its anti-pyroptosis effect after ICH.
Collapse
Affiliation(s)
- Lingui Gu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingjiang Sun
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ruihao Li
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xingyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ye Yuan
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xu Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
77
|
Mechanistic Insights into Gasdermin Pore Formation and Regulation in Pyroptosis. J Mol Biol 2022; 434:167297. [PMID: 34627790 PMCID: PMC8844191 DOI: 10.1016/j.jmb.2021.167297] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
The gasdermin family is a newly identified class of pore-forming proteins that play as the executioners of pyroptosis, a lytic pro-inflammatory type of cell death triggered by sensing cytosolic infections and danger signals. Upon activation, the gasdermin N-terminal domain translocates to the cell membrane to form pores, which allow the release of proinflammatory cytokines and alarmins, and cause cell lysis. Many structural studies have been conducted in the past few years to investigate the mechanisms of gasdermin proteins in the activation and pore formation. Here, we review these high-resolution structures and highlight the mechanistic insights into the gasdermin activation and regulation that are provided.
Collapse
|
78
|
Ivanova ME, Lukoyanova N, Malhotra S, Topf M, Trapani JA, Voskoboinik I, Saibil HR. The pore conformation of lymphocyte perforin. SCIENCE ADVANCES 2022; 8:eabk3147. [PMID: 35148176 PMCID: PMC8836823 DOI: 10.1126/sciadv.abk3147] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/17/2021] [Indexed: 05/05/2023]
Abstract
Perforin is a pore-forming protein that facilitates rapid killing of pathogen-infected or cancerous cells by the immune system. Perforin is released from cytotoxic lymphocytes, together with proapoptotic granzymes, to bind to a target cell membrane where it oligomerizes and forms pores. The pores allow granzyme entry, which rapidly triggers the apoptotic death of the target cell. Here, we present a 4-Å resolution cryo-electron microscopy structure of the perforin pore, revealing previously unidentified inter- and intramolecular interactions stabilizing the assembly. During pore formation, the helix-turn-helix motif moves away from the bend in the central β sheet to form an intermolecular contact. Cryo-electron tomography shows that prepores form on the membrane surface with minimal conformational changes. Our findings suggest the sequence of conformational changes underlying oligomerization and membrane insertion, and explain how several pathogenic mutations affect function.
Collapse
Affiliation(s)
- Marina E. Ivanova
- Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet St, London WC1E 7HX, UK
- Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Natalya Lukoyanova
- Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet St, London WC1E 7HX, UK
| | - Sony Malhotra
- Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet St, London WC1E 7HX, UK
- Scientific Computing Department, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Fermi Ave, Harwell, Didcot OX11 0QX, UK
| | - Maya Topf
- Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet St, London WC1E 7HX, UK
- Centre for Structural Systems Biology, Leibniz-Institut für Experimentelle Virologie and Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joseph A. Trapani
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Ilia Voskoboinik
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Helen R. Saibil
- Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet St, London WC1E 7HX, UK
| |
Collapse
|
79
|
AI-based atomic force microscopy image analysis allows to predict electrochemical impedance spectra of defects in tethered bilayer membranes. Sci Rep 2022; 12:1127. [PMID: 35064137 PMCID: PMC8783026 DOI: 10.1038/s41598-022-04853-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/24/2021] [Indexed: 01/08/2023] Open
Abstract
Atomic force microscopy (AFM) image analysis of supported bilayers, such as tethered bilayer membranes (tBLMs) can reveal the nature of the membrane damage by pore-forming proteins and predict the electrochemical impedance spectroscopy (EIS) response of such objects. However, automated analysis involving pore detection in such images is often non-trivial and can require AI-based object detection techniques. The specific object-detection algorithm we used to determine the defect coordinates in real AFM images was a convolutional neural network (CNN). Defect coordinates allow to predict the EIS response of tBLMs populated by the pore-forming toxins using finite element analysis (FEA) modeling. We tested if the accuracy of the CNN algorithm affected the EIS spectral features sensitive to defect densities and other physical parameters of tBLMs. We found that the EIS spectra can be predicted sufficiently well, however, systematic errors of characteristic spectral points were observed and need to be taken into account. Importantly, the comparison of predicted EIS curves with experimental ones allowed to estimate important physical parameters of tBLMs such as the specific resistance of submembrane reservoir. This reservoir separates phospholipid bilayer from the solid support. We found that the specific resistance of the reservoir amounts to \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$10^{4.25 \pm 0.10}$$\end{document}104.25±0.10\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\Omega \cdot cm$$\end{document}Ω·cm which is approximately two orders of a magnitude higher compared to the specific resistance of the buffer bathing tBLMs studied in this work. We hypothesize that such effect may be related in part due to decreased concentration of ionic carriers in the submembrane due to decreased relative dielectric permittivity in this region.
Collapse
|
80
|
Santa Cruz Garcia AB, Schnur KP, Malik AB, Mo GCH. Gasdermin D pores are dynamically regulated by local phosphoinositide circuitry. Nat Commun 2022; 13:52. [PMID: 35013201 PMCID: PMC8748731 DOI: 10.1038/s41467-021-27692-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Gasdermin D forms large, ~21 nm diameter pores in the plasma membrane to drive the cell death program pyroptosis. These pores are thought to be permanently open, and the resultant osmotic imbalance is thought to be highly damaging. Yet some cells mitigate and survive pore formation, suggesting an undiscovered layer of regulation over the function of these pores. However, no methods exist to directly reveal these mechanistic details. Here, we combine optogenetic tools, live cell fluorescence biosensing, and electrophysiology to demonstrate that gasdermin pores display phosphoinositide-dependent dynamics. We quantify repeated and fast opening-closing of these pores on the tens of seconds timescale, visualize the dynamic pore geometry, and identify the signaling that controls dynamic pore activity. The identification of this circuit allows pharmacological tuning of pyroptosis and control of inflammatory cytokine release by living cells.
Collapse
Affiliation(s)
| | - Kevin P Schnur
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA.,Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Gary C H Mo
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA. .,Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
81
|
Plasma membrane perforation by GSDME during apoptosis-driven secondary necrosis. Cell Mol Life Sci 2021; 79:19. [PMID: 34971436 PMCID: PMC8720079 DOI: 10.1007/s00018-021-04078-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022]
Abstract
Secondary necrosis has long been perceived as an uncontrolled process resulting in total lysis of the apoptotic cell. Recently, it was shown that progression of apoptosis to secondary necrosis is regulated by Gasdermin E (GSDME), which requires activation by caspase-3. Although the contribution of GSDME in this context has been attributed to its pore-forming capacity, little is known about the kinetics and size characteristics of this. Here we report on the membrane permeabilizing features of GSDME by monitoring the influx and efflux of dextrans of different sizes into/from anti-Fas-treated L929sAhFas cells undergoing apoptosis-driven secondary necrosis. We found that GSDME accelerates cell lysis measured by SYTOX Blue staining but does not affect the exposure of phosphatidylserine on the plasma membrane. Furthermore, loss of GSDME expression clearly hampered the influx of fluorescently labeled dextrans while the efflux happened independently of the presence or absence of GSDME expression. Importantly, both in- and efflux of dextrans were dependent on their molecular weight. Altogether, our results demonstrate that GSDME regulates the passage of compounds together with other plasma membrane destabilizing subroutines.
Collapse
|
82
|
Wang S, Liu Y, Zhang L, Sun Z. Methods for monitoring cancer cell pyroptosis. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0504. [PMID: 34931767 PMCID: PMC9088190 DOI: 10.20892/j.issn.2095-3941.2021.0504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/01/2021] [Indexed: 11/11/2022] Open
Abstract
Pyroptosis is a form of proinflammatory cell death that depends on the gasdermin family of proteins. The main features of pyroptosis are altered membrane permeability, cell swelling, membrane rupture, and the ability to mobilize a strong immune response. The relationship between pyroptosis and cancer has become a popular topic in immunological research. Multiple strategies for inducing pyroptosis in cancer cells have been developed for cancer therapy, including chemotherapy, small molecule drugs, and nanomedicines. In this review, we systematically discuss recent advances in research on the mechanisms of pyroptosis, and compare pyroptosis with apoptosis and necroptosis from several aspects. The development of various experimental systems has accompanied rapid progress in this field, but little consensus on monitoring pyroptosis is currently available. We focus on techniques commonly used to monitor pyroptosis, and describe future techniques that may be used to increase our knowledge in this field. Overall, the advancement of pyroptosis detection methods will help researchers to better investigate the relationships between pyroptosis and various cancers, and should provide insights into the use of these promising tools for cancer treatments.
Collapse
Affiliation(s)
- Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yuantong Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhijun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
83
|
Molecular and structural aspects of gasdermin family pores and insights into gasdermin-elicited programmed cell death. Biochem Soc Trans 2021; 49:2697-2710. [PMID: 34812891 PMCID: PMC8786298 DOI: 10.1042/bst20210672] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/04/2023]
Abstract
Pyroptosis is a highly inflammatory and lytic type of programmed cell death (PCD) commenced by inflammasomes, which sense perturbations in the cytosolic environment. Recently, several ground-breaking studies have linked a family of pore-forming proteins known as gasdermins (GSDMs) to pyroptosis. The human genome encodes six GSDM proteins which have a characteristic feature of forming pores in the plasma membrane resulting in the disruption of cellular homeostasis and subsequent induction of cell death. GSDMs have an N-terminal cytotoxic domain and an auto-inhibitory C-terminal domain linked together through a flexible hinge region whose proteolytic cleavage by various enzymes releases the N-terminal fragment that can insert itself into the inner leaflet of the plasma membrane by binding to acidic lipids leading to pore formation. Emerging studies have disclosed the involvement of GSDMs in various modalities of PCD highlighting their role in diverse cellular and pathological processes. Recently, the cryo-EM structures of the GSDMA3 and GSDMD pores were resolved which have provided valuable insights into the pore formation process of GSDMs. Here, we discuss the current knowledge regarding the role of GSDMs in PCD, structural and molecular aspects of autoinhibition, and pore formation mechanism followed by a summary of functional consequences of gasdermin-induced membrane permeabilization.
Collapse
|
84
|
Benton JT, Bayly-Jones C. Challenges and approaches to studying pore-forming proteins. Biochem Soc Trans 2021; 49:2749-2765. [PMID: 34747994 PMCID: PMC8892993 DOI: 10.1042/bst20210706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Pore-forming proteins (PFPs) are a broad class of molecules that comprise various families, structural folds, and assembly pathways. In nature, PFPs are most often deployed by their host organisms to defend against other organisms. In humans, this is apparent in the immune system, where several immune effectors possess pore-forming activity. Furthermore, applications of PFPs are found in next-generation low-cost DNA sequencing, agricultural crop protection, pest control, and biosensing. The advent of cryoEM has propelled the field forward. Nevertheless, significant challenges and knowledge-gaps remain. Overcoming these challenges is particularly important for the development of custom, purpose-engineered PFPs with novel or desired properties. Emerging single-molecule techniques and methods are helping to address these unanswered questions. Here we review the current challenges, problems, and approaches to studying PFPs.
Collapse
Affiliation(s)
- Joshua T. Benton
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Charles Bayly-Jones
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
85
|
Liu X, Ding S, Liu P. The Roles of Gasdermin D in Coronavirus Infection and Evasion. Front Microbiol 2021; 12:784009. [PMID: 34899666 PMCID: PMC8662355 DOI: 10.3389/fmicb.2021.784009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/22/2021] [Indexed: 11/17/2022] Open
Abstract
Pyroptosis is lytic, programmed cell death and plays a critical role against microbial invasion, functioning as an innate immune effector mechanism. The pore-forming protein gasdermin D (GSDMD), a member of gasdermin family proteins, is a primary effector of pyroptosis. The cleavage of inflammasome-associated inflammatory caspases activates GSDMD to liberate the N-terminal effector domain from the C-terminal inhibitory domain and form pores in the cellular plasma membrane. Emerging evidence shows that the pore-forming activity of GSDMD beyond pyroptosis and modifies non-lytic cytosolic protein secretion in living cells and innate immunity. While the essential roles of GSDMD in bacterial infection and cancer have been widely investigated, the importance of GSDMD in virus infection, including coronaviruses, remains elusive. Here, we review the current literature regarding the activation and functions of GSDMD during virus infections. Last, we further discuss the roles of GSDMD and the therapeutic potential of targeting this GSDMD pore-forming activity in coronavirus diseases.
Collapse
Affiliation(s)
- Xiang Liu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shihao Ding
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Pinghuang Liu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
86
|
Zou J, Zheng Y, Huang Y, Tang D, Kang R, Chen R. The Versatile Gasdermin Family: Their Function and Roles in Diseases. Front Immunol 2021; 12:751533. [PMID: 34858408 PMCID: PMC8632255 DOI: 10.3389/fimmu.2021.751533] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
The gasdermin (GSDM) family, a novel group of structure-related proteins, consists of GSDMA, GSDMB, GSDMC, GSDMD, GSDME/DNFA5, and PVJK/GSDMF. GSDMs possess a C-terminal repressor domain, cytotoxic N-terminal domain, and flexible linker domain (except for GSDMF). The GSDM-NT domain can be cleaved and released to form large oligomeric pores in the membrane that facilitate pyroptosis. The emerging roles of GSDMs include the regulation of various physiological and pathological processes, such as cell differentiation, coagulation, inflammation, and tumorigenesis. Here, we introduce the basic structure, activation, and expression patterns of GSDMs, summarize their biological and pathological functions, and explore their regulatory mechanisms in health and disease. This review provides a reference for the development of GSDM-targeted drugs to treat various inflammatory and tissue damage-related conditions.
Collapse
Affiliation(s)
- Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yixiang Zheng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
87
|
Apoptosis, Pyroptosis, and Necroptosis-Oh My! The Many Ways a Cell Can Die. J Mol Biol 2021; 434:167378. [PMID: 34838807 DOI: 10.1016/j.jmb.2021.167378] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/12/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022]
Abstract
Cell death is an essential process in all living organisms and occurs through different mechanisms. The three main types of programmed cell death are apoptosis, pyroptosis, and necroptosis, and each of these pathways employs complex molecular and cellular mechanisms. Although there are mechanisms and outcomes specific to each pathway, they share common components and features. In this review, we discuss recent discoveries in these three best understood modes of cell death, highlighting their singularities, and examining the intriguing notion that common players shape different individual pathways in this highly interconnected and coordinated cell death system. Understanding the similarities and differences of these cell death processes is crucial to enable targeted strategies to manipulate these pathways for therapeutic benefit.
Collapse
|
88
|
Li T, Zheng G, Li B, Tang L. Pyroptosis: A promising therapeutic target for noninfectious diseases. Cell Prolif 2021; 54:e13137. [PMID: 34590363 PMCID: PMC8560609 DOI: 10.1111/cpr.13137] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Pyroptosis, which is characterized by gasdermin family protein-mediated pore formation, cellular lysis and the release of pro-inflammatory cytokines, is a form of programmed cell death associated with intracellular pathogens-induced infection. However, emerging evidence indicates that pyroptosis also contributes to sterile inflammation. In this review, we will first illustrate the biological process of pyroptosis. Then, we will focus on the pathogenic effects of pyroptosis on multiple noninfectious disorders. At last, we will characterize several specific pyroptotic inhibitors targeting the pyroptotic signalling pathway. These data demonstrate that pyroptosis plays a prominent role in sterile diseases, thereby providing a promising approach to the treatment of noninfective inflammatory disorders.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pharmacy, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangjuan Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pathology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ben Li
- Department of Pharmacy, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lipeng Tang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
89
|
Rodríguez-Antonio I, López-Sánchez GN, Uribe M, Chávez-Tapia NC, Nuño-Lámbarri N. Role of the inflammasome, gasdermin D, and pyroptosis in non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2021; 36:2720-2727. [PMID: 34050551 DOI: 10.1111/jgh.15561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Pyroptosis is a type of programmed cell death mediated by a multiprotein complex called the inflammasome through the pro-inflammatory activity of gasdermin D. This study aimed to recognize the final biological product that leads to pore formation in the cell membrane, lysis, pro-inflammatory cytokines release, and the establishment of an immune response. An exhaustive search engine investigation of an elevated immune response can induce a sustained inflammation that directly links this mechanism to non-alcoholic fatty liver disease and its progression to non-alcoholic steatohepatitis. Clinical studies and systematic reviews suggest that gasdermin D is a critical molecule between the immune response and the disease manifestation, which could be considered a therapeutic target for highly prevalent diseases characterized by presenting perpetuated inflammatory processes. Both basic and clinical research show evidence on the expression and regulation of the inflammasome-gasdermin D-pyroptosis trinomial for the progression of non-alcoholic fatty liver disease to non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
| | | | - Misael Uribe
- Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | - Norberto C Chávez-Tapia
- Traslational Research Unit, Médica Sur Clinic Foundation, Mexico City, Mexico.,Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | | |
Collapse
|
90
|
Daskalov A, Glass NL. Gasdermin and Gasdermin-Like Pore-Forming Proteins in Invertebrates, Fungi and Bacteria. J Mol Biol 2021; 434:167273. [PMID: 34599942 DOI: 10.1016/j.jmb.2021.167273] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
The gasdermin family of pore-forming proteins (PFPs) has recently emerged as key molecular players controlling immune-related cell death in mammals. Characterized mammalian gasdermins are activated through proteolytic cleavage by caspases or serine proteases, which remove an inhibitory carboxy-terminal domain, allowing the pore-formation process. Processed gasdermins form transmembrane pores permeabilizing the plasma membrane, which often results in lytic and inflammatory cell death. While the gasdermin-dependent cell death (pyroptosis) has been predominantly characterized in mammals, it now has become clear that gasdermins also control cell death in early vertebrates (teleost fish) and invertebrate animals such as corals (Cnidaria). Moreover, gasdermins and gasdermin-like proteins have been identified and characterized in taxa outside of animals, notably Fungi and Bacteria. Fungal and bacterial gasdermins share many features with mammalian gasdermins including their mode of activation through proteolysis. It has been shown that in some cases the proteolytic activation is executed by evolutionarily related proteases acting downstream of proteins resembling immune receptors controlling pyroptosis in mammals. Overall, these findings establish gasdermins and gasdermin-regulated cell death as an extremely ancient mechanism of cellular suicide and build towards an understanding of the evolution of regulated cell death in the context of immunology. Here, we review the broader gasdermin family, focusing on recent discoveries in invertebrates, fungi and bacteria.
Collapse
Affiliation(s)
- Asen Daskalov
- Institut de Biochimie et Génétique Cellulaires, University of Bordeaux, France.
| | - N Louise Glass
- The Plant and Microbial Biology Department, The University of California, Berkeley, CA 94720-3102, United States
| |
Collapse
|
91
|
Américo-Da-Silva L, Aguilera J, Quinteros-Waltemath O, Sánchez-Aguilera P, Russell J, Cadagan C, Meneses-Valdés R, Sánchez G, Estrada M, Jorquera G, Barrientos G, Llanos P. Activation of the NLRP3 Inflammasome Increases the IL-1β Level and Decreases GLUT4 Translocation in Skeletal Muscle during Insulin Resistance. Int J Mol Sci 2021; 22:10212. [PMID: 34638553 PMCID: PMC8508423 DOI: 10.3390/ijms221910212] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Low-grade chronic inflammation plays a pivotal role in the pathogenesis of insulin resistance (IR), and skeletal muscle has a central role in this condition. NLRP3 inflammasome activation pathways promote low-grade chronic inflammation in several tissues. However, a direct link between IR and NLRP3 inflammasome activation in skeletal muscle has not been reported. Here, we evaluated the NLRP3 inflammasome components and their role in GLUT4 translocation impairment in skeletal muscle during IR. Male C57BL/6J mice were fed with a normal control diet (NCD) or high-fat diet (HFD) for 8 weeks. The protein levels of NLRP3, ASC, caspase-1, gasdermin-D (GSDMD), and interleukin (IL)-1β were measured in both homogenized and isolated fibers from the flexor digitorum brevis (FDB) or soleus muscle. GLUT4 translocation was determined through GLUT4myc-eGFP electroporation of the FBD muscle. Our results, obtained using immunofluorescence, showed that adult skeletal muscle expresses the inflammasome components. In the FDB and soleus muscles, homogenates from HFD-fed mice, we found increased protein levels of NLRP3 and ASC, higher activation of caspase-1, and elevated IL-1β in its mature form, compared to NCD-fed mice. Moreover, GSDMD, a protein that mediates IL-1β secretion, was found to be increased in HFD-fed-mice muscles. Interestingly, MCC950, a specific pharmacological NLRP3 inflammasome inhibitor, promoted GLUT4 translocation in fibers isolated from the FDB muscle of NCD- and HFD-fed mice. In conclusion, we found increased NLRP3 inflammasome components in adult skeletal muscle of obese insulin-resistant animals, which might contribute to the low-grade chronic metabolic inflammation of skeletal muscle and IR development.
Collapse
Affiliation(s)
- Luan Américo-Da-Silva
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Javiera Aguilera
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Oscar Quinteros-Waltemath
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Pablo Sánchez-Aguilera
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| | - Javier Russell
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad Autónoma de Chile, Santiago 8900000, Chile;
| | - Cynthia Cadagan
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Roberto Meneses-Valdés
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| | - Gina Sánchez
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Genaro Barrientos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Paola Llanos
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| |
Collapse
|
92
|
Nandi D, Farid NSS, Karuppiah HAR, Kulkarni A. Imaging Approaches to Monitor Inflammasome Activation. J Mol Biol 2021; 434:167251. [PMID: 34537231 DOI: 10.1016/j.jmb.2021.167251] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Inflammasomes are a critical component of innate immune response which plays an important role in the pathogenesis of various chronic and acute inflammatory disease conditions. An inflammasome complex consists of a multimeric protein assembly triggered by any form of pathogenic or sterile insult, resulting in caspase-1 activation. This active enzyme is further known to activate downstream pro-inflammatory cytokines along with a pore-forming protein, eventually leading to a lytic cell death called pyroptosis. Understanding the spatiotemporal kinetics of essential inflammasome components provides a better interpretation of the complex signaling underlying inflammation during several disease pathologies. This can be attained via in-vitro and in-vivo imaging platforms, which not only provide a basic understanding of molecular signaling but are also crucial to develop and screen targeted therapeutics. To date, numerous studies have reported platforms to image different signaling components participating in inflammasome activation. Here, we review several elements of inflammasome signaling, a common molecular mechanism combining these elements and their respective imaging tools. We anticipate that future needs will include developing new inflammasome imaging systems that can be utilized as clinical tools for diagnostics and monitoring treatment responses.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA. https://twitter.com/dipikanandi24
| | - Noorul Shaheen Sheikh Farid
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/Shaheen30n
| | - Hayat Anu Ranjani Karuppiah
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/AnuHayat
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA; Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA; Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
93
|
Kulma M, Anderluh G. Beyond pore formation: reorganization of the plasma membrane induced by pore-forming proteins. Cell Mol Life Sci 2021; 78:6229-6249. [PMID: 34387717 PMCID: PMC11073440 DOI: 10.1007/s00018-021-03914-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 12/23/2022]
Abstract
Pore-forming proteins (PFPs) are a heterogeneous group of proteins that are expressed and secreted by a wide range of organisms. PFPs are produced as soluble monomers that bind to a receptor molecule in the host cell membrane. They then assemble into oligomers that are incorporated into the lipid membrane to form transmembrane pores. Such pore formation alters the permeability of the plasma membrane and is one of the most common mechanisms used by PFPs to destroy target cells. Interestingly, PFPs can also indirectly manipulate diverse cellular functions. In recent years, increasing evidence indicates that the interaction of PFPs with lipid membranes is not only limited to pore-induced membrane permeabilization but is also strongly associated with extensive plasma membrane reorganization. This includes lateral rearrangement and deformation of the lipid membrane, which can lead to the disruption of target cell function and finally death. Conversely, these modifications also constitute an essential component of the membrane repair system that protects cells from the lethal consequences of pore formation. Here, we provide an overview of the current knowledge on the changes in lipid membrane organization caused by PFPs from different organisms.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia
| |
Collapse
|
94
|
Isoorientin Attenuated the Pyroptotic Hepatocyte Damage Induced by Benzo[a]pyrene via ROS/NF-κB/NLRP3/Caspase-1 Signaling Pathway. Antioxidants (Basel) 2021; 10:antiox10081275. [PMID: 34439523 PMCID: PMC8389279 DOI: 10.3390/antiox10081275] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 01/29/2023] Open
Abstract
Isoorientin (Iso), a natural bioactive flavonoid, possesses significant anti-tumor and anti-oxidant activities. Benzo[a]pyrene (BaP) is a food processing injurant with carcinogenicity, teratogenicity, and genotoxicity. Our preliminary study demonstrates that Iso attenuated the pyroptotic hepatocyte damage induced by BaP; however, the molecular mechanism remains unknown. The present study showed that Iso reduced the increase caused by BaP in the overflow of LDH, NO, and the electrical conductivity and the protein expressions of GSDMD-N, IL-18, and IL-1β, further showing that Iso could reduced the pyroptotic damage in HL-7702 cells induced by BaP. Caspase-1 inhibitor (Z-VAD-FMK) inhibited the characteristic pyroptosis protein expressions of Caspase-1, GSDMD-N, IL-18, and IL-1β, showing that the classic pyroptosis pathway depending on Caspase-1 was caused by BaP in HL-7702 cells. Consistent with the effects of the NLRP3 inhibitor (MCC950), NF-κB inhibitor (PDTC), ROS, and mtROS inhibitor (NAC and Mito-TEMPO), Iso weakened the stimulatory effects of BaP on the levels of ROS, the nuclear localization of NF-κB, and the activation of NLRP3 inflammasome and the characteristic indices of pyroptosis, demonstrating that Iso could alleviate the BaP-induced pyroptotic hepatocytes injury through inhibiting the ROS/NF-κB/NLRP3/Caspase-1 signaling pathway, which provides a new perspective and strategy to prevent liver injury induced by BaP.
Collapse
|
95
|
Chen MY, Ye XJ, He XH, Ouyang DY. The Signaling Pathways Regulating NLRP3 Inflammasome Activation. Inflammation 2021; 44:1229-1245. [PMID: 34009550 DOI: 10.1007/s10753-021-01439-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/30/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
The NLRP3 inflammasome is a multi-molecular complex that acts as a molecular platform to mediate caspase-1 activation, leading to IL-1β/IL-18 maturation and release in cells stimulated by various pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). This inflammasome plays an important role in the innate immunity as its activation can further promote the occurrence of inflammation, enhance the ability of host to remove pathogens, and thus facilitate the repair of injured tissues. But if the inflammasome activation is dysregulated, it will cause the development of various inflammatory diseases and metabolic disorders. Therefore, under normal conditions, the activation of inflammasome is tightly regulated by various positive and negative signaling pathways to respond to the stimuli without damaging the host itself while maintaining homeostasis. In this review, we summarize recent advances in the major signaling pathways (including TLRs, MAPK, mTOR, autophagy, PKA, AMPK, and IFNR) that regulate NLRP3 inflammasome activation, providing a brief view of the molecular network that regulates this inflammasome as a theoretical basis for therapeutic intervention of NLRP3 dysregulation-related diseases.
Collapse
Affiliation(s)
- Ming-Ye Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xun-Jia Ye
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Dong-Yun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
96
|
Wang XP, Zheng WC, Bai Y, Li Y, Xin Y, Wang JZ, Chang YL, Zhang LM. Carbon Monoxide-Releasing Molecule-3 Alleviates Kupffer Cell Pyroptosis Induced by Hemorrhagic Shock and Resuscitation via sGC-cGMP Signal Pathway. Inflammation 2021; 44:1330-1344. [PMID: 33575924 DOI: 10.1007/s10753-021-01419-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/30/2020] [Accepted: 01/11/2021] [Indexed: 12/23/2022]
Abstract
Following hepatic ischemia-reperfusion injury, Kupffer cells could be activated by inflammatory factors released from damaged hepatocytes. Carbon monoxide (CO)-releasing molecule (CORM)-3, a water-soluble transition metal carbonyl, exhibits excellent anti-inflammatory and anti-pyroptosis properties. We investigated whether CORM-3 attenuated hemorrhagic shock and resuscitation (HSR)-induced pyroptosis of Kupffer cells through the soluble guanylate-cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signal pathway. NS2028 (10 mg/kg), a blocker of sGC, was administrated at the onset of hemorrhage, but CORM-3 (4 mg/kg) was infused after resuscitation via femoral vein. Serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels, tumor necrosis Factor-α (TNF-α), and interleukin-1β (IL-1β) were measured at 3, 6, 12, and 24 h after HSR, respectively. Six hours post-HSR, liver injury, pyroptosis of Kupffer cells, and expressions in total caspase-1, cleaved caspase-1, gasdermin D (GSDMD) N-terminal fragment, IL-1β, and IL-18 were measured by hematoxylin-eosin (H&E), immunofluorescence and western blot assays, respectively (Fig. 1). The rats exposed to HSR exhibited significant upregulated levels of serum ALT, AST, TNF-α, and IL-1β, elevated liver injury score, increased pyroptosis of Kupffer cells, and accumulated expressions of pyroptosis-associated protein including cleaved caspase-1, GSDMD N-terminal fragment, IL-1β, and IL-18 than sham-treated rats. However, CORM-3 administration markedly reduced liver injury and pyroptosis of Kupffer cells, whereas these protective effects could be partially blocked by NS2028. CORM-3 can mitigate pyroptosis of Kupffer cells in a blood loss and re-infusion model of rats via sGC-cGMP signal pathway.
Collapse
Affiliation(s)
- Xu-Peng Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Yang Bai
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Jing-Zhou Wang
- Department of Neurosurgery, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Yu-Lin Chang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China.
| |
Collapse
|
97
|
Pandey A, Shen C, Feng S, Man SM. Cell biology of inflammasome activation. Trends Cell Biol 2021; 31:924-939. [PMID: 34284921 DOI: 10.1016/j.tcb.2021.06.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022]
Abstract
Organelles are critical structures in mediating the assembly and activation of inflammasomes in mammalian cells, resulting in inflammation and cell death. Assembly of inflammasomes can occur at the mitochondria, endoplasmic reticulum, nucleus, trans-Golgi network, or pathogen surface, facilitated by the overarching architecture of the cytoskeleton. NLRP3 and Pyrin inflammasome sensors may form smaller speckles and converge on a single larger speck at the microtubule-organizing center (MTOC). This signaling hub activates multiple mammalian inflammatory and apoptotic caspases, cytokine substrates, the pore-forming protein gasdermin D, and the plasma membrane rupture protein ninjurin-1 (NINJ1), allowing pyroptosis, cellular disintegration, and inflammation to ensue. In this review, we highlight the role of mammalian cell types and organellar architectures in executing inflammasome responses.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Cheng Shen
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Shouya Feng
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Si Ming Man
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
98
|
Fakhoury HMA, Kvietys PR, Shakir I, Shams H, Grant WB, Alkattan K. Lung-Centric Inflammation of COVID-19: Potential Modulation by Vitamin D. Nutrients 2021; 13:2216. [PMID: 34203190 PMCID: PMC8308422 DOI: 10.3390/nu13072216] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 infects the respiratory tract and leads to the disease entity, COVID-19. Accordingly, the lungs bear the greatest pathologic burden with the major cause of death being respiratory failure. However, organs remote from the initial site of infection (e.g., kidney, heart) are not spared, particularly in severe and fatal cases. Emerging evidence indicates that an excessive inflammatory response coupled with a diminished antiviral defense is pivotal in the initiation and development of COVID-19. A common finding in autopsy specimens is the presence of thrombi in the lungs as well as remote organs, indicative of immunothrombosis. Herein, the role of SARS-CoV-2 in lung inflammation and associated sequelae are reviewed with an emphasis on immunothrombosis. In as much as vitamin D is touted as a supplement to conventional therapies of COVID-19, the impact of this vitamin at various junctures of COVID-19 pathogenesis is also addressed.
Collapse
Affiliation(s)
- Hana. M. A. Fakhoury
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Peter R. Kvietys
- Department of Physiology, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (P.R.K.); (I.S.); (H.S.)
| | - Ismail Shakir
- Department of Physiology, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (P.R.K.); (I.S.); (H.S.)
| | - Hashim Shams
- Department of Physiology, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (P.R.K.); (I.S.); (H.S.)
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, P.O. Box 641603, San Francisco, CA 94164-1603, USA;
| | - Khaled Alkattan
- Department of Surgery, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
99
|
Kuc-Ciepluch D, Ciepluch K, Arabski M. Gasdermin family proteins as a permeabilization factor
of cell membrane in pyroptosis process. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.8985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The type of cell death, i.e. apoptosis, autophagy, necrosis or pyroptosis, depends on the inducing
factor and the phase of the cell cycle. The main role in immunological response to microorganisms
is played by a process called pyroptosis. Pyroptosis induces various types of inflammatory
factors in response to molecular patterns associated with pathogens, e.g., bacterial lipopolysaccharide
in the canonical or non-canonical pathway depending on the type of caspases involved.
In pyroptosis, the gasdermin D protein belonging to the gasdermin protein family (A, B, C, D, E
and DFNB59) plays an important role, which is characterized by specific tissue gene expression
mainly in epithelial cells, skin and the digestive system and is responsible for regulating the proliferation
and differentiation of cells and is responsible for inhibiting or developing cancers in
various organs. The GSDM family is responsible for the formation of pores in the cell membrane,
enabling the secretion of proinflammatory cytokines (IL-1β and IL-18) involved in initiating inflammatory
response pathways by recruiting and activating immune cells at the site of infection.
The gasdermin D protein plays an essential role in the non-canonical pyroptosis process, whose
N-terminal forming pores in the cell membrane leads to edema, osmotic lysis and, consequently,
to the death of the infected cell.
Collapse
Affiliation(s)
- Dorota Kuc-Ciepluch
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| | - Karol Ciepluch
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| | - Michał Arabski
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| |
Collapse
|
100
|
Bian K, Gerber C, Heinrich AJ, Müller DJ, Scheuring S, Jiang Y. Scanning probe microscopy. ACTA ACUST UNITED AC 2021. [DOI: 10.1038/s43586-021-00033-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|