51
|
Sheng Y, Hu W, Chen S, Zhu X. Efferocytosis by macrophages in physiological and pathological conditions: regulatory pathways and molecular mechanisms. Front Immunol 2024; 15:1275203. [PMID: 38779685 PMCID: PMC11109379 DOI: 10.3389/fimmu.2024.1275203] [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: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Efferocytosis is defined as the highly effective phagocytic removal of apoptotic cells (ACs) by professional or non-professional phagocytes. Tissue-resident professional phagocytes ("efferocytes"), such as macrophages, have high phagocytic capacity and are crucial to resolve inflammation and aid in homeostasis. Recently, numerous exciting discoveries have revealed divergent (and even diametrically opposite) findings regarding metabolic immune reprogramming associated with efferocytosis by macrophages. In this review, we highlight the key metabolites involved in the three phases of efferocytosis and immune reprogramming of macrophages under physiological and pathological conditions. The next decade is expected to yield further breakthroughs in the regulatory pathways and molecular mechanisms connecting immunological outcomes to metabolic cues as well as avenues for "personalized" therapeutic intervention.
Collapse
Affiliation(s)
- Yan−Ran Sheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen−Ting Hu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Siman Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao−Yong Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
52
|
Yang F, Chen M, Liu Y, Hu Y, Chen Y, Yu Y, Deng L. ANGPTL2 knockdown induces autophagy to relieve alveolar macrophage pyroptosis by reducing LILRB2-mediated inhibition of TREM2. J Cell Mol Med 2024; 28:e18280. [PMID: 38758159 PMCID: PMC11100552 DOI: 10.1111/jcmm.18280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/22/2024] [Accepted: 03/18/2024] [Indexed: 05/18/2024] Open
Abstract
Acute lung injury (ALI) is featured with a robust inflammatory response. Angiopoietin-like protein 2 (ANGPTL2), a pro-inflammatory protein, is complicated with various disorders. However, the role of ANGPTL2 in ALI remains to be further explored. The mice and MH-S cells were administrated with lipopolysaccharide (LPS) to evoke the lung injury in vivo and in vitro. The role and mechanism of ANGPTL was investigated by haematoxylin-eosin, measurement of wet/dry ratio, cell count, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling, reverse transcription quantitative polymerase chain reaction, immunofluorescence, enzyme-linked immunosorbent assay, detection of autophagic flux and western blot assays. The level of ANGPTL2 was upregulated in lung injury. Knockout of ANGPTL2 alleviated LPS-induced pathological symptoms, reduced pulmonary wet/dry weight ratio, the numbers of total cells and neutrophils in BALF, apoptosis rate and the release of pro-inflammatory mediators, and modulated polarization of alveolar macrophages in mice. Knockdown of ANGPTL2 downregulated the level of pyroptosis indicators, and elevated the level of autophagy in LPS-induced MH-S cells. Besides, downregulation of ANGPTL2 reversed the LPS-induced the expression of leukocyte immunoglobulin (Ig)-like receptor B2 (LILRB2) and triggering receptor expressed on myeloid cells 2 (TREM2), which was reversed by the overexpression of LILRB2. Importantly, knockdown of TREM2 reversed the levels of autophagy- and pyroptosis-involved proteins, and the contents of pro-inflammatory factors in LPS-induced MH-S cells transfected with si ANGPTL2, which was further inverted with the treatment of rapamycin. Therefore, ANGPTL2 silencing enhanced autophagy to alleviate alveolar macrophage pyroptosis via reducing LILRB2-mediated inhibition of TREM2.
Collapse
Affiliation(s)
- Fan Yang
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Muhu Chen
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Ying Liu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Yingchun Hu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Yangxi Chen
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Youwei Yu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Lu Deng
- Department of Thyroid SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
53
|
Nadjar J, Monnier S, Bastien E, Huber AL, Oddou C, Bardoulet L, Leloup HB, Ichim G, Vanbelle C, Py BF, Destaing O, Petrilli V. Optogenetically controlled inflammasome activation demonstrates two phases of cell swelling during pyroptosis. Sci Signal 2024; 17:eabn8003. [PMID: 38652763 DOI: 10.1126/scisignal.abn8003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Inflammasomes are multiprotein platforms that control caspase-1 activation, which process the inactive precursor forms of the inflammatory cytokines IL-1β and IL-18, leading to an inflammatory type of programmed cell death called pyroptosis. Studying inflammasome-driven processes, such as pyroptosis-induced cell swelling, under controlled conditions remains challenging because the signals that activate pyroptosis also stimulate other signaling pathways. We designed an optogenetic approach using a photo-oligomerizable inflammasome core adapter protein, apoptosis-associated speck-like containing a caspase recruitment domain (ASC), to temporally and quantitatively manipulate inflammasome activation. We demonstrated that inducing the light-sensitive oligomerization of ASC was sufficient to recapitulate the classical features of inflammasomes within minutes. This system showed that there were two phases of cell swelling during pyroptosis. This approach offers avenues for biophysical investigations into the intricate nature of cellular volume control and plasma membrane rupture during cell death.
Collapse
Affiliation(s)
- Julien Nadjar
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Sylvain Monnier
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, F-69622, Villeurbanne, France
| | - Estelle Bastien
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, F-69622, Villeurbanne, France
| | - Anne-Laure Huber
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Christiane Oddou
- DYSAD, Institut pour l'avancée des biosciences (IAB), Centre de Recherche UGA / Inserm U 1209/CNRS UMR 5309, 38700 La Tronche, France
| | - Léa Bardoulet
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Hubert B Leloup
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Gabriel Ichim
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Christophe Vanbelle
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| | - Bénédicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Olivier Destaing
- DYSAD, Institut pour l'avancée des biosciences (IAB), Centre de Recherche UGA / Inserm U 1209/CNRS UMR 5309, 38700 La Tronche, France
| | - Virginie Petrilli
- CRCL, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, F-69000 Lyon, France
| |
Collapse
|
54
|
Yang H, Park G, Lee S, Lee S, Kim Y, Zamora NA, Yi D, Kim S, Choi CW, Choi S, Park YH. Anti-inflammatory effect of Trichospira verticillata via suppression of the NLRP3 inflammasome in neutrophilic asthma. J Cell Mol Med 2024; 28:e18356. [PMID: 38668995 PMCID: PMC11048967 DOI: 10.1111/jcmm.18356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Trichospira verticillata is an annual herb that belongs to the family Asteraceae. Trichospira verticillata extract (TVE) elicits anti-plasmodial activity; however, there has been no detailed report about its anti-inflammatory effects and molecular mechanisms. In addition, herbal plants exhibit anti-inflammatory effects by suppressing the NLRP3 inflammasome. Therefore, the primary goal of this study was to examine the effects of TVE on NLRP3 inflammasome activation by measuring interleukin-1β (IL-1β) secretion. We treated lipopolysaccharides (LPS)-primed J774A.1 and THP-1 cells with TVE, which attenuated NLRP3 inflammasome activation. Notably, TVE did not affect nuclear factor-kappa B (NF-κB) signalling or intracellular reactive oxygen species (ROS) production and potassium efflux, suggesting that it inactivates the NLRP3 inflammasome via other mechanisms. Moreover, TVE suppressed the formation of apoptosis-associated speck-like protein (ASC) speck and oligomerization. Immunoprecipitation data revealed that TVE reduced the binding of NLRP3 to NIMA-related kinase 7 (NEK7), resulting in reduced ASC oligomerization and speck formation. Moreover, TVE alleviated neutrophilic asthma (NA) symptoms in mice. This study demonstrates that TVE modulates the binding of NLPR3 to NEK7, thereby reporting novel insights into the mechanism by which TVE inhibits NLRP3 inflammasome. These findings suggest TVE as a potential therapeutic of NLRP3 inflammasome-mediated diseases, particularly NA.
Collapse
Affiliation(s)
- Hyeyun Yang
- Department of MicrobiologyAjou University School of MedicineSuwonRepublic of Korea
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
| | - Gunwoo Park
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
- Department of Allergy and Clinical ImmunologyAjou University School of MedicineSuwonRepublic of Korea
| | - Sojung Lee
- Department of MicrobiologyAjou University School of MedicineSuwonRepublic of Korea
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
| | - Sumin Lee
- Department of MicrobiologyAjou University School of MedicineSuwonRepublic of Korea
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
| | - YeJi Kim
- Department of MicrobiologyAjou University School of MedicineSuwonRepublic of Korea
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
| | - Nelson A. Zamora
- Instituto Nacional de Biodiversidad (INBio)Santo DomingoCosta Rica
| | - Dong‐Keun Yi
- International Biological Material Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Soo‐Yong Kim
- International Biological Material Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Chun Whan Choi
- Natural Biomaterial TeamGyeonggi Bio‐CenterSuwonRepublic of Korea
| | - Sangho Choi
- International Biological Material Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Yong Hwan Park
- Department of MicrobiologyAjou University School of MedicineSuwonRepublic of Korea
- Department of Biomedical SciencesGraduate School of Ajou UniversitySuwonRepublic of Korea
| |
Collapse
|
55
|
Ahn H, Lee GS. Amino Sugar-Enriched Fraction of Korean Red Ginseng Extract Induces the Priming Step of NLRP3 Inflammasome. Molecules 2024; 29:1455. [PMID: 38611734 PMCID: PMC11013037 DOI: 10.3390/molecules29071455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Intracellular protein complexes, known as inflammasomes, activate caspase-1 and induce the secretion of pro-inflammatory cytokines, namely interleukin (IL)-1β and -18. Korean Red Ginseng extract (RGE) is a known immunomodulator and a potential candidate for the regulation of inflammasomes. The saponins, such as ginsenosides, of RGE inhibit inflammasome signaling, while non-saponin substances containing amino sugars promote the priming step, up-regulating inflammasome components (pro-IL-1β, NLRP3, caspase-1, and Asc). In this study, the amino sugar-enriched fraction (ASEF), which increases only non-saponin components, including amino sugars, without changing the concentration of saponin substances, was used to investigate whether saponin or non-saponin components of RGE would have a greater impact on the priming step. When murine macrophages were treated with ASEF, the gene expression of inflammatory cytokines (IL-1α, TNFα, IL-6, and IL-10) increased. Additionally, ASEF induced the priming step but did not affect the inflammasome activation step, such as the secretion of IL-1β, cleavage of caspase-1, and formation of Asc pyroptosome. Furthermore, the upregulation of gene expression of inflammasome components by ASEF was blocked by inhibitors of Toll-like receptor 4 signaling. Maltol, the main constituent of ASEF, promoted the priming step but inhibited the activation step of the inflammasome, while arginine, sugars, arginine-fructose-glucose, and fructose-arginine, the other main constituents of ASEF, had no effect on either step. Thus, certain amino sugars in RGE, excluding maltol, are believed to be the components that induce the priming step. The priming step that prepares the NLRP3 inflammasome for activation appears to be induced by amino sugars in RGE, thereby contributing to the immune-boosting effects of RGE.
Collapse
Affiliation(s)
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
56
|
Wu F, Du T, Jiang X, Liu S, Cheng Y, Zhang Z, Miao W, Wang T. Lactococcus garvieae exerts a critical role in inducing inflammation in dairy mastitis by triggering NLRP3 inflammasome-mediated pyroptosis in MAC-T cells. World J Microbiol Biotechnol 2024; 40:132. [PMID: 38470533 DOI: 10.1007/s11274-024-03947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
Lactococcus garvieae (L. garvieae) is a pathogenic bacterium that is Gram-positive and catalase-negative (GPCN), and it is capable of growing in a wide range of environmental conditions. This bacterium is associated with significant mortality and losses in fisheries, and there are concerns regarding its potential as a zoonotic pathogen, given its presence in cattle and dairy products. While we have identified and characterized virulent strains of L. garvieae through phenotyping and molecular typing studies, their impact on mammary tissue remains unknown. This study aims to investigate the pathogenicity of strong and weak virulent strains of L. garvieae using in vivo mouse models. We aim to establish MAC-T cell model to examine potential injury caused by the strong virulent strain LG41 through the TLR2/NLRP3/NF-kB pathway. Furthermore, we assess the involvement of NLRP3 inflammasome-mediated pyroptosis in dairy mastitis by silencing NLRP3. The outcomes of this study will yield crucial theoretical insights into the potential mechanisms involved in mastitis in cows caused by the L. garvieae-induced inflammatory response in MAC-T cells.
Collapse
Affiliation(s)
- Fan Wu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Tao Du
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Xiaodan Jiang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Shuhong Liu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yiru Cheng
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Zhe Zhang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Wei Miao
- Agronomy college, Shenyang Agricultural University, Shenyang, 110866, China
| | - Tiancheng Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| |
Collapse
|
57
|
Lee KG, Hong BK, Lee S, Lee N, Kim SW, Kim D, Kim WU. Nuclear receptor coactivator 6 is a critical regulator of NLRP3 inflammasome activation and gouty arthritis. Cell Mol Immunol 2024; 21:227-244. [PMID: 38195836 PMCID: PMC10902316 DOI: 10.1038/s41423-023-01121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
Transcriptional coactivators regulate the rate of gene expression in the nucleus. Nuclear receptor coactivator 6 (NCOA6), a coactivator, has been implicated in embryonic development, metabolism, and cancer pathogenesis, but its role in innate immunity and inflammatory diseases remains unclear. Here, we demonstrated that NCOA6 was expressed in monocytes and macrophages and that its level was increased under proinflammatory conditions. Unexpectedly, nuclear NCOA6 was found to translocate to the cytoplasm in activated monocytes and then become incorporated into the inflammasome with NLRP3 and ASC, forming cytoplasmic specks. Mechanistically, NCOA6 associated with the ATP hydrolysis motifs in the NACHT domain of NLRP3, promoting the oligomerization of NLRP3 and ASC and thereby instigating the production of IL-1β and active caspase-1. Of note, Ncoa6 deficiency markedly inhibited NLRP3 hyperactivation caused by the Nlrp3R258W gain-of-function mutation in macrophages. Genetic ablation of Ncoa6 substantially attenuated the severity of two NLRP3-dependent diseases, folic-induced acute tubular necrosis and crystal-induced arthritis, in mice. Consistent with these findings, NCOA6 was highly expressed in macrophages derived from gout patients, and NCOA6-positive macrophages were significantly enriched in gout macrophages according to the transcriptome profiling results. Conclusively, NCOA6 is a critical regulator of NLRP3 inflammasome activation and is therefore a promising target for NLRP3-dependent diseases, including gout.
Collapse
Affiliation(s)
- Kang-Gu Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Naeun Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seung-Whan Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan, Seoul, 05505, Republic of Korea
| | - Donghyun Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
58
|
Stergiou IE, Tsironis C, Papadakos SP, Tsitsilonis OE, Dimopoulos MA, Theocharis S. Unraveling the Role of the NLRP3 Inflammasome in Lymphoma: Implications in Pathogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:2369. [PMID: 38397043 PMCID: PMC10889189 DOI: 10.3390/ijms25042369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammasomes are multimeric protein complexes, sensors of intracellular danger signals, and crucial components of the innate immune system, with the NLRP3 inflammasome being the best characterized among them. The increasing scientific interest in the mechanisms interconnecting inflammation and tumorigenesis has led to the study of the NLRP3 inflammasome in the setting of various neoplasms. Despite a plethora of data regarding solid tumors, NLRP3 inflammasome's implication in the pathogenesis of hematological malignancies only recently gained attention. In this review, we investigate its role in normal lymphopoiesis and lymphomagenesis. Considering that lymphomas comprise a heterogeneous group of hematologic neoplasms, both tumor-promoting and tumor-suppressing properties were attributed to the NLRP3 inflammasome, affecting neoplastic cells and immune cells in the tumor microenvironment. NLRP3 inflammasome-related proteins were associated with disease characteristics, response to treatment, and prognosis. Few studies assess the efficacy of NLRP3 inflammasome therapeutic targeting with encouraging results, though most are still at the preclinical level. Further understanding of the mechanisms regulating NLRP3 inflammasome activation during lymphoma development and progression can contribute to the investigation of novel treatment approaches to cover unmet needs in lymphoma therapeutics.
Collapse
Affiliation(s)
- Ioanna E. Stergiou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (C.T.)
| | - Christos Tsironis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (C.T.)
| | - Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece;
| | - Ourania E. Tsitsilonis
- Flow Cytometry Unit, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15784 Athens, Greece;
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, 11528 Athens, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece;
| |
Collapse
|
59
|
Jiang X, Zhu W, Sun Y, Wang S, Sun M, Tang R, Tang Z, Ma T. Tandem mass tag-based quantitative proteomics analyses of the spermatogenesis-ameliorating effect of Youjing granule on rats. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9679. [PMID: 38211349 DOI: 10.1002/rcm.9679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/15/2023] [Accepted: 11/11/2023] [Indexed: 01/13/2024]
Abstract
RATIONALE Male infertility is a common reproductive system disease manifested as aberrant spermatogenesis and identified as "kidney deficiency and dampness" in Chinese traditional medicine. Youjing granule (YG) is a Chinese material medica based on tonifying kidneys and removing dampness. It has proven to be able to regulate semen quality in clinical application, but the underlying mechanism has not been clarified. METHODS Using serum containing YG to treat primarily cultured spermatogonial stem cells (SSCs), the apoptotic rate and mitosis phase ratio of SSCs were measured. The liquid chromatography-tandem mass spectrometry with tandem mass tags method was applied for analyzing the serum of rats treated with YG/distilled water, and proteomic analyses were performed to clarify the mechanisms of YG. RESULTS Totally, 111 proteins in YG-treated serum samples were differentially expressed compared with control groups, and 43 of them were identified as potential target proteins, which were further annotated based on their enrichment in Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways. Proteomic analyses showed that the mechanisms of YG may involve regulation of glycolysis, gluconeogenesis and nucleotide-binding and oligomerization domain-like receptor signaling pathway. In addition, RhoA and Lamp2 were found to be possible responders of YG through reviewing the literature. CONCLUSIONS The results demonstrate that our serum proteomics platform is clinically useful in understanding the mechanisms of YG.
Collapse
Affiliation(s)
- Xuping Jiang
- Department of Traditional Chinese Medicine, Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
- Department of Urology, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Wenjiao Zhu
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Yaoxiang Sun
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
- Department of Clinical Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Sijia Wang
- Department of Traditional Chinese Medicine, Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Miaomiao Sun
- Department of Traditional Chinese Medicine, Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
| | - Ruijie Tang
- School of Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhian Tang
- Department of Traditional Chinese Medicine, Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Tieliang Ma
- Department of Traditional Chinese Medicine, Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| |
Collapse
|
60
|
Huang W, Wang B, Ou Q, Zhang X, He Y, Mao X, Wei X, Kou X. ASC-expressing pyroptotic extracellular vesicles alleviate sepsis by protecting B cells. Mol Ther 2024; 32:395-410. [PMID: 38093517 PMCID: PMC10861962 DOI: 10.1016/j.ymthe.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Pyroptosis is an inflammatory programmed cell death process characterized by membrane rupture. Interestingly, pyroptotic cells can generate plenty of nanosized vesicles. Non-inflammatory apoptotic cell death-derived apoptotic vesicles (apoVs) were systemically characterized and displayed multiple physiological functions and therapeutic potentials. However, the characteristics of pyroptotic cell-generated extracellular vesicles (EVs) are largely unknown. Here, we identified a group of pyroptotic EVs (pyroEVs) from in vitro cultured pyroptotic mesenchymal stem cells (MSCs), as well as from septic mouse blood. Compared with apoVs, pyroEVs express similar levels of annexin V, calreticulin, and common EV markers, but express a decreased level of apoptotic marker cleave caspase-3. PyroEVs, but not apoVs and exosomes, specifically express pyroptotic maker apoptosis-associated speck-like protein containing CARD (ASC). More importantly, MSC-derived pyroEVs protect B cells in the spleen and bone marrow to relieve inflammatory responses and enhance the survival rate of the septic mice. Mechanistically, pyroEV membrane-expressed ASC binds to B cells to repress cell death by repressing Toll-like receptor 4. This study uncovered the characteristics of pyroEVs and their therapeutic role in sepsis and B cell-mediated immune response.
Collapse
Affiliation(s)
- Weiying Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Ben Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiao Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, China.
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China.
| |
Collapse
|
61
|
He W, Mu X, Wu X, Liu Y, Deng J, Liu Y, Han F, Nie X. The cGAS-STING pathway: a therapeutic target in diabetes and its complications. BURNS & TRAUMA 2024; 12:tkad050. [PMID: 38312740 PMCID: PMC10838060 DOI: 10.1093/burnst/tkad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 02/06/2024]
Abstract
Diabetic wound healing (DWH) represents a major complication of diabetes where inflammation is a key impediment to proper healing. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a central mediator of inflammatory responses to cell stress and damage. However, the contribution of cGAS-STING activation to impaired healing in DWH remains understudied. In this review, we examine the evidence that cGAS-STING-driven inflammation is a critical factor underlying defective DWH. We summarize studies revealing upregulation of the cGAS-STING pathway in diabetic wounds and discuss how this exacerbates inflammation and senescence and disrupts cellular metabolism to block healing. Partial pharmaceutical inhibition of cGAS-STING has shown promise in damping inflammation and improving DWH in preclinical models. We highlight key knowledge gaps regarding cGAS-STING in DWH, including its relationships with endoplasmic reticulum stress and metal-ion signaling. Elucidating these mechanisms may unveil new therapeutic targets within the cGAS-STING pathway to improve healing outcomes in DWH. This review synthesizes current understanding of how cGAS-STING activation contributes to DWH pathology and proposes future research directions to exploit modulation of this pathway for therapeutic benefit.
Collapse
Affiliation(s)
- Wenjie He
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Xingrui Mu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Xingqian Wu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Ye Liu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Junyu Deng
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Yiqiu Liu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| |
Collapse
|
62
|
Guo X, Qiao G, Wang J, Yang C, Zhao M, Zhang Q, Wan Y. TIFA contributes to periodontitis in diabetic mice via activating the NF‑κB signaling pathway. Mol Med Rep 2024; 29:23. [PMID: 38099344 PMCID: PMC10784739 DOI: 10.3892/mmr.2023.13146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic periodontitis (DP) refers to destruction of periodontal tissue and absorption of bone tissue in diabetic patients. Tumor necrosis factor receptor‑associated factor (TRAF)‑interacting protein with forkhead‑associated domain (TIFA) as a crucial regulator of inflammation activates the NF‑κB signaling pathway to regulate cell biological behavior. However, the function and mechanism of TIFA on DP suffer from a lack of research. In the present study, TIFA was upregulated in the periodontal tissue of a DP mouse model. In addition, the expression of TIFA in RAW264.7 cells was induced by high glucose (HG) culture and increased by lipopolysaccharide (LPS) from Porphyromonas gingivalis treatment in a time‑dependent manner. Knockdown of TIFA significantly reduced the levels of inflammatory cytokines, including TNF‑α, IL‑6, IL‑1β and monocyte chemoattractant protein‑1, in HG and LPS‑induced RAW264.7 cells. The nuclear translocation of NF‑κB p65 was induced by HG and LPS and was clearly suppressed by absence of TIFA. The expression of downstream factors Nod‑like receptor family pyrin domain‑containing 3 and apoptosis‑associated speck‑like protein was inhibited by silencing TIFA. Moreover, TIFA was increased by receptor activator of NF‑κB (RANK) ligand (RANKL) in a concentration dependent manner. The expression of cathepsin K, MMP9 and nuclear factor of activated T cells cytoplasmic 1 was downregulated by depletion of TIFA. RANKL‑induced osteoclast differentiation was inhibited by silencing of TIFA. Meanwhile, the decrease of TIFA blocked activation of the NF‑κB pathway in RANKL‑treated RAW264.7 cells. In conclusion, TIFA as a promoter regulates the inflammation and osteoclast differentiation via activating the NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Xiaoqian Guo
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Ningxia Key Laboratory of Oral Disease Research, School of Stomatology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Guangwei Qiao
- Department of Oral and Maxillofacial Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jingjiao Wang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Changyi Yang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Min Zhao
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qian Zhang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yingbiao Wan
- Department of Prosthodontics and Oral Implantology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
63
|
Huang J, Li W. Molecular crosstalk between circadian clock and NLRP3 inflammasome signaling in Parkinson's disease. Heliyon 2024; 10:e24752. [PMID: 38268831 PMCID: PMC10803942 DOI: 10.1016/j.heliyon.2024.e24752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Research has recently found that both animal models and patients with PD have circadian dysfunction, accompanied by abnormal expression of circadian genes and proteins, which implies that the circadian clock plays a crucial role in PD etiopathogenesis. In addition, a strong relationship between NLRP3 inflammasome signaling and PD has been observed. Meanwhile, the activation of the NLRP3 inflammasome is highly relevant to dysfunctions of the molecular clock. Therefore, alleviating the neuroinflammation caused by NLRP3 inflammasome signaling by adjusting the abnormal molecular clock may be a potential strategy for preventing and treating PD. In this article, we have reviewed the potential or direct relationship between abnormalities of the circadian clock and NLRP3 inflammasome signaling in PD.
Collapse
Affiliation(s)
- Jiahua Huang
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| | - Wenwei Li
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| |
Collapse
|
64
|
Coombs JR, Zamoshnikova A, Holley CL, Maddugoda MP, Teo DET, Chauvin C, Poulin LF, Vitak N, Ross CM, Mellacheruvu M, Coll RC, Heinz LX, Burgener SS, Emming S, Chamaillard M, Boucher D, Schroder K. NLRP12 interacts with NLRP3 to block the activation of the human NLRP3 inflammasome. Sci Signal 2024; 17:eabg8145. [PMID: 38261657 DOI: 10.1126/scisignal.abg8145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome-nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form "specks," and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant-associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1β in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.
Collapse
Affiliation(s)
- Jared R Coombs
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Alina Zamoshnikova
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Caroline L Holley
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Madhavi P Maddugoda
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Daniel Eng Thiam Teo
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Camille Chauvin
- U1019, Institut Pasteur de Lille, University of Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalo-Universitaire Lille, Lille 59019, France
| | - Lionel F Poulin
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille 59000, France
| | - Nazarii Vitak
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Connie M Ross
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Manasa Mellacheruvu
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Rebecca C Coll
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Leonhard X Heinz
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Sabrina S Burgener
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Stefan Emming
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Mathias Chamaillard
- U1019, Institut Pasteur de Lille, University of Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalo-Universitaire Lille, Lille 59019, France
| | - Dave Boucher
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
65
|
Yue Z, Zhang X, Gu Y, Liu Y, Lan LM, Liu Y, Li Y, Yang G, Wan P, Chen X. Regulation and functions of the NLRP3 inflammasome in RNA virus infection. Front Cell Infect Microbiol 2024; 13:1309128. [PMID: 38249297 PMCID: PMC10796458 DOI: 10.3389/fcimb.2023.1309128] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Virus infection is one of the greatest threats to human life and health. In response to viral infection, the host's innate immune system triggers an antiviral immune response mostly mediated by inflammatory processes. Among the many pathways involved, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome has received wide attention in the context of viral infection. The NLRP3 inflammasome is an intracellular sensor composed of three components, including the innate immune receptor NLRP3, adaptor apoptosis-associated speck-like protein containing CARD (ASC), and the cysteine protease caspase-1. After being assembled, the NLRP3 inflammasome can trigger caspase-1 to induce gasdermin D (GSDMD)-dependent pyroptosis, promoting the maturation and secretion of proinflammatory cytokines such as interleukin-1 (IL-1β) and interleukin-18 (IL-18). Recent studies have revealed that a variety of viruses activate or inhibit the NLRP3 inflammasome via viral particles, proteins, and nucleic acids. In this review, we present a variety of regulatory mechanisms and functions of the NLRP3 inflammasome upon RNA viral infection and demonstrate multiple therapeutic strategies that target the NLRP3 inflammasome for anti-inflammatory effects in viral infection.
Collapse
Affiliation(s)
- Zhaoyang Yue
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Xuelong Zhang
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yu Gu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ying Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Lin-Miaoshen Lan
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yilin Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yongkui Li
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xin Chen
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| |
Collapse
|
66
|
Glück IM, Mathias GP, Strauss S, Rat V, Gialdini I, Ebert TS, Stafford C, Agam G, Manley S, Hornung V, Jungmann R, Sieben C, Lamb DC. Nanoscale organization of the endogenous ASC speck. iScience 2023; 26:108382. [PMID: 38047065 PMCID: PMC10690566 DOI: 10.1016/j.isci.2023.108382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 06/15/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
The NLRP3 inflammasome is a central component of the innate immune system. Its activation leads to formation of the ASC speck, a supramolecular assembly of the inflammasome adaptor protein ASC. Different models, based on ASC overexpression, have been proposed for the structure of the ASC speck. Using dual-color 3D super-resolution imaging (dSTORM and DNA-PAINT), we visualized the ASC speck structure following NLRP3 inflammasome activation using endogenous ASC expression. A complete structure was only obtainable by labeling with both anti-ASC antibodies and nanobodies. The complex varies in diameter between ∼800 and 1000 nm, and is composed of a dense core with emerging filaments. Dual-color confocal fluorescence microscopy indicated that the ASC speck does not colocalize with the microtubule-organizing center at late time points after Nigericin stimulation. From super-resolution images of whole cells, the ASC specks were sorted into a pseudo-time sequence indicating that they become denser but not larger during formation.
Collapse
Affiliation(s)
- Ivo M. Glück
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Grusha Primal Mathias
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Virgile Rat
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Irene Gialdini
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Thomas Sebastian Ebert
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Che Stafford
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ganesh Agam
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Suliana Manley
- Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, BSP 427 (Cubotron UNIL), Rte de la Sorge, CH-1015 Lausanne, Switzerland
| | - Veit Hornung
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Christian Sieben
- Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, BSP 427 (Cubotron UNIL), Rte de la Sorge, CH-1015 Lausanne, Switzerland
| | - Don C. Lamb
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| |
Collapse
|
67
|
Sharma M, de Alba E. Assembly mechanism of the inflammasome sensor AIM2 revealed by single molecule analysis. Nat Commun 2023; 14:7957. [PMID: 38042863 PMCID: PMC10693601 DOI: 10.1038/s41467-023-43691-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/06/2023] [Indexed: 12/04/2023] Open
Abstract
Pathogenic dsDNA prompts AIM2 assembly leading to the formation of the inflammasome, a multimeric complex that triggers the inflammatory response. The recognition of foreign dsDNA involves AIM2 self-assembly concomitant with dsDNA binding. However, we lack mechanistic and kinetic information on the formation and propagation of the assembly, which can shed light on innate immunity's time response and specificity. Combining optical traps and confocal fluorescence microscopy, we determine here the association and dissociation rates of the AIM2-DNA complex at the single molecule level. We identify distinct mechanisms for oligomer growth via the binding of incoming AIM2 molecules to adjacent dsDNA or direct interaction with bound AIM2 assemblies, resembling primary and secondary nucleation. Through these mechanisms, the size of AIM2 oligomers can increase fourfold in seconds. Finally, our data indicate that single AIM2 molecules do not diffuse/scan along the DNA, suggesting that oligomerization depends on stochastic encounters with DNA and/or DNA-bound AIM2.
Collapse
Affiliation(s)
- Meenakshi Sharma
- Department of Bioengineering, School of Engineering, University of California Merced, Merced, California, USA
| | - Eva de Alba
- Department of Bioengineering, School of Engineering, University of California Merced, Merced, California, USA.
| |
Collapse
|
68
|
Quan JH, Gao FF, Ma TZ, Ye W, Gao X, Deng MZ, Yin LL, Choi IW, Yuk JM, Cha GH, Lee YH, Chu JQ. Toxoplasma gondii Induces Pyroptosis in Human Placental Trophoblast and Amniotic Cells by Inducing ROS Production and Activation of Cathepsin B and NLRP1/NLRP3/NLRC4/AIM2 Inflammasome. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2047-2065. [PMID: 37741453 DOI: 10.1016/j.ajpath.2023.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/25/2023]
Abstract
Toxoplasma gondii infection in pregnant women may cause fetal anomalies; however, the underlying mechanisms remain unclear. The current study investigated whether T. gondii induces pyroptosis in human placental cells and the underlying mechanisms. Human placental trophoblast (BeWo and HTR-8/SVneo) and amniotic (WISH) cells were infected with T. gondii, and then reactive oxygen species (ROS) production, cathepsin B (CatB) release, inflammasome activation, and pyroptosis induction were evaluated. The molecular mechanisms of these effects were investigated by treating the cells with ROS scavengers, a CatB inhibitor, or inflammasome-specific siRNA. T. gondii infection induced ROS generation and CatB release into the cytosol in placental cells but decreased mitochondrial membrane potential. T. gondii-infected human placental cells and villi exhibited NLRP1, NLRP3, NLRC4, and AIM2 inflammasome activation and subsequent pyroptosis induction, as evidenced by increased expression of ASC, cleaved caspase-1, and mature IL-1β and gasdermin D cleavage. In addition to inflammasome activation and pyroptosis induction, adverse pregnancy outcome was shown in a T. gondii-infected pregnant mouse model. Administration of ROS scavengers, CatB inhibitor, or inflammasome-specific siRNA into T. gondii-infected cells reversed these effects. Collectively, these findings show that T. gondii induces NLRP1/NLRP3/NLRC4/AIM2 inflammasome-dependent caspase-1-mediated pyroptosis via induction of ROS production and CatB activation in placental cells. This mechanism may play an important role in inducing cell injury in congenital toxoplasmosis.
Collapse
Affiliation(s)
- Juan-Hua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Fei Fei Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Tian-Zhong Ma
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Wei Ye
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Ming-Zhu Deng
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Lan-Lan Yin
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - In-Wook Choi
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Guang-Ho Cha
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young-Ha Lee
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| | - Jia-Qi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.
| |
Collapse
|
69
|
Terzioglu G, Young-Pearse TL. Microglial function, INPP5D/SHIP1 signaling, and NLRP3 inflammasome activation: implications for Alzheimer's disease. Mol Neurodegener 2023; 18:89. [PMID: 38017562 PMCID: PMC10685641 DOI: 10.1186/s13024-023-00674-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Recent genetic studies on Alzheimer's disease (AD) have brought microglia under the spotlight, as loci associated with AD risk are enriched in genes expressed in microglia. Several of these genes have been recognized for their central roles in microglial functions. Increasing evidence suggests that SHIP1, the protein encoded by the AD-associated gene INPP5D, is an important regulator of microglial phagocytosis and immune response. A recent study from our group identified SHIP1 as a negative regulator of the NLRP3 inflammasome in human iPSC-derived microglial cells (iMGs). In addition, we found evidence for a connection between SHIP1 activity and inflammasome activation in the AD brain. The NLRP3 inflammasome is a multiprotein complex that induces the secretion of pro-inflammatory cytokines as part of innate immune responses against pathogens and endogenous damage signals. Previously published studies have suggested that the NLRP3 inflammasome is activated in AD and contributes to AD-related pathology. Here, we provide an overview of the current understanding of the microglial NLRP3 inflammasome in the context of AD-related inflammation. We then review the known intracellular functions of SHIP1, including its role in phosphoinositide signaling, interactions with microglial phagocytic receptors such as TREM2 and evidence for its intersection with NLRP3 inflammasome signaling. Through rigorous examination of the intricate connections between microglial signaling pathways across several experimental systems and postmortem analyses, the field will be better equipped to tailor newly emerging therapeutic strategies targeting microglia in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gizem Terzioglu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, Boston, MA, 02115, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, Boston, MA, 02115, USA.
| |
Collapse
|
70
|
Meng H, Zhou J, Wang M, Zheng M, Xing Y, Wang Y. SARS-CoV-2 Papain-like Protease Negatively Regulates the NLRP3 Inflammasome Pathway and Pyroptosis by Reducing the Oligomerization and Ubiquitination of ASC. Microorganisms 2023; 11:2799. [PMID: 38004809 PMCID: PMC10673202 DOI: 10.3390/microorganisms11112799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The interaction of viruses with hosts is complex, especially so with the antiviral immune systems of hosts, and the underlying mechanisms remain perplexing. Infection with SARS-CoV-2 may result in cytokine syndrome in the later stages, reflecting the activation of the antiviral immune response. However, viruses also encode molecules to negatively regulate the antiviral immune systems of hosts to achieve immune evasion and benefit viral replication during the early stage of infection. It has been observed that the papain-like protease (PLP) encoded by coronavirus could negatively regulate the host's IFNβ innate immunity. In this study, we first found that eight inflammasome-related genes were downregulated in CD14+ monocytes from COVID-19 patients. Subsequently, we observed that SARS-CoV-2 PLP negatively regulated the NLRP3 inflammasome pathway, inhibited the secretion of IL-1β, and decreased the caspase-1-mediated pyroptosis of human monocytes. The mechanisms for this may arise because PLP coimmunoprecipitates with ASC, reduces ASC ubiquitination, and inhibits ASC oligomerization and the formation of ASC specks. These findings suggest that PLP may inhibit strong immune defenses and provide the maximum advantage for viral replication. This research may allow us to better understand the flex function of CoV-encoding proteases and provide a new perspective on the innate immune responses against SARS-CoV-2 and other viruses.
Collapse
Affiliation(s)
- Huan Meng
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Chaoyang District, Beijing 100015, China
- Bioinformatics Center of Academy of Military Medicine Science, Beijing 100850, China
| | - Jianglin Zhou
- Bioinformatics Center of Academy of Military Medicine Science, Beijing 100850, China
| | - Mingyu Wang
- Bioinformatics Center of Academy of Military Medicine Science, Beijing 100850, China
| | - Mei Zheng
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Chaoyang District, Beijing 100015, China
| | - Yaling Xing
- Bioinformatics Center of Academy of Military Medicine Science, Beijing 100850, China
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Chaoyang District, Beijing 100015, China
| |
Collapse
|
71
|
Ramos-Tovar E, Muriel P. NLRP3 inflammasome in hepatic diseases: A pharmacological target. Biochem Pharmacol 2023; 217:115861. [PMID: 37863329 DOI: 10.1016/j.bcp.2023.115861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome pathway is mainly responsible for the activation and release of a cascade of proinflammatory mediators that contribute to the development of hepatic diseases. During alcoholic liver disease development, the NLRP3 inflammasome pathway contributes to the maturation of caspase-1, interleukin (IL)-1β, and IL-18, which induce a robust inflammatory response, leading to fibrosis by inducing profibrogenic hepatic stellate cell (HSC) activation. Substantial evidence demonstrates that nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) via NLRP3 inflammasome activation, ultimately leading to fibrosis and hepatocellular carcinoma (HCC). Activation of the NLRP3 inflammasome in NASH can be attributed to several factors, such as reactive oxygen species (ROS), gut dysbiosis, leaky gut, which allow triggers such as cardiolipin, cholesterol crystals, endoplasmic reticulum stress, and uric acid to reach the liver. Because inflammation triggers HSC activation, the NLRP3 inflammasome pathway performs a central function in fibrogenesis regardless of the etiology. Chronic hepatic activation of the NLRP3 inflammasome can ultimately lead to HCC; however, inflammation also plays a role in decreasing tumor growth. Some data indicate that NLRP3 inflammasome activation plays an important role in autoimmune hepatitis, but the evidence is scarce. Most researchers have reported that NLRP3 inflammasome activation is essential in liver injury induced by a variety of drugs and hepatotropic virus infection; however, few reports indicate that this pathway can play a beneficial role by inducing liver regeneration. Modulation of the NLRP3 inflammasome appears to be a suitable strategy to treat liver diseases.
Collapse
Affiliation(s)
- Erika Ramos-Tovar
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Apartado Postal 11340, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México.
| |
Collapse
|
72
|
Hempel A, D'Osualdo A, Snipas S, Salvesen G. Cell organelles are retained inside pyroptotic corpses during inflammatory cell death. Biosci Rep 2023; 43:BSR20231265. [PMID: 37797233 PMCID: PMC10611922 DOI: 10.1042/bsr20231265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/07/2023] Open
Abstract
Many proinflammatory proteins are released via the necrotic form of cell death known as pyroptosis. Sometimes known as gasdermin D (GSDMD) dependent cell death, pyroptosis results from the formation of pores in the plasma membrane leading to eventual cell lysis. Seeking to understand the magnitude of this cell lysis we measured the size of proteins released during pyroptosis. We demonstrate that there is no restriction on the size of soluble proteins released during pyroptosis even at early timepoints. However, even though large molecules can exit the dying cell, organelles are retained within it. This observation indicates that complete cell rupture may not be a consequence of pyroptosis, and that plasma membrane architecture is retained.
Collapse
Affiliation(s)
- Anne Hempel
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, U.S.A
| | - Andrea D'Osualdo
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, U.S.A
| | - Scott J. Snipas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, U.S.A
| | - Guy S. Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, U.S.A
| |
Collapse
|
73
|
Feng M, Luo F, Wu H, Chen Y, Zuo J, Weng X, Chen G, Zhong J. Network Pharmacology Analysis and Machine-Learning Models Confirmed the Ability of YiShen HuoXue Decoction to Alleviate Renal Fibrosis by Inhibiting Pyroptosis. Drug Des Devel Ther 2023; 17:3169-3192. [PMID: 37900883 PMCID: PMC10612518 DOI: 10.2147/dddt.s420135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/07/2023] [Indexed: 10/31/2023] Open
Abstract
Purpose YiShen HuoXue decoction (YSHXD) is a formulation that has been used clinically for the treatment of renal fibrosis (RF) for many years. We aimed to clarify therapeutic effects of YSHXD against RF and potential pharmacological mechanisms. Materials and Methods We used network pharmacology analysis and machine-learning to screen the core components and core targets of YSHXD against RF, followed by molecular docking and molecular dynamics simulations to confirm the reliability of the results. Finally, we validated the network pharmacology analysis experimentally in HK-2 cells and a rat model of RF established by unilateral ureteral ligation (UUO). Results Quercetin, kaempferol, luteolin, beta-sitosterol, wogonin, stigmasterol, isorhamnetin, baicalein, and dihydrotanshinlactone progesterone were identified as the main active components of YSHXD in the treatment of unilateral ureteral ligation-induced RF, with IL-6, IL1β, TNF, AR, and PTGS2 as core target proteins. Molecular docking and molecular dynamics simulations further confirmed the relationship between compounds and target proteins. The potential molecular mechanism of YSHXD predicted by network pharmacology analysis was confirmed in HK-2 cells and UUO rats. YSHXD downregulated NLRP3, ASC, NF-κBp65, Caspase-1, GSDMD, PTGS2, IL-1β, IL-6, IL-18, TNF-α, α-SMA and upregulated HGF, effectively alleviating the RF process. Conclusion YSHXD exerts important anti-inflammatory and anti-cellular inflammatory necrosis effects by inhibiting the NLRP3/caspase-1/GSDMD-mediated pyroptosis pathway, indicating that YSHXD represents a new strategy and complementary approach to RF therapy.
Collapse
Affiliation(s)
- MinChao Feng
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - Fang Luo
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - HuiMin Wu
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - Yushan Chen
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - Jinjin Zuo
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - Xueying Weng
- The First Clinical Medical College, Guangxi University of Traditional Chinese Medicine, Nannig, People’s Republic of China
| | - Guozhong Chen
- Department of Gastroenterology, the First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, People’s Republic of China
| | - Jian Zhong
- Department of Nephrology, the First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, People’s Republic of China
| |
Collapse
|
74
|
Xie D, Guo H, Li M, Jia L, Zhang H, Liang D, Wu N, Yang Z, Tian Y. Splenic monocytes mediate inflammatory response and exacerbate myocardial ischemia/reperfusion injury in a mitochondrial cell-free DNA-TLR9-NLRP3-dependent fashion. Basic Res Cardiol 2023; 118:44. [PMID: 37814087 DOI: 10.1007/s00395-023-01014-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
The spleen contributes importantly to myocardial ischemia/reperfusion (MI/R) injury. Nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) recruits inflammasomes, initiating inflammatory responses and mediating tissue injury. We hypothesize that myocardial cell-free DNA (cfDNA) activates the splenic NLRP3 inflammasome during early reperfusion, increases systemic inflammatory response, and exacerbates myocardial infarct. Mice were subjected to 40 min of ischemia followed by 0, 1, 5, or 15 min, or 24 h of reperfusion. Splenic leukocyte adoptive transfer was performed by injecting isolated splenocytes to mice with splenectomy performed prior to left coronary artery occlusion. CY-09 (4 mg/kg) was administered 5 min before reperfusion. During post-ischemic reperfusion, splenic protein levels of NLRP3, cleaved caspase-1, and interleukin-1β (IL-1β) were significantly elevated and peaked (2.1 ± 0.2-, 3.4 ± 0.4-, and 3.2 ± 0.2-fold increase respectively, p < 0.05) within 5 min of reperfusion. In myocardial tissue, NLRP3 was not upregulated until 24 h after reperfusion. Suppression by CY09, a specific NLRP3 inflammasome inhibitor, or deficiency of NLRP3 significantly reduced myocardial infarct size (17.3% ± 4.2% and 33.2% ± 1.8% decrease respectively, p < 0.01). Adoptive transfer of NLRP3-/- splenocytes to WT mice significantly decreased infarct size compared to transfer of WT splenocytes (19.1% ± 2.8% decrease, p < 0.0001). NLRP3 was mainly activated at 5 min after reperfusion in CD11b+ and LY6G- splenocytes, which significantly increased during reperfusion (24.8% ± 0.7% vs.14.3% ± 0.6%, p < 0.0001). The circulating cfDNA level significantly increased in patients undergoing cardiopulmonary bypass (CPB) (43.3 ± 5.3 ng/mL, compared to pre-CPB 23.8 ± 3.5 ng/mL, p < 0.01). Mitochondrial cfDNA (mt-cfDNA) contributed to NLRP3 activation in macrophages (2.1 ± 0.2-fold increase, p < 0.01), which was inhibited by a Toll-like receptor 9(TLR9) inhibitor. The NLRP3 inflammasome in splenic monocytes is activated and mediates the inflammatory response shortly after reperfusion onset, exacerbating MI/R injury in mt-cfDNA/TLR9-dependent fashion. The schema reveals splenic NLRP3 mediates the inflammatory response in macrophages and exacerbates MI/R in a mitochondrial cfDNA/ TLR9-dependent fashion.
Collapse
Affiliation(s)
- Dina Xie
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hanliang Guo
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Mingbiao Li
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Liqun Jia
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hao Zhang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Degang Liang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Naishi Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zequan Yang
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Yikui Tian
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
75
|
Gao Y, Yu S, Chen M, Wang X, Pan L, Wei B, Meng G. cFLIP S regulates alternative NLRP3 inflammasome activation in human monocytes. Cell Mol Immunol 2023; 20:1203-1215. [PMID: 37591930 PMCID: PMC10541859 DOI: 10.1038/s41423-023-01077-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
The innate immune responses, including inflammasome activation, are paramount for host defense against pathogen infection. In contrast to canonical and noncanonical inflammasome activation, in this study, heat-killed gram-negative bacteria (HK bacteria) were identified as single-step stimulators of the NLRP3 inflammasome in human monocytes, and they caused a moderate amount of IL-1β to be released from cells. Time course experiments showed that this alternative inflammasome response was finished within a few hours. Further analysis showed that the intrinsically limited NLRP3 inflammasome activation response was due to the negative regulation of caspase-8 by the short isoform of cFLIP (cFLIPs), which was activated by NF-κB. In contrast, overexpressed cFLIPS, but not overexpressed cFLIPL, inhibited the activation of caspase-8 and the release of IL-1β in response to HK bacteria infection in human monocytes. Furthermore, we demonstrated that TAK1 activity mediated the expression of cFLIPs and was upstream and essential for the caspase-8 cleavage induced by HK bacteria in human monocytes. The functional specificity of cFLIPs and TAK1 revealed unique responses of human monocytes to a noninvasive pathogen, providing novel insights into an alternative regulatory pathway of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yuhui Gao
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shi Yu
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Department of Basic Research, Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, Guangdong, China
| | - Mengdan Chen
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xun Wang
- Shanghai Blood Center, Shanghai, 200051, China
| | - Lei Pan
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Pasteurien College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Bin Wei
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Pasteurien College, Soochow University, Suzhou, 215006, Jiangsu, China.
- Nanjing Advanced Academy of Life and Health, Nanjing, 211135, Jiangsu, China.
| |
Collapse
|
76
|
Liu X, Luo P, Zhang W, Zhang S, Yang S, Hong F. Roles of pyroptosis in atherosclerosis pathogenesis. Biomed Pharmacother 2023; 166:115369. [PMID: 37643484 DOI: 10.1016/j.biopha.2023.115369] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Pyroptosis is a pro-inflammatory type of regulated cell death (RCD) characterized by gasdermin protein-mediated membrane pore formation, cell swelling, and rapid lysis. Recent studies have suggested that pyroptosis is closely related to atherosclerosis (AS). Previous studies reported that pyroptosis involving endothelial cells (ECs), macrophages, and smooth muscle cells (SMCs) plays an important role in the formation and development of AS. Pyroptosis not only causes local inflammation but also amplifies the inflammatory response and it aggravates plaque instability, leading to plaque rupture and thrombosis, eventually resulting in acute cardiovascular events. In this review, we clarified some novel pathways and mechanics and presented some potential drugs.
Collapse
Affiliation(s)
- Xiaohan Liu
- Pathogen Biology Experimental Center, College of Medicine, Nanchang University, Jiangxi 330000, China
| | - Peiyi Luo
- Pathogen Biology Experimental Center, College of Medicine, Nanchang University, Jiangxi 330000, China; Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Weiyun Zhang
- Pathogen Biology Experimental Center, College of Medicine, Nanchang University, Jiangxi 330000, China; Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Shuxian Zhang
- Pathogen Biology Experimental Center, College of Medicine, Nanchang University, Jiangxi 330000, China; Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Shulong Yang
- School of basic medical sciences, Fuzhou Medical College of Nanchang University, Fuzhou 344000, China; Key Laboratory of Chronic Diseases, Fuzhou Medical University, Fuzhou 344000, China; Technology Innovation Center of Chronic Disease Research in Fuzhou City, Fuzhou Science and Technology Bureau, Fuzhou 344000, China.
| | - Fenfang Hong
- Pathogen Biology Experimental Center, College of Medicine, Nanchang University, Jiangxi 330000, China.
| |
Collapse
|
77
|
Wang L, Li W, Xin S, Wu S, Peng C, Ding H, Feng S, Zhao C, Wu J, Wang X. Soybean glycinin and β-conglycinin damage the intestinal barrier by triggering oxidative stress and inflammatory response in weaned piglets. Eur J Nutr 2023; 62:2841-2854. [PMID: 37358571 DOI: 10.1007/s00394-023-03188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/31/2023] [Indexed: 06/27/2023]
Abstract
PURPOSE Soybean glycinin (11S) and β-conglycinin (7S) are major antigenic proteins in soybean and can induce a variety of allergic reactions in the young animals. This study aimed to investigate the effect of 7S and 11S allergens on the intestine of piglets. METHODS Thirty healthy 21-day-old weaned "Duroc × Long White × Yorkshire" piglets were randomly divided into three groups fed with the basic diet, the 7S supplemented basic diet, or the 11S supplemented basic diet for 1 week. Allergy markers, intestinal permeability, oxidative stress, and inflammatory reactions were detected, and we observed different sections of intestinal tissue. The expressions of genes and proteins related to NOD-like receptor thermal protein domain associated protein 3 (NLRP-3) signaling pathway were detected by IHC, RT-qPCR, and WB. RESULTS Severe diarrhea and decreased growth rate were observed in the 7S and 11S groups. Typical allergy markers include IgE production and significant elevations of histamine and 5-hydroxytryptamine (5-HT). More aggressive intestinal inflammation and barrier dysfunction were observed in the experimental weaned piglets. In addition, 7S and 11S supplementation increased the levels of 8-hydroxy-2 deoxyguanosine (8-OHdG) and nitrotyrosine, triggering oxidative stress. Furthermore, higher expression levels of NLRP-3 inflammasome ASC, caspase-1, IL-1β, and IL-18 were observed in the duodenum, jejunum, and ileum. CONCLUSION We confirmed that 7S and 11S damaged the intestinal barrier of weaned piglets and may be associated with the onset of oxidative stress and inflammatory response. However, the molecular mechanism underlying these reactions deserves further study.
Collapse
Affiliation(s)
- Lei Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Wen Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Shuzhen Xin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Shuang Wu
- Wolong District Animal Health Supervision Institute, Nanyang, 473000, China
| | - Chenglu Peng
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongyan Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Shibing Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China.
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230061, China.
| |
Collapse
|
78
|
Metcalfe S, Panasiewicz M, Kay JG. Inflammatory macrophages exploited by oral streptococcus increase IL-1B release via NLRP6 inflammasome. J Leukoc Biol 2023; 114:347-357. [PMID: 37497744 PMCID: PMC10533225 DOI: 10.1093/jleuko/qiad089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic inflammatory periodontal disease develops in part from the infiltration of a large number of classically activated inflammatory macrophages that release inflammatory cytokines important for disease progression, including inflammasome-dependent interleukin (IL)-1β. Streptococcus gordonii is a normally commensal oral microorganism; while not causative, recent evidence indicates that commensal oral microbes are required for the full development of periodontal disease. We have recently reported that inflammatory macrophages counterintuitively allow for the increased survival of phagocytosed S. gordonii over nonactivated or alternatively activated macrophages. This survival is dependent on increased reactive oxygen species production within the phagosome of the inflammatory macrophages, and resistance by the bacterium and can result in S. gordonii damaging the phagolysosomes. Here, we show that activated macrophages infected with live S. gordonii release more IL-1β than non-activated macrophages infected with either live or dead S. gordonii, and that the survival of oral Streptococci are more dependent on macrophage activation than other Gram positive microbes, both classical pathogens and commensals. We also find that S. gordonii-dependent inflammatory macrophage inflammasome activation requires the cytoplasmic NLRP6. Overall, our results suggest S. gordonii is capable of evading immune destruction, increasing inflammatory mediators, and increasing inflammatory macrophage response, and that this ability is increased under conditions of inflammation. This work reveals additional mechanisms by which normally commensal oral streptococci-macrophage interactions can change, resulting in increased release of mature IL-1β, potentially contributing to an environment that perpetuates inflammation.
Collapse
Affiliation(s)
- Sarah Metcalfe
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main street, Buffalo, NY 14214, United States
| | - Michelle Panasiewicz
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main street, Buffalo, NY 14214, United States
| | - Jason G Kay
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main street, Buffalo, NY 14214, United States
| |
Collapse
|
79
|
Acevedo W, Morán-Figueroa R, Vargas-Chacoff L, Morera FJ, Pontigo JP. Revealing the Salmo salar NLRP3 Inflammasome: Insights from Structural Modeling and Transcriptome Analysis. Int J Mol Sci 2023; 24:14556. [PMID: 37834004 PMCID: PMC10572965 DOI: 10.3390/ijms241914556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
The NLRP3, one of the most heavily studied inflammasome-related proteins in mammals, remains inadequately characterized in Atlantic salmon (Salmo salar), despite the significant commercial importance of this salmonid. The NLRP3 inflammasome is composed of the NLRP3 protein, which is associated with procaspase-1 via an adapter molecule known as ASC. This work aims to characterize the Salmo salar NLRP3 inflammasome through in silico structural modeling, functional transcript expression determination in the SHK-1 cell line in vitro, and a transcriptome analysis on Atlantic salmon. The molecular docking results suggested a similar arrangement of the ternary complex between NLRP3, ASC, and caspase-1 in both the Atlantic salmon and the mammalian NLRP3 inflammasomes. Moreover, the expression results confirmed the functionality of the SsNLRP3 inflammasome in the SHK-1 cells, as evidenced by the lipopolysaccharide-induced increase in the transcription of genes involved in inflammasome activation, including ASC and NLRP3. Additionally, the transcriptome results revealed that most of the inflammasome-related genes, including ASC, NLRP3, and caspase-1, were down-regulated in the Atlantic salmon following its adaptation to seawater (also known as parr-smolt transformation). This is correlated with a temporary detrimental effected on the immune system. Collectively, these findings offer novel insights into the evolutionarily conserved role of NLRP3.
Collapse
Affiliation(s)
- Waldo Acevedo
- Biological Chemistry Laboratory, Institute of Chemistry, Faculty of Science, Pontificia Universidad Católica de Valparaíso, Valparaiso 2373223, Chile;
| | - Rodrigo Morán-Figueroa
- Escuela de Medicina Veterinaria, Facultad de Agronomía y Sistemas Naturales, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile;
- Escuela de Medicina Veterinaria, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Luis Vargas-Chacoff
- Institute of Marine Sciences and Limnology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5110566, Chile;
- IDEAL Research Center for Dynamics of High Latitude Marine Ecosystems, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute Biodiversity of Antarctic and Subantarctic Ecosystems, BASE, University Austral of Chile, Valdivia 5090000, Chile
- Integrative Biology Group, Valdivia 5110566, Chile
| | - Francisco J. Morera
- Escuela de Medicina Veterinaria, Facultad de Agronomía y Sistemas Naturales, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile;
- Escuela de Medicina Veterinaria, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
- Integrative Biology Group, Valdivia 5110566, Chile
| | - Juan Pablo Pontigo
- Laboratorio Institucional de Investigación, Facultad Ciencias de la Naturaleza, Medicina Veterinaria, Universidad San Sebastián, Lago Panguipulli 1390, Puerto Montt 5090000, Chile
| |
Collapse
|
80
|
Zhu R, Wang Y, Ouyang Z, Hao W, Zhou F, Lin Y, Cheng Y, Zhou R, Hu W. Targeting regulated chondrocyte death in osteoarthritis therapy. Biochem Pharmacol 2023; 215:115707. [PMID: 37506921 DOI: 10.1016/j.bcp.2023.115707] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
In vivo articular cartilage degeneration is an essential hallmark of osteoarthritis (OA), involving chondrocyte senescence, extracellular matrix degradation, chondrocyte death, cartilage loss, and bone erosion. Among them, chondrocyte death is one of the major factors leading to cartilage degeneration. Many studies have reported that various cell death modes, including apoptosis, ferroptosis, and autophagy, play a key role in OA chondrocyte death. Currently, there is insufficient understanding of OA pathogenesis, and there remains a lack of treatment methods to prevent OA and inhibit its progression. Studies suggest that OA prevention and treatment are mainly directed to arrest premature or excessive chondrocyte death. In this review, we a) discuss the forms of death of chondrocytes and the associations between them, b) summarize the critical factors in chondrocyte death, c) discuss the vital role of chondrocyte death in OA, d) and, explore new approaches for targeting the regulation of chondrocyte death in OA treatment.
Collapse
Affiliation(s)
- Rendi Zhu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yan Wang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ziwei Ouyang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wenjuan Hao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Fuli Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yi Lin
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuanzhi Cheng
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
81
|
Althagafy HS, Sharawi ZW, Batawi AH, Almohaimeed HM, Al-Thubiani WS, Hassanein EHM, Rateb A. Buspirone attenuated methotrexate-induced hippocampal toxicity in rats by regulating Nrf2/HO-1, PPAR-γ, NF-κB/nNOS, and ROS/NLRP3/caspase-1 signaling pathways. J Biochem Mol Toxicol 2023; 37:e23414. [PMID: 37341015 DOI: 10.1002/jbt.23414] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Methotrexate (MTX) is a chemotherapeutic agent widely used to treat a variety of tumors. Nonetheless, MTX-induced hippocampal neurotoxicity is a well-defined dose-limiting adverse effect that limits clinical utility. Proinflammatory cytokine production and oxidative stress are possible mechanisms for MTX-induced neurotoxicity. Buspirone (BSP), a partial agonist of the 5-HT1a receptor (5-HT1aR), has emerged as an anxiolytic drug. BSP has been shown to possess antioxidant and anti-inflammatory effects. The current study investigated BSP's potential anti-inflammatory and antioxidant effects in attenuating MTX-induced hippocampal toxicity. Rats received either BSP (1.5 mg/kg) orally for 10 days and MTX (20 mg/kg) i.p. on Day 5. BSP administration markedly protected hippocampal neurons from drastic degenerated neuronal changes induced by MTX. BSP significantly attenuated oxidative injury by downregulating Kelch-like ECH-associated protein 1 expression while potently elevating hippocampal Nrf2, heme oxygenase-1, and peroxisome proliferator-activated receptor expression. BSP dampened inflammation by reducing NO2 - , tumor necrosis factor-alpha, IL-6, and interleukin 1 beta levels mediated by downregulating NF-κB and neuronal nitric oxides synthase expression. Moreover, BSP potently counteracted hippocampal pyroptosis by downregulating NLRP3, ASC, and cleaved-caspase-1 proteins. Therefore, BSP may represent a promising approach to attenuate neurotoxicity in patients receiving MTX.
Collapse
Affiliation(s)
- Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zeina W Sharawi
- Department Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashwaq H Batawi
- Department Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hailah M Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdul Rahman University, Riyadh, Saudi Arabia
| | - Wafa S Al-Thubiani
- Department of Biology, Faculty of Applied Sciences, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Amal Rateb
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assuit University, Assiut, Egypt
- Department of Basic Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arbia
| |
Collapse
|
82
|
Li B, Xu L, Liu J, Zhou M, Jiang X. Phloretin ameliorates heart function after myocardial infarction via NLRP3/Caspase-1/IL-1β signaling. Biomed Pharmacother 2023; 165:115083. [PMID: 37413902 DOI: 10.1016/j.biopha.2023.115083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVES/AIMS Inflammation is crucial in structural and electrical remodeling after myocardial infarction (MI), affecting cardiac pump function and conduction pathways. Phloretin possesses an anti-inflammation role by inhibiting the NLRP3/Caspase-1/IL-1β pathway. However, the effects of Phloretin on cardiac contractile and electrical conduction function after MI remained unclear. Therefore, we aimed to investigate the potential role of Phloretin in a rat model of MI. METHODS Rats were assigned into four groups: Sham, Sham+Phloretin, MI and MI+Phloretin, with ad libitum food and water. In the MI and MI+Phloretin groups, the left anterior descending coronary artery was occluded for 4 weeks, while the Sham and Sham+Phloretin groups received sham operation. The Sham+Phloretin group and the MI+Phloretin group received oral administration of Phloretin. In vitro, H9c2 cells were subjected to hypoxic conditions to simulate an MI model, with Phloretin for 24 h. Cardiac electrophysiological properties were assessed following MI, including the effective refractory period (ERP), action potential duration (APD)90 and ventricular fibrillation (VF) incidence. Echocardiography evaluated left ventricular ejection fraction (LVEF), left ventricular fraction shortening (LVFS), left ventricular internal diameter at end-diastole (LVIDd), left ventricular internal diameter at end-systole (LVIDs), left ventricular end-systolic volume (LVESV) and left ventricular end-diastolic volume (LVEDV) to assess cardiac function. Serum type B natriuretic peptide (BNP) level was applied to evaluate the degree of Heart failure (HF). The fibrosis area and severity were assessed by Masson staining and protein expression levels of collagen 3, collagen 1, TGF-β and α-SMA. Western blot analysis estimated the protein expression levels of NLRP3, Pro Caspase-1, Caspase-1, ASC, IL-18, IL-1β, pp38, p38, and Connexin43(Cx43) to elucidate the influence of inflammation on electrical remodeling after MI. RESULTS Our findings demonstrate that Phloretin inhibits the NLRP3/Caspase-1/IL-1β pathway, leading to the upregulation of Cx43 by limiting p38 phosphorylation, which further decreases susceptibility to ventricular arrhythmias (VAs). Additionally, Phloretin attenuated fibrosis by inhibiting inflammation to prevent HF. In vitro experiments also provided strong evidence supporting the inhibitory effects of Phloretin on the NLRP3/Caspase-1/IL-1β pathway. CONCLUSION Our results suggest that Phloretin could suppress the NLRP3/Caspase-1/IL-1β pathway to reverse structural and electrical remodeling after MI to prevent the occurrence of VAs and HF.
Collapse
Affiliation(s)
- Bin Li
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jiangwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mingmin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
83
|
Van Bruggen S, Jarrot PA, Thomas E, Sheehy CE, Silva CMS, Hsu AY, Cunin P, Nigrovic PA, Gomes ER, Luo HR, Waterman CM, Wagner DD. NLRP3 is essential for neutrophil polarization and chemotaxis in response to leukotriene B4 gradient. Proc Natl Acad Sci U S A 2023; 120:e2303814120. [PMID: 37603754 PMCID: PMC10468616 DOI: 10.1073/pnas.2303814120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
Neutrophil recruitment to sites of infection and inflammation is an essential process in the early innate immune response. Upon activation, a subset of neutrophils rapidly assembles the multiprotein complex known as the NLRP3 inflammasome. The NLRP3 inflammasome forms at the microtubule organizing center, which promotes the formation of interleukin (IL)-1β and IL-18, essential cytokines in the immune response. We recently showed that mice deficient in NLRP3 (NLRP3-/-) have reduced neutrophil recruitment to the peritoneum in a model of thioglycolate-induced peritonitis. Here, we tested the hypothesis that this diminished recruitment could be, in part, the result of defects in neutrophil chemotaxis. We find that NLRP3-/- neutrophils show loss of cell polarization, as well as reduced directionality and velocity of migration toward increasing concentrations of leukotriene B4 (LTB4) in a chemotaxis assay in vitro, which was confirmed through intravital microscopy of neutrophil migration toward a laser-induced burn injury of the liver. Furthermore, pharmacologically blocking NLRP3 inflammasome assembly with MCC950 in vitro reduced directionality but preserved nondirectional movement, indicating that inflammasome assembly is specifically required for polarization and directional chemotaxis, but not cell motility per se. In support of this, pharmacological breakdown of the microtubule cytoskeleton via nocodazole treatment induced cell polarization and restored nondirectional cell migration in NLRP3-deficient neutrophils in the LTB4 gradient. Therefore, NLRP3 inflammasome assembly is required for establishment of cell polarity to guide the directional chemotactic migration of neutrophils.
Collapse
Affiliation(s)
- Stijn Van Bruggen
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
| | - Pierre-André Jarrot
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
| | - Eline Thomas
- Department of Life Science Technology, Imec, Leuven3001, Belgium
- Department of Biophysics, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Casey E. Sheehy
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Camila M. S. Silva
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
| | - Alan Y. Hsu
- Department of Pathology, Harvard Medical School, Boston, MA02115
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Boston, MA02115
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Pierre Cunin
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Peter A. Nigrovic
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Edgar R. Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon1649-028, Portugal
| | - Hongbo R. Luo
- Department of Pathology, Harvard Medical School, Boston, MA02115
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Boston, MA02115
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Clare M. Waterman
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute of the NIH, Bethesda, MD20892
| | - Denisa D. Wagner
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA02115
| |
Collapse
|
84
|
Lamichhane PP, Samir P. Cellular Stress: Modulator of Regulated Cell Death. BIOLOGY 2023; 12:1172. [PMID: 37759572 PMCID: PMC10525759 DOI: 10.3390/biology12091172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Cellular stress response activates a complex program of an adaptive response called integrated stress response (ISR) that can allow a cell to survive in the presence of stressors. ISR reprograms gene expression to increase the transcription and translation of stress response genes while repressing the translation of most proteins to reduce the metabolic burden. In some cases, ISR activation can lead to the assembly of a cytoplasmic membraneless compartment called stress granules (SGs). ISR and SGs can inhibit apoptosis, pyroptosis, and necroptosis, suggesting that they guard against uncontrolled regulated cell death (RCD) to promote organismal homeostasis. However, ISR and SGs also allow cancer cells to survive in stressful environments, including hypoxia and during chemotherapy. Therefore, there is a great need to understand the molecular mechanism of the crosstalk between ISR and RCD. This is an active area of research and is expected to be relevant to a range of human diseases. In this review, we provided an overview of the interplay between different cellular stress responses and RCD pathways and their modulation in health and disease.
Collapse
Affiliation(s)
| | - Parimal Samir
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
85
|
Boccaccio GL, Thomas MG, García CC. Membraneless Organelles and Condensates Orchestrate Innate Immunity Against Viruses. J Mol Biol 2023; 435:167976. [PMID: 36702393 DOI: 10.1016/j.jmb.2023.167976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023]
Abstract
The cellular defense against viruses involves the assembly of oligomers, granules and membraneless organelles (MLOs) that govern the activation of several arms of the innate immune response. Upon interaction with specific pathogen-derived ligands, a number of pattern recognition receptors (PRRs) undergo phase-separation thus triggering downstream signaling pathways. Among other relevant condensates, inflammasomes, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) specks, cyclic GMP-AMP synthase (cGAS) foci, protein kinase R (PKR) clusters, ribonuclease L-induced bodies (RLBs), stress granules (SGs), processing bodies (PBs) and promyelocytic leukemia protein nuclear bodies (PML NBs) play different roles in the immune response. In turn, viruses have evolved diverse strategies to evade the host defense. Viral DNA or RNA, as well as viral proteases or proteins carrying intrinsically disordered regions may interfere with condensate formation and function in multiple ways. In this review we discuss current and hypothetical mechanisms of viral escape that involve the disassembly, repurposing, or inactivation of membraneless condensates that govern innate immunity. We summarize emerging interconnections between these diverse condensates that ultimately determine the cellular outcome.
Collapse
Affiliation(s)
- Graciela Lidia Boccaccio
- Laboratorio de Biología Celular del ARN, Instituto Leloir (FIL) and Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Departamento de Fisiología y Biología Molecular y Celular (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - María Gabriela Thomas
- Laboratorio de Biología Celular del ARN, Instituto Leloir (FIL) and Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina. https://www.twitter.com/_gabithomas
| | - Cybele Carina García
- Departamento de Química Biológica (QB), Facultad de Ciencias Exactas y Naturales (FCEN), and IQUIBICEN, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) and Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
86
|
Shi W, Jin M, Chen H, Wu Z, Yuan L, Liang S, Wang X, Memon FU, Eldemery F, Si H, Ou C. Inflammasome activation by viral infection: mechanisms of activation and regulation. Front Microbiol 2023; 14:1247377. [PMID: 37608944 PMCID: PMC10440708 DOI: 10.3389/fmicb.2023.1247377] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023] Open
Abstract
Viral diseases are the most common problems threatening human health, livestock, and poultry industries worldwide. Viral infection is a complex and competitive dynamic biological process between a virus and a host/target cell. During viral infection, inflammasomes play important roles in the host and confer defense mechanisms against the virus. Inflammasomes are polymeric protein complexes and are considered important components of the innate immune system. These immune factors recognize the signals of cell damage or pathogenic microbial infection after activation by the canonical pathway or non-canonical pathway and transmit signals to the immune system to initiate the inflammatory responses. However, some viruses inhibit the activation of the inflammasomes in order to replicate and proliferate in the host. In recent years, the role of inflammasome activation and/or inhibition during viral infection has been increasingly recognized. Therefore, in this review, we describe the biological properties of the inflammasome associated with viral infection, discuss the potential mechanisms that activate and/or inhibit NLRP1, NLRP3, and AIM2 inflammasomes by different viruses, and summarize the reciprocal regulatory effects of viral infection on the NLRP3 inflammasome in order to explore the relationship between viral infection and inflammasomes. This review will pave the way for future studies on the activation mechanisms of inflammasomes and provide novel insights for the development of antiviral therapies.
Collapse
Affiliation(s)
- Wen Shi
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Mengyun Jin
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hao Chen
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | | | - Liuyang Yuan
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Si Liang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaohan Wang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Fareed Uddin Memon
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Fatma Eldemery
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| | - Changbo Ou
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| |
Collapse
|
87
|
Li Z, Lin C, Cai X, Hu S, Lv F, Yang W, Zhu X, Ji L. Anti-inflammatory therapies were associated with reduced risk of myocardial infarction in patients with established cardiovascular disease or high cardiovascular risks: A systematic review and meta-analysis of randomized controlled trials. Atherosclerosis 2023; 379:117181. [PMID: 37527612 DOI: 10.1016/j.atherosclerosis.2023.06.972] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND AIMS We aimed to evaluate the association between anti-inflammatory therapies and the incidence of cardiovascular events in patients with established cardiovascular disease (CVD) or high cardiovascular risks. METHODS Literature retrieval was conducted in PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials and Clinicaltrial.gov website from the inception to December 2022. Randomized controlled trials comparing anti-inflammatory therapies with placebo in patients with established CVD or high cardiovascular risks were included. The results of the meta-analysis were computed as the risk ratio (RR) with 95% confidence interval (CI). RESULTS Compared with placebo, anti-inflammatory therapies were associated with decreased incidence of myocardial infarction (MI) (RR = 0.93, 95% CI, 0.88 to 0.98), which was mainly driven by therapies targeting central IL-6 signaling pathway (RR = 0.83, 95% CI, 0.74 to 0.93). IL-1 inhibitors treatment was associated with reduced risks of heart failure (RR = 0.38, 95% CI, 0.18 to 0.80) while lower incidence of stroke was observed in patients with colchicine treatment (RR = 0.47, 95% CI, 0.28 to 0.77). MI events were less frequent in patients over 65 years of age (RR = 0.90, 95% CI, 0.83 to 0.98) or with follow-up duration over 1 year (RR = 0.90, 95% CI, 0.85 to 0.96) when comparing anti-inflammatory therapies with placebo. CONCLUSIONS Anti-inflammatory therapies, especially those targeting the central IL-6 signaling pathway, may serve as promising treating strategies to ameliorate the risk of MI. IL-1 inhibitor and colchicine were associated with decreased risks of heart failure and stroke, respectively. MI risk reduction by anti-inflammatory therapies seemed to be more prominent in older patients with long follow-up duration.
Collapse
Affiliation(s)
- Zonglin Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| | - Suiyuan Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xingyun Zhu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
88
|
Han J, Wu J, Liu H, Huang Y, Ju W, Xing Y, Zhang X, Yang J. Inhibition of pyroptosis and apoptosis by capsaicin protects against LPS-induced acute kidney injury through TRPV1/UCP2 axis in vitro. Open Life Sci 2023; 18:20220647. [PMID: 37528882 PMCID: PMC10389676 DOI: 10.1515/biol-2022-0647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/11/2023] [Accepted: 05/31/2023] [Indexed: 08/03/2023] Open
Abstract
Acute kidney injury is a fatal disease characterized by a rapid deterioration of kidney function. Capsaicin (trans-8-methyl-N-vanillyl-6-nonenamide) is a natural product extracted from Capsicum. The aim of this study was to explore the protective effect of capsaicin on inflammation, apoptosis, and mitochondrial dysfunction in an in vitro model of acute kidney injury. Lipopolysaccharide (LPS)-induced acute kidney injury model was established in HK-2 cells to investigate the protective effect of capsaicin. Cell viability was assessed using CCK-8 assay, and protein expression was detected using western blot and immunofluorescence assay. Intracellular reactive oxygen species (ROS) level and mitochondrial membrane potential were analyzed by flow cytometry. Cell apoptosis was detected by propidium iodide staining. The results showed that capsaicin ameliorated LPS-induced cytotoxicity in vitro and attenuated the release of interleukin (IL)-1β and IL-18. Intriguingly, genipin abolished the protective effect of capsaicin. Molecularly, capsaicin activated transient receptor potential cation channel subfamily V member 1 -mitochondrial uncoupling protein 2 axis and inhibited caspase-1-mediated pyroptosis. In addition, capsaicin alleviated LPS-induced ROS production and mitochondrial membrane potential disruption and inhibited apoptosis. These findings suggest that capsaicin shows a protective effect in in vitro acute kidney injury model.
Collapse
Affiliation(s)
- Jinrun Han
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei, China
| | - Jinhao Wu
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei, China
| | - Hong Liu
- The Intensive Care Unit, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Huang
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Ju
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yifei Xing
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Zhang
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Yang
- The Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
89
|
Song Y, Chung J. Zingerone-Induced Autophagy Suppresses IL-1β Production by Increasing the Intracellular Killing of Aggregatibacter actinomycetemcomitans in THP-1 Macrophages. Biomedicines 2023; 11:2130. [PMID: 37626627 PMCID: PMC10452316 DOI: 10.3390/biomedicines11082130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Periodontitis is caused by the inflammation of tooth-supporting tissue by pathogens such as Aggregatibacter actinomycetemcomitans. Interleukin-1β (IL-1β), a pro-inflammatory cytokine, triggers a series of inflammatory reactions and promotes bone resorption. The aim of this study was to examine the molecular mechanism and anti-inflammatory function of zingerone, a dietary phenolic found in Zingiber officinale, on periodontal inflammation induced by A. actinomycetemcomitans. Zingerone attenuated A. actinomycetemcomitans-induced nitric oxide (NO) production by inhibiting the expression of inducible nitric oxide synthase (iNOS) in THP-1 macrophages. Zingerone also inhibited the expression of tumor necrosis factor (TNF)-α, IL-1β, and their signal pathway molecules including the toll-like receptor (TLR)/mitogen-activated protein kinase (MAPKase). In particular, zingerone suppressed the expression of absent in melanoma 2 (AIM2) inflammasome components on IL-1β production. Moreover, zingerone enhanced autophagosome formation and the expressions of autophagy-associated molecules. Interestingly, zingerone reduced the intracellular survival of A. actinomycetemcomitans. This was blocked by an autophagy inhibitor, which reversed the decrease in IL-1β production by zingerone. Finally, zingerone alleviated alveolar bone absorption in an A. actnomycetemcomitans-induced periodontitis mice model. Our data suggested that zingerone has potential use as a treatment for periodontal inflammation induced by A. actinomycetemcomitans.
Collapse
Affiliation(s)
- Yuri Song
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan-si 50612, Republic of Korea;
- Oral Genomics Research Center, Pusan National University, Yangsan-si 50612, Republic of Korea
| | - Jin Chung
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan-si 50612, Republic of Korea;
- Oral Genomics Research Center, Pusan National University, Yangsan-si 50612, Republic of Korea
| |
Collapse
|
90
|
Nagar A, Bharadwaj R, Shaikh MOF, Roy A. What are NLRP3-ASC specks? an experimental progress of 22 years of inflammasome research. Front Immunol 2023; 14:1188864. [PMID: 37564644 PMCID: PMC10411722 DOI: 10.3389/fimmu.2023.1188864] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
Speck assembly is the hallmark of NLRP3 inflammasome activation. The 1µm structure comprising of NLRP3 and ASC is the first observable phenotype of NLRP3 activation. While the common consensus is that the specks are the site of inflammasome activity, no direct experimental evidence exists to support this notion. In these 22 years, since the inflammasome discovery, several research studies have been published which directly or indirectly support or refute the idea of speck being the inflammasome. This review compiles the data from two decades of research to answer a long-standing question: "What are NLRP3-ASC specks?"
Collapse
Affiliation(s)
- Abhinit Nagar
- Department of Flow Cytometry, Cytek Biosciences, Fremont, CA, United States
| | - Ravi Bharadwaj
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, United States
| | - Mohammad Omar Faruk Shaikh
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia, Harvard Medical School, Boston, MA, United States
| | - Abhishek Roy
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
91
|
Tozetto-Mendoza TR, Mendes-Correa MC, Linhares IM, de Cássia Raymundi V, de Oliveira Paião HG, Barbosa EMG, Luna-Muschi A, Honorato L, Correa GF, da Costa AC, Costa SF, Witkin SS. Association between development of severe COVID-19 and a polymorphism in the CIAS1 gene that codes for an inflammasome component. Sci Rep 2023; 13:11252. [PMID: 37438453 DOI: 10.1038/s41598-023-38095-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023] Open
Abstract
An elevated pro-inflammatory cytokine response is associated with severe life-threatening symptoms in individuals with Coronavirus Disease-2019 (COVID). The inflammasome is an intracellular structure responsible for generation of interleukin (IL)-1β and IL-18. NALP3, a product of the CIAS1 gene, is the rate-limiting component for inflammasome activity. We evaluated if a CIAS1 42 base pair length polymorphism (rs74163773) was associated with severe COVID. DNA from 93 individuals with severe COVID, 38 with mild COVID, and 98 controls were analyzed for this polymorphism. The 12 unit repeat allele is associated with the highest inflammasome activity. Five alleles, corresponding to 6, 7, 9, 12 or 13 repeat units, divided into 12 genotypes were identified. The frequency of the 12 unit repeat allele was 45.3% in those with severe disease as opposed to 30.0% in those with mild disease and 26.0% in controls (p < 0.0001, severe vs. controls). In contrast, the 7 unit repeat allele frequency was 30.1% in controls as opposed to 14.0% and 12.5% in those with severe or mild disease, respectively (p ≤ 0.0017). We conclude that individuals positive for the CIAS1 12 allele may be at elevated risk for development of severe COVID due to an increased level of induced pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Tania R Tozetto-Mendoza
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Maria Cassia Mendes-Correa
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Iara Moreno Linhares
- Departamento de Ginecologia e Obstetricia, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa de Cássia Raymundi
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Heuder Gustavo de Oliveira Paião
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Erick Matheus Garcia Barbosa
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alessandra Luna-Muschi
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Layla Honorato
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Giovanna Francisco Correa
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Antonio Charlys da Costa
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Figueiredo Costa
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Steven S Witkin
- Laboratório de Virologia LIM 52, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
92
|
Salami A, Bettadapura S, Wang S. Gasdermin D kills bacteria. Microbiol Res 2023; 272:127383. [PMID: 37062105 PMCID: PMC10192060 DOI: 10.1016/j.micres.2023.127383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/07/2023] [Accepted: 04/08/2023] [Indexed: 04/18/2023]
Abstract
The recognition of pathogen- or damage- associated molecular patterns (PAMPs/DAMPs) signals a series of coordinated responses as part of innate immunity or host cell defense during infection. The inflammasome is an assemblage of multiprotein complexes in the cytosol that activate inflammatory caspases and release pro-inflammatory mediators. This review examines the two-edged sword activity of gasdermin D (GSDMD). Since its discovery in 2015, GSDMD has played a crucial role in the programmed necrotic type of cell death called pyroptosis. Pyroptosis is an important response in host self-protection against danger signals and infection. Although excessive pyroptosis has a deleterious effect on the host, it proves to have a game-changing therapeutic application against pathogenic invasion when controlled. Here, we explore the mechanism utilized by GSDMD, the best studied member of the gasdermin protein family, in host immune defense against many bacteria. While the protein contributes to the clearance of some bacteria, we also discussed results from previous studies and research, that its presence might hinder effective immunity against other pathogens, thus aiding pathogenic invasion and spread.
Collapse
Affiliation(s)
- Abosede Salami
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States
| | - Sahana Bettadapura
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, United States
| | - Shanzhi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States.
| |
Collapse
|
93
|
Loevenich S, Montaldo NP, Wickenhagen A, Sherstova T, van Loon B, Boyartchuk V, Anthonsen MW. Human metapneumovirus driven IFN-β production antagonizes macrophage transcriptional induction of IL1-β in response to bacterial pathogens. Front Immunol 2023; 14:1173605. [PMID: 37435074 PMCID: PMC10330783 DOI: 10.3389/fimmu.2023.1173605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1β transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-β. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1β transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-β-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1β mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1β. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.
Collapse
Affiliation(s)
- Simon Loevenich
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Nicola P. Montaldo
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Arthur Wickenhagen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Tatyana Sherstova
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Barbara van Loon
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Victor Boyartchuk
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Surgery, St Olav Hospital HF, Trondheim, Norway
| | - Marit W. Anthonsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
94
|
Ramon-Luing LA, Palacios Y, Ruiz A, Téllez-Navarrete NA, Chavez-Galan L. Virulence Factors of Mycobacterium tuberculosis as Modulators of Cell Death Mechanisms. Pathogens 2023; 12:839. [PMID: 37375529 PMCID: PMC10304248 DOI: 10.3390/pathogens12060839] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/29/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) modulates diverse cell death pathways to escape the host immune responses and favor its dissemination, a complex process of interest in pathogenesis-related studies. The main virulence factors of Mtb that alter cell death pathways are classified according to their origin as either non-protein (for instance, lipomannan) or protein (such as the PE family and ESX secretion system). The 38 kDa lipoprotein, ESAT-6 (early antigen-secreted protein 6 kDa), and another secreted protein, tuberculosis necrotizing toxin (TNT), induces necroptosis, thereby allowing mycobacteria to survive inside the cell. The inhibition of pyroptosis by blocking inflammasome activation by Zmp1 and PknF is another pathway that aids the intracellular replication of Mtb. Autophagy inhibition is another mechanism that allows Mtb to escape the immune response. The enhanced intracellular survival (Eis) protein, other proteins, such as ESX-1, SecA2, SapM, PE6, and certain microRNAs, also facilitate Mtb host immune escape process. In summary, Mtb affects the microenvironment of cell death to avoid an effective immune response and facilitate its spread. A thorough study of these pathways would help identify therapeutic targets to prevent the survival of mycobacteria in the host.
Collapse
Affiliation(s)
- Lucero A. Ramon-Luing
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico; (L.A.R.-L.); (A.R.)
| | - Yadira Palacios
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Mexico City 11200, Mexico;
- Department of Biological Systems, Universidad Autónoma Metropolitana, Campus Xochimilco, Mexico City 04960, Mexico
| | - Andy Ruiz
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico; (L.A.R.-L.); (A.R.)
| | - Norma A. Téllez-Navarrete
- Department of Healthcare Coordination, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico; (L.A.R.-L.); (A.R.)
| |
Collapse
|
95
|
Zhong Y, Wang S, Yin Y, Yu J, Liu Y, Gao H. Dexmedetomidine suppresses hippocampal astrocyte pyroptosis in cerebral hypoxic-ischemic neonatal rats by upregulating microRNA-148a-3p to inactivate the STAT/JMJD3 axis. Int Immunopharmacol 2023; 121:110440. [PMID: 37327511 DOI: 10.1016/j.intimp.2023.110440] [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: 04/14/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Dexmedetomidine (DEX), a selective α2-adrenoceptor agonist, is an anesthetic and sedative agent and has been reported to confer neuroprotective effects after cerebral hypoxic ischemia (CHI). This study was undertaken to elucidate the mechanisms by which microRNA (miR)-148a-3p is involved in the neuroprotective effect of DEX on hypoxic-ischemic brain damage in neonatal rats. METHODS Neonatal rats were exposed to CHI conditions, a miR-148a-3p inhibitor, and DEX. Hippocampal astrocytes were isolated to construct an oxygen-glucose deprivation (OGD) model. qRT-PCR and western blot were utilized to inspect miR-148a-3p, STAT1, STAT3, JMJD3, cleaved-Caspase-1, ASC, NLRP3, GSDMD, and GSDMD-N expression in rats and astrocytes. TUNEL staining was employed to measure astrocyte apoptosis rate, immunofluorescence to inspect cleaved-Caspase-1 and ASC levels, and ELISA to determine IL-1β and IL-18 expression. The target genes of miR-148a-3p were predicted using online software and verified by a dual-luciferase reporter gene assay. RESULTS A prominent increase in astrocyte apoptosis rate and the expression of pyroptosis- and inflammation-related factors were found in rats with CHI and OGD-treated astrocytes. DEX suppressed astrocyte apoptosis rate and decreased expression of pyroptosis- and inflammation-related factors. Knockdown of miR-148a-3p facilitated astrocyte pyroptosis, indicating that DEX exerted its protective effect by upregulating miR-148a-3p. miR-148a-3p negatively mediated STAT to inactivate JMJD3. Overexpression of STAT1 and STAT3 facilitated pyroptosis in astrocytes, which was negated by the overexpression of miR-148a-3p. CONCLUSION DEX inhibited hippocampal astrocyte pyroptosis by upregulating miR-148a-3p to inactivate the STAT/JMJD3 axis, thereby alleviating cerebral damage in neonatal rats with CHI.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Shengzhao Wang
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yongqiang Yin
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Jialu Yu
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yang Liu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Hong Gao
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou 558000, PR China.
| |
Collapse
|
96
|
Niu J, Meng G. Roles and Mechanisms of NLRP3 in Influenza Viral Infection. Viruses 2023; 15:1339. [PMID: 37376638 DOI: 10.3390/v15061339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Pathogenic viral infection represents a major challenge to human health. Due to the vast mucosal surface of respiratory tract exposed to the environment, host defense against influenza viruses has perpetually been a considerable challenge. Inflammasomes serve as vital components of the host innate immune system and play a crucial role in responding to viral infections. To cope with influenza viral infection, the host employs inflammasomes and symbiotic microbiota to confer effective protection at the mucosal surface in the lungs. This review article aims to summarize the current findings on the function of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) in host response to influenza viral infection involving various mechanisms including the gut-lung crosstalk.
Collapse
Affiliation(s)
- Junling Niu
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| |
Collapse
|
97
|
Sidor K, Jeznach A, Hoser G, Skirecki T. 1-Methylnicotinamide (1-MNA) inhibits the activation of the NLRP3 inflammasome in human macrophages. Int Immunopharmacol 2023; 121:110445. [PMID: 37290319 DOI: 10.1016/j.intimp.2023.110445] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
The NLRP3 inflammasome is among the most potent intracellular sensors of danger and disturbances of cellular homeostasis that can lead to the release of IL-1β and cell death, or pyroptosis. Despite its protective role, this mechanism is involved in the pathogenesis of numerous inflammatory diseases; therefore, it is seen as a potential therapeutic target. 1-methylnicotinamide (1-MNA) is a direct metabolite of nicotinamide and was previously shown to display several immunomodulatory properties, including a reduction in the reactive oxygen species (ROS). Here, we investigated whether 1-MNA could influence the activation of the NLRP3 inflammasome in human macrophages. In differentiated human macrophages we observed that 1-MNA specifically reduced the activation of the NLRP3 inflammasome. This effect was related to the scavenging of ROS, as exogenous H2O2 was able to restore NLRP3 activation. Additionally, 1-MNA increased the mitochondrial membrane potential, indicating that it did not inhibit oxidative phosphorylation. Moreover, at high but not low concentrations, 1-MNA decreased NF-κB activation and the level of pro-IL-1β. Interestingly, 1-MNA did not reduce the secretion of IL-6 upon endotoxin stimulation, confirming that its primary immunomodulatory effect on human macrophages is dependent on the NLRP3 inflammasome. Taken together, we have shown for the first time that 1-MNA reduced the activation of the NLRP3 inflammasome in human macrophages via an ROS-dependent pathway. Our results indicate a novel potential use of 1-MNA in NLRP3-related disorders.
Collapse
Affiliation(s)
- Karolina Sidor
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Aldona Jeznach
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
98
|
Kowalski S, Karska J, Łapińska Z, Hetnał B, Saczko J, Kulbacka J. An overview of programmed cell death: Apoptosis and pyroptosis-Mechanisms, differences, and significance in organism physiology and pathophysiology. J Cell Biochem 2023. [PMID: 37269535 DOI: 10.1002/jcb.30413] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 06/05/2023]
Abstract
Regulated cell death is an essential and heterogeneous process occurring in the life cycle of organisms, from embryonic development and aging to the regulation of homeostasis and organ maintenance. Under this term, we can distinguish many distinct pathways, including apoptosis and pyroptosis. Recently, there has been an increasing comprehension of the mechanisms governing these phenomena and their characteristic features. The coexistence of different types of cell death and the differences and similarities between them has been the subject of many studies. This review aims to present the latest literature in the field of pyroptosis and apoptosis and compare their molecular pathway's elements and significance in the physiology and pathophysiology of the organism.
Collapse
Affiliation(s)
- Szymon Kowalski
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Julia Karska
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Zofia Łapińska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Hetnał
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
99
|
Huang D, Guo Y, Li X, Pan M, Liu J, Zhang W, Mai K. Vitamin D 3/VDR inhibits inflammation through NF-κB pathway accompanied by resisting apoptosis and inducing autophagy in abalone Haliotis discus hannai. Cell Biol Toxicol 2023; 39:885-906. [PMID: 34637036 DOI: 10.1007/s10565-021-09647-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023]
Abstract
Vitamin D3 is believed to be a contributing factor to innate immunity. Vitamin D receptor (VDR) has a positive effect on inhibiting nuclear factor κB (NF-κB)-mediated inflammation. The underlying molecular mechanisms remain unclear, particularly in mollusks. Consequently, this study will investigate the process of vitamin D3/VDR regulating NF-κB pathway and further explore their functions on inflammation, autophagy, and apoptosis in abalone Haliotis discus hannai. Results showed that knockdown of VDR by using siRNA and dsRNA of VDR in vitro and in vivo led to more intense response of NF-κB signaling to lipopolysaccharide and higher level of apoptosis and autophagy. In addition, 1,25(OH)2D3 stimulation after VDR silencing could partially alleviate apoptosis and induce autophagy. Overexpression of VDR restricted the K48-polyubiquitin chain-dependent inhibitor of κB (IκB) ubiquitination and apoptosis-associated speck-like protein containing CARD (ASC) oligomerization. Besides, VDR silencing resulted in increase of ASC speck formation. In further mechanistic studies, we showed that VDR can directly bind to IκB and IKK1 in vitro and in vivo. In the feeding trial, H&E staining, TUNEL, and electron microscope results showed that vitamin D3 deficiency (0 IU/kg) could recruit more basophilic cells and increase more TUNEL-positive apoptotic cells and lipid droplets (LDs) than vitamin D3 supplement (1000 IU/kg and 5000 IU/kg). In summary, abalone VDR plays a negative regulator role in NF-κB-mediated inflammation via interacting with IκB and inhibiting ubiquitin-dependent degradation of IκB. Vitamin D3 in combination with VDR is essential to establish a delicate balance between autophagy and apoptosis in response to inflammation.
Collapse
Affiliation(s)
- Dong Huang
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Yanlin Guo
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Xinxin Li
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Mingzhu Pan
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Jiahuan Liu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China.
| | - Kangsen Mai
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
100
|
Ding C, Yang X, Li S, Zhang E, Fan X, Huang L, He Z, Sun J, Ma J, Zang L, Zheng M. Exploring the role of pyroptosis in shaping the tumor microenvironment of colorectal cancer by bulk and single-cell RNA sequencing. Cancer Cell Int 2023; 23:95. [PMID: 37198617 DOI: 10.1186/s12935-023-02897-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/12/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Emerging studies have shown that pyroptosis plays a non-negligible role in the development and treatment of tumors. However, the mechanism of pyroptosis in colorectal cancer (CRC) remains still unclear. Therefore, this study investigated the role of pyroptosis in CRC. METHODS A pyroptosis-related risk model was developed using univariate Cox regression and LASSO Cox regression analyses. Based on this model, pyroptosis-related risk scores (PRS) of CRC samples with OS time > 0 from Gene Expression Omnibus (GEO) database and The Cancer Genome Atlas (TCGA) database were calculated. The abundance of immune cells in CRC tumor microenvironment (TME) was predicted by single-sample gene-set enrichment analysis (ssGSEA). Then, the responses to chemotherapy and immunotherapy were predicted by pRRophetic algorithm, the tumor immune dysfunction and exclusion (TIDE) and SubMap algorithms, respectively. Moreover, the Cancer Therapeutics Response Portal (CTRP) and PRISM Repurposing dataset (PRISM) were used to explore novel drug treatment strategies of CRC. Finally, we investigated pyroptosis-related genes in the level of single-cell and validated the expression levels of these genes between normal and CRC cell lines by RT-qPCR. RESULTS Survival analysis showed that CRC samples with low PRS had better overall survival (OS) and progression-free survival (PFS). CRC samples with low PRS had higher immune-related gene expression and immune cell infiltration than those with high PRS. Besides, CRC samples with low PRS were more likely to benefit from 5-fluorouracil based chemotherapy and anti-PD-1 immunotherapy. In novel drug prediction, some compounds such as C6-ceramide and noretynodrel, were inferred as potential drugs for CRC with different PRS. Single-cell analysis revealed pyroptosis-related genes were highly expressed in tumor cells. RT-qPCR also demonstrated different expression levels of these genes between normal and CRC cell lines. CONCLUSIONS Taken together, this study provides a comprehensive investigation of the role of pyroptosis in CRC at the bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) levels, advances our understanding of CRC characteristics, and guides more effective treatment regimens.
Collapse
Affiliation(s)
- Chengsheng Ding
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Enkui Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xiaodong Fan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ling Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zirui He
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Junjun Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Lu Zang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|