1
|
Datta S, Rahman MA, Koka S, Boini KM. Mitigation of nicotine-induced podocyte injury through inhibition of thioredoxin interacting protein. Biomed Pharmacother 2025; 187:118110. [PMID: 40311224 DOI: 10.1016/j.biopha.2025.118110] [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: 01/20/2025] [Revised: 03/20/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Nicotine has been reported to initiate NLRP3 inflammasome formation and activation in different pathological conditions. The current study assessed whether thioredoxin-interacting protein (TXNIP) mediates nicotine-induced NLRP3 inflammasome activation and consequent podocyte injury. Co-immunoprecipitation analysis demonstrated that nicotine-induced TXNIP/NLRP3 interaction in podocytes relative to control groups. However, pre-treatment with TXNIP inhibitors, verapamil (Vera) or SRI-37330 (SRI) attenuates nicotine-induced TXNIP/NLRP3 interaction. Confocal microscopic analysis showed that nicotine treatment significantly increased the colocalization of Nlrp3 with Asc, Nlrp3 with caspase-1 and Nlrp3 with TXNIP in podocytes compared to control cells. Pretreatment with TXNIP inhibitor Vera or SRI abolished nicotine-induced Nlrp3/Asc, Nlrp3/caspase-1 or Nlrp3/TXNIP colocalization. Correspondingly, nicotine treatment significantly increased the caspase-1 activity and IL-1β production compared to control cells. However, prior treatment with TXNIP inhibiting Vera or SRI significantly attenuated the nicotine-induced caspase-1 activity and IL-1β production. Further immunofluorescence analysis showed that nicotine treatment significantly decreased podocin and nephrin expression compared to control cells. However, pretreatment with TXNIP inhibiting Vera or SRI attenuated the nicotine-induced podocin and nephrin reduction. In addition, confocal, flow cytometry and biochemical analysis showed that nicotine treatment significantly increased desmin expression, apoptosis and cell permeability compared to control cells. However, prior treatment with TXNIP inhibiting Vera or SRI significantly attenuated the nicotine-induced desmin expression, apoptosis and cell permeability. Taken together, our results demonstrate that TXNIP/NLRP3 interaction constitutes a potentially key signalling mechanism driving nicotine-induced NLRP3 inflammasome formation, activation and subsequent podocyte damage.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Mohammad Atiqur Rahman
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX, United States
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.
| |
Collapse
|
2
|
Shang DF, Xu WQ, Zhao Q, Zhao CL, Wang SY, Han YL, Li HG, Liu MH, Zhao WX. Molecular mechanisms of pyroptosis in non-alcoholic steatohepatitis and feasible diagnosis and treatment strategies. Pharmacol Res 2025; 216:107754. [PMID: 40306603 DOI: 10.1016/j.phrs.2025.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a distinct form of cell death that plays a critical role in intensifying inflammatory responses. It primarily occurs via the classical pathway, non-classical pathway, caspase-3/6/7/8/9-mediated pathways, and granzyme-mediated pathways. Key effector proteins involved in the pyroptosis process include gasdermin family proteins and pannexin-1 protein. Pyroptosis is intricately linked to the onset and progression of non-alcoholic steatohepatitis (NASH). During the development of NASH, factors such as pyroptosis, innate immunity, lipotoxicity, endoplasmic reticulum stress, and gut microbiota imbalance interact and interweave, collectively driving disease progression. This review analyzes the molecular mechanisms of pyroptosis and its role in the pathogenesis of NASH. Furthermore, it explores potential diagnostic and therapeutic strategies targeting pyroptosis, offering new avenues for improving the diagnosis and treatment of NASH.
Collapse
Affiliation(s)
- Dong-Fang Shang
- Henan University of CM, Zhengzhou 450000, China; The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Wen-Qian Xu
- Henan University of CM, Zhengzhou 450000, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Chen-Lu Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Si-Ying Wang
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Yong-Li Han
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - He-Guo Li
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Ming-Hao Liu
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Wen-Xia Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| |
Collapse
|
3
|
Gu F, Wang Z, Ding H, Tao X, Zhang J, Dai K, Li X, Shen H, Li H, Chen Z, Wang Z. Microglial mitochondrial DNA release contributes to neuroinflammation after intracerebral hemorrhage through activating AIM2 inflammasome. Exp Neurol 2024; 382:114950. [PMID: 39278588 DOI: 10.1016/j.expneurol.2024.114950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
Intracerebral hemorrhage (ICH) is a severe disease that often leads to disability and death. Neuroinflammatory response is a key causative factor of early secondary brain injury after ICH. AIM2 is a DNA-sensing protein that recognizes cytosolic double-stranded DNA and take a significant part in neuroinflammation. Mitochondrial DNA participates in the translation of proteins such as the respiratory chain in the mitochondria. Whether mtDNA is involved in forming AIM2 inflammasome after ICH remains unclear. We used mice to construct ICH model in vivo and we used BV2 microglial cells treated with oxyhemoglobin to simulate ICH in vitro. Following lentiviral transfection to overexpress AIM2 antagonist P202, a notable decrease was observed in the levels of AIM2 inflammasome-associated proteins, leading to a reduction in dead neurons surrounding the hematoma and an enhancement in long-term and short-term behavior of neurological deficits. We further explored whether mtDNA took part in the AIM2 activation after ICH. The cytosolic mtDNA level was down-regulated by the mitochondrial division protector Mdivi-1 and up-regulated by transfection of mtDNA into cytoplasm. We found the expression level of AIM2 inflammasome-related proteins and inflammatory cytokines release were regulated by the cytosolic mtDNA level. In conclusion, after ICH, the mtDNA content in the cytoplasm of microglia around the hematoma rises, causing AIM2 inflammation leading to neuronal apoptosis, which leads to neurological deficits in mice. On the other hand, P202 was able to block inflammatory vesicle activation and improve neurological function by preventing the interaction between AIM2 protein and mitochondrial DNA.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haojie Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xinyu Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Kun Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
4
|
Chen J, Hao Z, Li H, Wang J, Chen T, Wang Y, Shi G, Wang J, Wang Z, Zhang Z, Li J. Osteoporotic osseointegration: therapeutic hallmarks and engineering strategies. Theranostics 2024; 14:3859-3899. [PMID: 38994021 PMCID: PMC11234277 DOI: 10.7150/thno.96516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Osteoporosis is a systemic skeletal disease caused by an imbalance between bone resorption and formation. Current treatments primarily involve systemic medication and hormone therapy. However, these systemic treatments lack directionality and are often ineffective for locally severe osteoporosis, with the potential for complex adverse reactions. Consequently, treatment strategies using bioactive materials or external interventions have emerged as the most promising approaches. This review proposes twelve microenvironmental treatment targets for osteoporosis-related pathological changes, including local accumulation of inflammatory factors and reactive oxygen species (ROS), imbalance of mitochondrial dynamics, insulin resistance, disruption of bone cell autophagy, imbalance of bone cell apoptosis, changes in neural secretions, aging of bone cells, increased local bone tissue vascular destruction, and decreased regeneration. Additionally, this review examines the current research status of effective or potential biophysical and biochemical stimuli based on these microenvironmental treatment targets and summarizes the advantages and optimal parameters of different bioengineering stimuli to support preclinical and clinical research on osteoporosis treatment and bone regeneration. Finally, the review addresses ongoing challenges and future research prospects.
Collapse
Affiliation(s)
- Jiayao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jianping Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zepu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zheyuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
5
|
Li N, Zhang L, Wang X, Zhou Y, Gong L. Exploring exercise-driven inhibition of pyroptosis: novel insights into treating diabetes mellitus and its complications. Front Endocrinol (Lausanne) 2023; 14:1230646. [PMID: 37859981 PMCID: PMC10582706 DOI: 10.3389/fendo.2023.1230646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Diabetes mellitus (DM) and its complications are important, worldwide public health issues, exerting detrimental effects on human health and diminishing both quality of life and lifespan. Pyroptosis, as a new form of programmed cell death, plays a critical role in DM and its complications. Exercise has been shown to be an effective treatment for improving insulin sensitivity or preventing DM. However, the molecular mechanisms underlying the effects of exercise on pyroptosis-related diseases remain elusive. In this review, we provided a comprehensive elucidation of the molecular mechanisms underlying pyroptosis and the potential mechanism of exercise in the treatment of DM and its complications through the modulation of anti-pyroptosis-associated inflammasome pathways. Based on the existing evidence, further investigation into the mechanisms by which exercise inhibits pyroptosis through the regulation of inflammasome pathways holds promising potential for expanding preventive and therapeutic strategies for DM and facilitating the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Nan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Liang Zhang
- School of Strength and Conditioning Training, Beijing Sport University, Beijing, China
| | - Xintang Wang
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lijing Gong
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| |
Collapse
|
6
|
Yeung ST, Ovando LJ, Russo AJ, Rathinam VA, Khanna KM. CD169+ macrophage intrinsic IL-10 production regulates immune homeostasis during sepsis. Cell Rep 2023; 42:112171. [PMID: 36867536 PMCID: PMC10123955 DOI: 10.1016/j.celrep.2023.112171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 09/23/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
Abstract
Macrophages facilitate critical functions in regulating pathogen clearance and immune homeostasis in tissues. The remarkable functional diversity exhibited by macrophage subsets is dependent on tissue environment and the nature of the pathological insult. Our current knowledge of the mechanisms that regulate the multifaceted counter-inflammatory responses mediated by macrophages remains incomplete. Here, we report that CD169+ macrophage subsets are necessary for protection under excessive inflammatory conditions. We show that in the absence of these macrophages, even under mild septic conditions, mice fail to survive and exhibit increased production of inflammatory cytokines. Mechanistically, CD169+ macrophages control inflammatory responses via interleukin-10 (IL-10), as CD169+ macrophage-specific deletion of IL-10 was lethal during septic conditions, and recombinant IL-10 treatment reduced lipopolysaccharide (LPS)-induced lethality in mice lacking CD169+ macrophages. Collectively, our findings show a pivotal homeostatic role for CD169+ macrophages and suggest they may serve as an important target for therapy under damaging inflammatory conditions.
Collapse
Affiliation(s)
- Stephen T Yeung
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Luis J Ovando
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, Farmington, CT 06032, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT 06032, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA; Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
7
|
Wei H, Cui D. Pyroptosis and Insulin Resistance in Metabolic Organs. Int J Mol Sci 2022; 23:11638. [PMID: 36232938 PMCID: PMC9570443 DOI: 10.3390/ijms231911638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Skeletal muscle serves as the optimal effective organ to balance glucose homeostasis, but insulin resistance (IR) in skeletal muscle breaks this balance by impeding glucose uptake and causes metabolic disorders. IR in skeletal muscle is caused by multiple factors, and it has been reported that systemic low-grade inflammation is related to skeletal muscle IR, though its molecular mechanisms need to be ulteriorly studied. Pyroptosis is a novel inflammatory-mediated type of cell death. It has recently been reported that pyroptosis is associated with a decline in insulin sensitivity in skeletal muscle. The appropriate occurrence of pyroptosis positively eliminates pathogenic factors, whereas its excessive activation may aggravate inflammatory responses and expedite disease progression. The relationship between pyroptosis and IR in skeletal muscle and its underlined mechanism need to be further illustrated. The role of pyroptosis during the process of IR alleviation induced by non-drug interventions, such as exercise, also needs to be clarified. In this paper, we review and describe the molecular mechanisms of pyroptosis and further comb the roles of its relevant key factors in skeletal muscle IR, aiming to propose a novel theoretical basis for the relationship between pyroptosis and muscle IR and provide new research targets for the improvement of IR-related diseases.
Collapse
Affiliation(s)
| | - Di Cui
- College of Physical Education, Hunan University, Changsha 410012, China
| |
Collapse
|
8
|
Innate Immune Sensing of Nucleic Acid in Endodontic Infection. Int Endod J 2022; 55:1335-1346. [DOI: 10.1111/iej.13831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/26/2022]
|
9
|
Gómez‐Apo E, Silva‐Pereyra J, Soto‐Abraham V, Mondragón‐Maya A, Sanchez‐Lopez J. Immunohistochemical analysis of caspase expression in the brains of individuals with obesity or overweight. Obes Sci Pract 2022; 9:137-144. [PMID: 37034564 PMCID: PMC10073824 DOI: 10.1002/osp4.632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/03/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
Mechanisms underlying the negative effects of obesity on the brain are still unknown. Obesity is associated with oxidative stress in the brain and neuroinflammation that promotes neurodegenerative diseases. Chronic low-grade neuroinflammation in obesity could be associated with lower volumes of gray matter and lower neuronal density. If neuroinflammation mediated by the expression of cytokines and chemokines leads to apoptosis, this can be assessed by examining caspase expression. The aim of this study was to compare the expression of caspases in the 16 brains of donors with obesity/overweight (n = 8; Body Mass Index [BMI] = 31.6 ± 4.35 kg/m2; 2 females; Age = 52.9 ± 4.76 years) and normal weight (n = 8; BMI = 21.8 ± 1.5 kg/m2; 3 females; Age = 37.8 ± 19.2 years). Sixteen human brain samples were processed. Serial paraffin sections were examined by anti-caspase immunochemistry (caspase-3, caspase-4, caspase-6, caspase-1, caspase-8, and caspase-9 antibodies). Postmortem samples of cerebral cortex tissue were captured as photomicrographs and the images obtained were analyzed using ImageJ software to obtain the percentage of positive caspase expression. Nonparametric Mann-Whitney U tests were performed to compare caspase expression between samples from donors with obesity/overweight and normal weight. Taking into consideration the immunohistochemistry results, the Search Tool for the Retrieval of Interacting Genes was used to model molecular interactions. Results showed that brain samples from individuals with obesity/overweight exhibited significantly greater values of positive expression for Caspase-1 (U = 16.5, p = 0.05, Cohen d = 0.89) and -8 (U = 15, p = 0.03, Cohen d = 0.99) than those from donors with normal weight. This study contributes to the knowledge about the inflammatory effects of obesity/overweight on brain, suggesting the activation of the alternative inflammasome pathway in which interact caspase-1 and -8.
Collapse
Affiliation(s)
- Erick Gómez‐Apo
- Servicio de Anatomía Patológica Hospital General de México “Dr. Eduardo Liceaga” Ciudad de México México
- Programa de Maestría y Doctorado en Ciencias Médicas y Odontológicas y de la Salud Universidad Nacional Autónoma de México Ciudad de México México
| | - Juan Silva‐Pereyra
- Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México Tlalnepantla Estado de México México
| | - Virgilia Soto‐Abraham
- Servicio de Anatomía Patológica Hospital General de México “Dr. Eduardo Liceaga” Ciudad de México México
| | - Alejandra Mondragón‐Maya
- Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México Tlalnepantla Estado de México México
| | - Javier Sanchez‐Lopez
- Escuela Nacional de Estudios Superiores Unidad Juriquilla Universidad Nacional Autónoma de México Querétaro Querétaro México
| |
Collapse
|
10
|
Shidid S, Bluth MH, Smith-Norowitz TA. The Role of Inflammasomes in Mediating Urological Disease: A Short Literature Review. J Inflamm Res 2022; 15:4359-4365. [PMID: 35937918 PMCID: PMC9354909 DOI: 10.2147/jir.s370451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/23/2022] [Indexed: 11/23/2022] Open
Abstract
Inflammasome dysfunction may be responsible for underlying inflammatory diseases, which include renal and urological pathologies. Five inflammasomes have been described, including nucleotide-binding domain leucine-rich repeat (NLR), NL pyrin domain containing receptor 1(NLPR1), NLRP3, NLR and caspase recruitment domain containing receptor 4 (NLRC4), and the AIM2-like receptor. The purpose of this study was to review literature sources regarding how innate immunity and inflammasomes contribute to urologic disease and infection. A literature search of PubMed/MEDLINE, EMBASE and Google Scholar articles. Articles were selected for review if their content included (1) inflammasomes and (2) urology in the adult population. The initiation of specific cytokine cascades, which include IL-1β and IL-18, appear responsible for a repertoire of urologic pathologies. Inflammation mediates a wide range of uropathies (urologic disorders and infections) which are found in the bladder, prostate, or kidney and inflammasomes appear to be particularly responsible for urological and renal pathologies. Understanding the role of inflammasomes in urologic disorders can help improve treatment and overall quality of life in patients with these disorders.
Collapse
Affiliation(s)
- Sarah Shidid
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, New York, NY, 11203, USA
- Correspondence: Sarah Shidid, Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, Brooklyn, New York, NY, 11203, USA, Tel +1718 270-1295, Fax +1718 270-3289, Email
| | - Martin H Bluth
- Department of Pathology, Maimonides Medical Center, New York, NY, 11219, USA
| | - Tamar A Smith-Norowitz
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, New York, NY, 11203, USA
| |
Collapse
|
11
|
Anton-Pampols P, Diaz-Requena C, Martinez-Valenzuela L, Gomez-Preciado F, Fulladosa X, Vidal-Alabro A, Torras J, Lloberas N, Draibe J. The Role of Inflammasomes in Glomerulonephritis. Int J Mol Sci 2022; 23:ijms23084208. [PMID: 35457026 PMCID: PMC9029880 DOI: 10.3390/ijms23084208] [Citation(s) in RCA: 15] [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: 02/28/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 02/07/2023] Open
Abstract
The inflammasome is an immune multiprotein complex that activates pro-caspase 1 in response to inflammation-inducing stimuli and it leads to IL-1β and IL-18 proinflammatory cytokine production. NLRP1 and NLRP3 inflammasomes are the best characterized and they have been related to several autoimmune diseases. It is well known that the kidney expresses inflammasome genes, which can influence the development of some glomerulonephritis, such as lupus nephritis, ANCA glomerulonephritis, IgA nephropathy and anti-GBM nephropathy. Polymorphisms of these genes have also been described to play a role in autoimmune and kidney diseases. In this review, we describe the main characteristics, activation mechanisms, regulation and functions of the different inflammasomes. Moreover, we discuss the latest findings about the role of the inflammasome in several glomerulonephritis from three different points of view: in vitro, animal and human studies.
Collapse
Affiliation(s)
- Paula Anton-Pampols
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
| | - Clara Diaz-Requena
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
| | - Laura Martinez-Valenzuela
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
| | - Francisco Gomez-Preciado
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
| | - Xavier Fulladosa
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
- Clinical Sciences Department, Campus de Bellvitge, Barcelona University, Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Anna Vidal-Alabro
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
| | - Joan Torras
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
- Clinical Sciences Department, Campus de Bellvitge, Barcelona University, Hospitalet de Llobregat, 08907 Barcelona, Spain
- Correspondence:
| | - Núria Lloberas
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
- Department of Physiological Sciences, Campus de Bellvitge, Barcelona University, Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Juliana Draibe
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain; (P.A.-P.); (L.M.-V.); (F.G.-P.); (X.F.); (J.D.)
- IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain; (C.D.-R.); (A.V.-A.); (N.L.)
| |
Collapse
|
12
|
Mechanisms and Consequences of Noncanonical Inflammasome-Mediated Pyroptosis. J Mol Biol 2022; 434:167245. [PMID: 34537239 PMCID: PMC8844060 DOI: 10.1016/j.jmb.2021.167245] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
The noncanonical inflammasome, comprising inflammatory caspases 4, 5, or 11, monitors the cytosol for bacterial lipopolysaccharide (LPS). Intracellular LPS-elicited autoproteolysis of these inflammatory caspases leads to the cleavage of the pore-forming protein gasdermin D (GSDMD). GSDMD pore formation induces a lytic form of cell death known as pyroptosis and the release of inflammatory cytokines and DAMPs, thereby promoting inflammation. The noncanonical inflammasome-dependent innate sensing of cytosolic LPS plays important roles in bacterial infections and sepsis pathogenesis. Exciting studies in the recent past have significantly furthered our understanding of the biochemical and structural basis of the caspase-4/11 activation of GSDMD, caspase-4/11's substrate specificity, and the biological consequences of noncanonical inflammasome activation of GSDMD. This review will discuss these recent advances and highlight the remaining gaps in our understanding of the noncanonical inflammasome and pyroptosis.
Collapse
|
13
|
Epithelial GPR35 protects from Citrobacter rodentium infection by preserving goblet cells and mucosal barrier integrity. Mucosal Immunol 2022; 15:443-458. [PMID: 35264769 PMCID: PMC9038528 DOI: 10.1038/s41385-022-00494-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023]
Abstract
Goblet cells secrete mucin to create a protective mucus layer against invasive bacterial infection and are therefore essential for maintaining intestinal health. However, the molecular pathways that regulate goblet cell function remain largely unknown. Although GPR35 is highly expressed in colonic epithelial cells, its importance in promoting the epithelial barrier is unclear. In this study, we show that epithelial Gpr35 plays a critical role in goblet cell function. In mice, cell-type-specific deletion of Gpr35 in epithelial cells but not in macrophages results in goblet cell depletion and dysbiosis, rendering these animals more susceptible to Citrobacter rodentium infection. Mechanistically, scRNA-seq analysis indicates that signaling of epithelial Gpr35 is essential to maintain normal pyroptosis levels in goblet cells. Our work shows that the epithelial presence of Gpr35 is a critical element for the function of goblet cell-mediated symbiosis between host and microbiota.
Collapse
|
14
|
Ye Z, Liang L, Lu H, Shen Y, Zhou W, Li Y. Nanotechnology-Employed Bacteria-Based Delivery Strategy for Enhanced Anticancer Therapy. Int J Nanomedicine 2021; 16:8069-8086. [PMID: 34934313 PMCID: PMC8684392 DOI: 10.2147/ijn.s329855] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Bacteria and their derivatives (membrane vesicles, MVs) exhibit great advantages for targeting hypoxic tumor cores, strong penetration ability and activating immune responses, holding great potential as auspicious candidates for therapeutic and drug-delivery applications. However, the safety issues and low therapeutic efficiency by single administration still need to be solved. To further optimize their performance and to utilize their natural abilities, scientists have strived to modify bacteria with new moieties on their surface while preserving their advantages. The aim of this review is to give a comprehensive overview of a non-genetic engineering modification strategy that can be used to optimize the bacteria with nanomaterials and the design strategy that can be used to optimize MVs for better targeted therapy. Here, the advantages and disadvantages of these processes and their applicability for the development of bacteria-related delivery system as antitumor therapeutic agents are discussed. The prospect and the challenges of the above targeted delivery system are also proposed.
Collapse
Affiliation(s)
- Zixuan Ye
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Lizhen Liang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Huazhen Lu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Yan Shen
- State Key Laboratory of Natural Medicines, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Wenwu Zhou
- National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| |
Collapse
|
15
|
Differential involvement of the canonical and noncanonical inflammasomes in the immune response against infection by the periodontal bacteria Porphyromonas gingivalis and Fusobacterium nucleatum. CURRENT RESEARCH IN MICROBIAL SCIENCES 2021; 2:100023. [PMID: 34841314 PMCID: PMC8610350 DOI: 10.1016/j.crmicr.2021.100023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/30/2021] [Accepted: 02/16/2021] [Indexed: 12/30/2022] Open
Abstract
The canonical P2 × 7-Caspase-1 pathway is necessary for secretion of IL-1β in oral tissues and macrophages infected with P. gingivalis. P2 × 7 receptor controls bacterial load of F. nucleatum and P. gingivalis in macrophages and in mice. Caspase-11 is essential for F. nucleatum-induced secretion of IL-1β in macrophages, limits F. nucleatum infection in macrophages and in mice, and is required for cell death induced by F. nucleatum infection. The canonical inflammasome is activated preferentially in response to P. gingivalis infection, while the noncanonical inflammasome plays a predominant role during F. nucleatum infection.
We examined the involvement of the P2 × 7 receptor and the canonical and noncanonical inflammasomes in the control of single-species or dual-species infection by the periodontal bacteria Porphyromonas gingivalis and Fusobacterium nucleatum in cells and mice. Stimulation of the P2 × 7 receptor leads to activation of the canonical NLRP3 inflammasome and activation of caspase-1, which leads to cleavage of pro-IL-1β to IL-1β, a key cytokine in the host inflammatory response in periodontal disease. The non-canonical inflammasome pathway involves caspase-11. Thus, wildtype (WT), P2 × 7−/−, caspase-11−/− and caspase-1/11−/− mice were co-infected with both bacterial species. In parallel, bone marrow-derived macrophages (BMDMs) from WT mice and the different knockout mice were infected with P. gingivalis and/or F. nucleatum, and treated or not with extracellular ATP, which is recognized by P2 × 7. F. nucleatum infection alone promoted secretion of IL-1β in BMDMs. Conversely, the canonical pathway involving P2 × 7 and caspase-1 was necessary for secretion of IL-1β in BMDMs infected with P. gingivalis and in the mandible of mice coinfected with P. gingivalis and F. nucleatum. The P2 × 7 pathway can limit bacterial load in single-species and dual-species infection with P. gingivalis and F. nucleatum in BMDMs and in mice. The non-canonical pathway involving caspase-11 was required for secretion of IL-1β induced by F. nucleatum infection in BMDMs, without treatment with ATP. Caspase-11 was also required for induction of cell death during infection with F. nucleatum and contributed to limiting bacterial load during F. nucleatum infection in BMDMs and in the gingival tissue of mice coinfected with P. gingivalis and F. nucleatum. Together, these data suggest that the P2 × 7-caspase-1 and caspase-11 pathways are involved in the immune response against infection by P. gingivalis and F. nucleatum, respectively.
Collapse
|
16
|
Kasti AN, Synodinou KD, Pyrousis IA, Nikolaki MD, Triantafyllou KD. Probiotics Regulating Inflammation via NLRP3 Inflammasome Modulation: A Potential Therapeutic Approach for COVID-19. Microorganisms 2021; 9:2376. [PMID: 34835501 PMCID: PMC8624812 DOI: 10.3390/microorganisms9112376] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/21/2022] Open
Abstract
Inflammasomes are cytoplasmic multiprotein complexes formed by the host's immune system as a response to microbial infection and cellular damage. Many studies have revealed various regulators of NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation, while it has been recently shown that NLRP3 is implicated in COVID-19 pathogenesis. At the same time, probiotics counteract the inflammatory process and modulate cytokine release, thus influencing both innate and adaptive immune systems. Herein, we review the immunomodulatory potential of probiotics on the assembly of NLRP3 inflammasome, as well as the pathophysiological mechanisms supporting the use of probiotic bacteria for SARS-CoV-2 infection management, presenting evidence from preclinical studies of the last decade: in vivo, ex vivo, and mixed trials. Data show that probiotics intake is related to NLRP3 inflammasome attenuation and lower levels of inflammation markers, highlighting the beneficial effects of probiotics on inflammatory conditions. Currently, none of the ongoing clinical trials evaluating the effectiveness of probiotics intake in humans with COVID-19 has been completed. However, evidence from preclinical studies indicates that probiotics may block virus invasion and replication through their metabolites, bacteriocins, and their ability to block Angiotensin-Converting Enzyme 2 (ACE2), and by stimulating the immune response through NLRP3 inflammasome regulation. In this review, the beneficial effects of probiotics in the inflammatory process through NLRP3 inflammasome attenuation are presented. Furthermore, probiotics may target SARS-CoV-2 both by blocking virus invasion and replication and by stimulating the immune response through NLRP3 inflammasome regulation. Heterogeneity of the results-due to, among others, different bacterial strains and their metabolites, forms, dosage, and experimental designs-indicates the need for more extensive research.
Collapse
Affiliation(s)
- Arezina N. Kasti
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.N.K.); (K.D.S.); (I.A.P.); (M.D.N.)
| | - Kalliopi D. Synodinou
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.N.K.); (K.D.S.); (I.A.P.); (M.D.N.)
| | - Ioannis A. Pyrousis
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.N.K.); (K.D.S.); (I.A.P.); (M.D.N.)
- Medical School, University of Patras, 26504 Patras, Greece
| | - Maroulla D. Nikolaki
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.N.K.); (K.D.S.); (I.A.P.); (M.D.N.)
| | - Konstantinos D. Triantafyllou
- Hepatogastroenterology Unit, 2nd Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, 12462 Athens, Greece
| |
Collapse
|
17
|
Park JM, Han YM, Hahm KB. Rejuvenation of Helicobacter pylori-Associated Atrophic Gastritis Through Concerted Actions of Placenta-Derived Mesenchymal Stem Cells Prevented Gastric Cancer. Front Pharmacol 2021; 12:675443. [PMID: 34483897 PMCID: PMC8416416 DOI: 10.3389/fphar.2021.675443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/22/2021] [Indexed: 01/06/2023] Open
Abstract
Chronic Helicobacter pylori infection causes gastric cancer via the progression of precancerous chronic atrophic gastritis (CAG). Therefore, repairing gastric atrophy could be a useful strategy in preventing H. pylori-associated gastric carcinogenesis. Although eradication of the bacterial pathogen offers one solution to this association, this study was designed to evaluate an alternative approach using mesenchymal stem cells to treat CAG and prevent carcinogenesis. Here, we used human placenta-derived mesenchymal stem cells (PD-MSCs) and their conditioned medium (CM) to treat H. pylori-associated CAG in a mice/cell model to explore their therapeutic effects and elucidate their molecular mechanisms. We compared the changes in the fecal microbiomes in response to PD-MSC treatments, and chronic H. pylori-infected mice were given ten treatments with PD-MSCs before being sacrificed for end point assays at around 36 weeks of age. These animals presented with significant reductions in the mean body weights of the control group, which were eradicated following PD-MSC treatment (p < 0.01). Significant changes in various pathological parameters including inflammation, gastric atrophy, erosions/ulcers, and dysplastic changes were noted in the control group (p < 0.01), but these were all significantly reduced in the PD-MSC/CM-treated groups. Lgr5+, Ki-67, H+/K+-ATPase, and Musashi-1 expressions were all significantly increased in the treated animals, while inflammatory mediators, MMP, and apoptotic executors were significantly decreased in the PD-MSC group compared to the control group (p < 0.001). Our model showed that H. pylori-initiated, high-salt diet-promoted gastric atrophic gastritis resulted in significant changes in the fecal microbiome at the phylum/genus level and that PD-MSC/CM interventions facilitated a return to more normal microbial communities. In conclusion, administration of PD-MSCs or their conditioned medium may present a novel rejuvenating agent in preventing the progression of H. pylori-associated premalignant lesions.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daejeon, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Seoul, Korea.,CHA Cancer Preventive Research Center, CHA Bio Complex, Seongnam, Korea
| |
Collapse
|
18
|
Li Y, Ling J, Jiang Q. Inflammasomes in Alveolar Bone Loss. Front Immunol 2021; 12:691013. [PMID: 34177950 PMCID: PMC8221428 DOI: 10.3389/fimmu.2021.691013] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Bone remodeling is tightly controlled by osteoclast-mediated bone resorption and osteoblast-mediated bone formation. Fine tuning of the osteoclast-osteoblast balance results in strict synchronization of bone resorption and formation, which maintains structural integrity and bone tissue homeostasis; in contrast, dysregulated bone remodeling may cause pathological osteolysis, in which inflammation plays a vital role in promoting bone destruction. The alveolar bone presents high turnover rate, complex associations with the tooth and periodontium, and susceptibility to oral pathogenic insults and mechanical stress, which enhance its complexity in host defense and bone remodeling. Alveolar bone loss is also involved in systemic bone destruction and is affected by medication or systemic pathological factors. Therefore, it is essential to investigate the osteoimmunological mechanisms involved in the dysregulation of alveolar bone remodeling. The inflammasome is a supramolecular protein complex assembled in response to pattern recognition receptors and damage-associated molecular patterns, leading to the maturation and secretion of pro-inflammatory cytokines and activation of inflammatory responses. Pyroptosis downstream of inflammasome activation also facilitates the clearance of intracellular pathogens and irritants. However, inadequate or excessive activity of the inflammasome may allow for persistent infection and infection spreading or uncontrolled destruction of the alveolar bone, as commonly observed in periodontitis, periapical periodontitis, peri-implantitis, orthodontic tooth movement, medication-related osteonecrosis of the jaw, nonsterile or sterile osteomyelitis of the jaw, and osteoporosis. In this review, we present a framework for understanding the role and mechanism of canonical and noncanonical inflammasomes in the pathogenesis and development of etiologically diverse diseases associated with alveolar bone loss. Inappropriate inflammasome activation may drive alveolar osteolysis by regulating cellular players, including osteoclasts, osteoblasts, osteocytes, periodontal ligament cells, macrophages, monocytes, neutrophils, and adaptive immune cells, such as T helper 17 cells, causing increased osteoclast activity, decreased osteoblast activity, and enhanced periodontium inflammation by creating a pro-inflammatory milieu in a context- and cell type-dependent manner. We also discuss promising therapeutic strategies targeting inappropriate inflammasome activity in the treatment of alveolar bone loss. Novel strategies for inhibiting inflammasome signaling may facilitate the development of versatile drugs that carefully balance the beneficial contributions of inflammasomes to host defense.
Collapse
Affiliation(s)
- Yang Li
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Junqi Ling
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
19
|
Mamun AA, Wu Y, Nasrin F, Akter A, Taniya MA, Munir F, Jia C, Xiao J. Role of Pyroptosis in Diabetes and Its Therapeutic Implications. J Inflamm Res 2021; 14:2187-2206. [PMID: 34079327 PMCID: PMC8164340 DOI: 10.2147/jir.s291453] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Pyroptosis is mainly considered as a new pro-inflammatory mediated-programmed cell death. In addition, pyroptosis is described by gasdermin-induced pore formation on the membrane, cell swelling and rapid lysis, and several pro-inflammatory mediators interleukin-1β (IL-1β) and interleukin-18 (IL-18) release. Extensive studies have shown that pyroptosis is commonly involved by activating the caspase-1-dependent canonical pathway and caspase-4/5/11-dependent non-canonical pathway. However, pyroptosis facilitates local inflammation and inflammatory responses. Current researches have reported that pyroptosis promotes the progression of several diabetic complications. Emerging studies have suggested that some potential molecules targeting the pyroptosis and inflammasome signaling pathways could be a novel therapeutic avenue for managing and treating diabetes and its complications in the near future. Our narrative review concisely describes the possible mechanism of pyroptosis and its progressive understanding of the development of diabetic complications.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou, Zhejiang Province, 325035, People's Republic of China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang Province, 325035, People's Republic of China
| | - Fatema Nasrin
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia.,School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Afroza Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Masuma Afrin Taniya
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh, Dhaka, 1229, Bangladesh
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, People's Republic of China
| | - Jian Xiao
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou, Zhejiang Province, 325035, People's Republic of China
| |
Collapse
|
20
|
Ren H, Yang H, Yang X, Zhang G, Rong X, Huang J, Zhang L, Fu Y, Allain JP, Li C, Wang W. Brucella Outer Membrane Lipoproteins 19 and 16 Differentially Induce IL-18 Response or Pyroptosis in Human Monocytic Cells. J Infect Dis 2021; 224:2148-2159. [PMID: 34013337 DOI: 10.1093/infdis/jiab272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/14/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Brucella species (B. spp.) are Gram-negative intracellular bacteria, causing severe inflammatory diseases in animals and humans. Two major lipoproteins (L19) and (L16) of Brucella outer membrane proteins (OMPs) were extensively explored in associating with inflammatory response of human monocytes (THP-1). METHODS Activated THP-1 cells induced with recombinant L19 and L16 were analyzed in comparison with unlipidated forms (U19 and U16) and lipopolysaccharide (LPS) of B. melitensis, respectively. RESULTS Secretion of inflammatory factors TNF-α, IL-6 and IL-1β was significantly increased from L19, L16 or both stimulated THP-1 cells. High secretion of IL-18 was detected only from L19-induced cells. Signaling of those cytokine responses was identified mainly through P38-MAPK pathway, and signaling of L19-induced IL-1β response was partly occurred via NF-κB. Exploration for different forms of IL-18 found that L19-induced production of active IL-18 (18 kD) was through up-regulating NLRP3 and activating caspase-1, while L16-induced production of inactive IL-18 fragments (15 kD and 16 kD) occurred through activating caspase-8/3. Additionally, L19 up-regulated phosphorylation of XIAP for inhibiting caspase-3 activity to cleave IL-18, while L16 activated caspase-3 for producing GSDME-N and leading to pyroptosis of THP-1 cells. CONCLUSION Brucella L19 and L16 differentially induce IL-18 response or pyroptosis in THP-1 cells, respectively.
Collapse
Affiliation(s)
- Hui Ren
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Heng Yang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.,Department of blood Transfusion, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xin Yang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Guoxia Zhang
- Department of Infectious Disease, Hei Longjiang General Hospital of Agriculture reclamation Bureau, Harbin 150088, China
| | - Xia Rong
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jiaheng Huang
- Department of Surgery, First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yongshui Fu
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jean-Pierre Allain
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.,Emeritus professor of Transfusion Medicine, University of Cambridge, Cambridge CB2 2PT, Cambridge, UK
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Wenjing Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
21
|
Cheng K, Zhao R, Li Y, Qi Y, Wang Y, Zhang Y, Qin H, Qin Y, Chen L, Li C, Liang J, Li Y, Xu J, Han X, Anderson GJ, Shi J, Ren L, Zhao X, Nie G. Bioengineered bacteria-derived outer membrane vesicles as a versatile antigen display platform for tumor vaccination via Plug-and-Display technology. Nat Commun 2021; 12:2041. [PMID: 33824314 PMCID: PMC8024398 DOI: 10.1038/s41467-021-22308-8] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
An effective tumor vaccine vector that can rapidly display neoantigens is urgently needed. Outer membrane vesicles (OMVs) can strongly activate the innate immune system and are qualified as immunoadjuvants. Here, we describe a versatile OMV-based vaccine platform to elicit a specific anti-tumor immune response via specifically presenting antigens onto OMV surface. We first display tumor antigens on the OMVs surface by fusing with ClyA protein, and then simplify the antigen display process by employing a Plug-and-Display system comprising the tag/catcher protein pairs. OMVs decorated with different protein catchers can simultaneously display multiple, distinct tumor antigens to elicit a synergistic antitumour immune response. In addition, the bioengineered OMVs loaded with different tumor antigens can abrogate lung melanoma metastasis and inhibit subcutaneous colorectal cancer growth. The ability of the bioengineered OMV-based platform to rapidly and simultaneously display antigens may facilitate the development of these agents for personalized tumour vaccines.
Collapse
Affiliation(s)
- Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian, China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian, China
| | - Yingqiu Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yazhou Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yuting Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Long Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Chen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yujing Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
22
|
Kumari P, Russo AJ, Wright SS, Muthupalani S, Rathinam VA. Hierarchical cell-type-specific functions of caspase-11 in LPS shock and antibacterial host defense. Cell Rep 2021; 35:109012. [PMID: 33882312 PMCID: PMC8451177 DOI: 10.1016/j.celrep.2021.109012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/06/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022] Open
Abstract
Caspase-11 sensing of intracellular lipopolysaccharide (LPS) plays critical roles during infections and sepsis. However, the key cell types that sense intracellular LPS and their contributions to the host responses at the organismal level are not completely clear. Here, we show that macrophage/monocyte-specific caspase-11 plays a dominant role in mediating the pathological manifestations of endotoxemia, including gasdermin D (GSDMD) activation, interleukin (IL)-1β, IL-18, and damage-associated molecular pattern (DAMP) release, tissue damage, and death. Surprisingly, caspase-11 expression in CD11c+ cells and intestinal epithelial cells (IECs) plays minor detrimental roles in LPS shock. In contrast, caspase-11 expression in neutrophils is dispensable for LPS-induced lethality. Importantly, caspase-11 sensing of intracellular LPS in LyzM+ myeloid cells and MRP8+ neutrophils, but not CD11c+ cells and IECs, is necessary for bacterial clearance and host survival during intracellular bacterial infection. Thus, we reveal hierarchical cell-type-specific roles of caspase-11 that govern the host-protective and host-detrimental functions of the cytosolic LPS surveillance. Kumari et al. reveal hierarchical cell-type-specific roles of caspase-11 that govern the host-protective and host-detrimental functions of the cytosolic LPS surveillance pathway during bacterial infections and sepsis, respectively.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Skylar S Wright
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|
23
|
Ta A, Vanaja SK. Inflammasome activation and evasion by bacterial pathogens. Curr Opin Immunol 2021; 68:125-133. [PMID: 33338767 PMCID: PMC7925435 DOI: 10.1016/j.coi.2020.11.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/25/2020] [Accepted: 11/22/2020] [Indexed: 12/21/2022]
Abstract
Innate immune system plays an essential role in combating infectious diseases by recognizing invading pathogens and activating host defense response. Inflammasomes complexes are a central component of the cytosolic innate immune surveillance and are vital in host defense against bacterial pathogens. Bacterial products or pathogen-induced modifications in the intracellular environment are sensed by the inflammasome receptors that form complexes that serve as a platform for caspase-1-dependent or caspase-11-dependent induction of pyroptosis and secretion of cytokines, IL-1β and IL-18. However, several pathogenic bacteria have developed strategies to evade inflammasome activation. This review highlights the recent advances in the mechanism of inflammasome activation by bacterial pathogens and some of the bacterial evasion strategies of inflammasome activation.
Collapse
Affiliation(s)
- Atri Ta
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA.
| |
Collapse
|
24
|
Pyroptosis by caspase-11 inflammasome-Gasdermin D pathway in autoimmune diseases. Pharmacol Res 2021; 165:105408. [PMID: 33412278 DOI: 10.1016/j.phrs.2020.105408] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023]
Abstract
Inflammasomes are a group of supramolecular complexes primarily comprise a sensor, adaptor protein and an effector. Among them, canonical inflammasomes are assembled by one specific pattern recognition receptor, the adaptor protein apoptosis-associated speck-like protein containing a CARD and procaspase-1. Murine caspase-11 and its human ortholog caspase-4/5 are identified as cytosolic sensors which directly responds to LPS. Once gaining access to cytosol, LPS further trigger inflammasome activation in noncanonical way. Downstream pore-forming Gasdermin D is a pyroptosis executioner. Emerging evidence announced in recent years demonstrate the vital role played by caspase-11 non-canonical inflammasome in a range of autoimmune diseases. Pharmacological ablation of caspase-11 and its related effector results in potent therapeutic effects. Though recent advances have highlighted the potential of caspase-11 as a drug target, the understanding of caspase-11 molecular activation and regulation mechanism remains to be limited and thus hampered the discovery and progression of novel inhibitors. Here in this timeline review, we explored how caspase-11 get involved in the pathogenesis of autoimmune diseases, we also collected the reported small-molecular caspase-11 inhibitors. Moreover, the clinical implications and therapeutic potential of caspase-11 inhibitors are discussed. Targeting non-canonical inflammasomes is a promising strategy for autoimmune diseases treatment, while information about the toxicity and physiological disposition of the promising caspase-11 inhibitors need to be supplemented before they can be translated from bench to bedside.
Collapse
|
25
|
Ding PH, Yang MX, Wang NN, Jin LJ, Dong Y, Cai X, Chen LL. Porphyromonas gingivalis-Induced NLRP3 Inflammasome Activation and Its Downstream Interleukin-1β Release Depend on Caspase-4. Front Microbiol 2020; 11:1881. [PMID: 32903638 PMCID: PMC7438778 DOI: 10.3389/fmicb.2020.01881] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Oral commensals contribute to microbe-host symbiosis in periodontal homeostasis, and Porphyromonas gingivalis (P. gingivalis) as the keystone pathogen critically accounts for the shift of symbiosis to dysbiosis and periodontal destruction. Nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome-mediated interleukin-1β (IL-1β) is significantly involved in periodontal diseases, and notably P. gingivalis enables to modulate the induction and expression of NLRP3. Whereas, the exact mechanism by which NLRP3 inflammasome is regulated in response to commensal and pathogenic bacteria remains unclear. Methods: To examine the expression of IL-1β and NLRPs inflammasome in tissues with severe chronic periodontitis, and further investigate how Caspase-4-dependent non-canonical NLRP3 inflammasome pathways functioned during the interactions of Streptococcus mitis (S. mitis) and P. gingivalis with human THP-1 cells. Results: IL-1β and NLRP3, NLRP6, NLRP12, and absent in melanoma 2 (AIM2) inflammasomes are highly expressed in gingival tissues with severe chronic periodontitis. In human THP-1 cells, P. gingivalis activates the synthesis and secretion of IL-1β to higher levels than S. mitis. Importantly, NLRP3-, Caspase-1-, and Caspase-4-siRNA knockdown THP-1 cells treated with P. gingivalis exhibited a lower expression level of IL-1β as compared to the control cells. In addition, silencing of either CASP4 or CASP1 can lead to a concurrent or reciprocal decrease in the expression of the other. Of note, the IL-1β induction is not affected in the S. mitis-treated THP-1 cells with the silence of NLRP3, Caspase-1, and Caspase-4 genes. Conclusion: NLRP3/Caspase-4 and NLRP3/Caspase-1 dependent IL-1β production may crucially contribute to the dysregulated immuno-inflammatory response in periodontal pathogenesis.
Collapse
Affiliation(s)
- Pei-Hui Ding
- Department of Periodontology, Stomatology Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Meng-Xin Yang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China.,Department of Periodontology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na-Na Wang
- Department of Periodontology, Fuyang People's Hospital, Fuyang, China
| | - Li-Jian Jin
- Division of Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yan Dong
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China.,Department of Prosthodontics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Cai
- Department of Periodontology, Stomatology Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Li-Li Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China.,Department of Periodontology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Kumari P, Russo AJ, Shivcharan S, Rathinam VA. AIM2 in health and disease: Inflammasome and beyond. Immunol Rev 2020; 297:83-95. [PMID: 32713036 DOI: 10.1111/imr.12903] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022]
Abstract
Nucleic acid sensing is a critical mechanism by which the immune system monitors for pathogen invasion. A set of germline-encoded innate immune receptors detect microbial DNA in various compartments of the cell, such as endosomes, the cytosol, and the nucleus. Sensing of microbial DNA through these receptors stimulates, in most cases, interferon regulatory factor-dependent type I IFN synthesis followed by JAK/STAT-dependent interferon-stimulated gene expression. In contrast, the detection of DNA in the cytosol by AIM2 assembles a macromolecular complex called the inflammasome, which unleashes the proteolytic activity of a cysteine protease caspase-1. Caspase-1 cleaves and activates the pro-inflammatory cytokines such as IL-1β and IL-18 and a pore-forming protein, gasdermin D, which triggers pyroptosis, an inflammatory form of cell death. Research over the past decade has revealed that AIM2 plays essential roles not only in host defense against pathogens but also in inflammatory diseases, autoimmunity, and cancer in inflammasome-dependent and inflammasome-independent manners. This review discusses the latest advancements in our understanding of AIM2 biology and its functions in health and disease.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Sonia Shivcharan
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| |
Collapse
|
27
|
Outer Membrane Lipid Secretion and the Innate Immune Response to Gram-Negative Bacteria. Infect Immun 2020; 88:IAI.00920-19. [PMID: 32253250 DOI: 10.1128/iai.00920-19] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The outer membrane (OM) of Gram-negative bacteria is an asymmetric lipid bilayer that consists of inner leaflet phospholipids and outer leaflet lipopolysaccharides (LPS). The asymmetric character and unique biochemistry of LPS molecules contribute to the OM's ability to function as a molecular permeability barrier that protects the bacterium against hazards in the environment. Assembly and regulation of the OM have been extensively studied for understanding mechanisms of antibiotic resistance and bacterial defense against host immunity; however, there is little knowledge on how Gram-negative bacteria release their OMs into their environment to manipulate their hosts. Discoveries in bacterial lipid trafficking, OM lipid homeostasis, and host recognition of microbial patterns have shed new light on how microbes secrete OM vesicles (OMVs) to influence inflammation, cell death, and disease pathogenesis. Pathogens release OMVs that contain phospholipids, like cardiolipins, and components of LPS molecules, like lipid A endotoxins. These multiacylated lipid amphiphiles are molecular patterns that are differentially detected by host receptors like the Toll-like receptor 4/myeloid differentiation factor 2 complex (TLR4/MD-2), mouse caspase-11, and human caspases 4 and 5. We discuss how lipid ligands on OMVs engage these pattern recognition receptors on the membranes and in the cytosol of mammalian cells. We then detail how bacteria regulate OM lipid asymmetry, negative membrane curvature, and the phospholipid-to-LPS ratio to control OMV formation. The goal is to highlight intersections between OM lipid regulation and host immunity and to provide working models for how bacterial lipids influence vesicle formation.
Collapse
|
28
|
Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discov 2020; 6:36. [PMID: 32550001 PMCID: PMC7280307 DOI: 10.1038/s41421-020-0167-x] [Citation(s) in RCA: 606] [Impact Index Per Article: 121.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/05/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are cytoplasmic multiprotein complexes comprising a sensor protein, inflammatory caspases, and in some but not all cases an adapter protein connecting the two. They can be activated by a repertoire of endogenous and exogenous stimuli, leading to enzymatic activation of canonical caspase-1, noncanonical caspase-11 (or the equivalent caspase-4 and caspase-5 in humans) or caspase-8, resulting in secretion of IL-1β and IL-18, as well as apoptotic and pyroptotic cell death. Appropriate inflammasome activation is vital for the host to cope with foreign pathogens or tissue damage, while aberrant inflammasome activation can cause uncontrolled tissue responses that may contribute to various diseases, including autoinflammatory disorders, cardiometabolic diseases, cancer and neurodegenerative diseases. Therefore, it is imperative to maintain a fine balance between inflammasome activation and inhibition, which requires a fine-tuned regulation of inflammasome assembly and effector function. Recently, a growing body of studies have been focusing on delineating the structural and molecular mechanisms underlying the regulation of inflammasome signaling. In the present review, we summarize the most recent advances and remaining challenges in understanding the ordered inflammasome assembly and activation upon sensing of diverse stimuli, as well as the tight regulations of these processes. Furthermore, we review recent progress and challenges in translating inflammasome research into therapeutic tools, aimed at modifying inflammasome-regulated human diseases.
Collapse
Affiliation(s)
- Danping Zheng
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001 Israel
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Timur Liwinski
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001 Israel
- 1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001 Israel
- Cancer-Microbiome Division Deutsches Krebsforschungszentrum (DKFZ), Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
29
|
Shenker BJ, Walker LM, Zekavat Z, Ojcius DM, Huang PR, Boesze-Battaglia K. Cytolethal distending toxin-induced release of interleukin-1β by human macrophages is dependent upon activation of glycogen synthase kinase 3β, spleen tyrosine kinase (Syk) and the noncanonical inflammasome. Cell Microbiol 2020; 22:e13194. [PMID: 32068949 DOI: 10.1111/cmi.13194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 12/19/2022]
Abstract
Cytolethal distending toxins (Cdt) are a family of toxins produced by several human pathogens which infect mucocutaneous tissue and induce inflammatory disease. We have previously demonstrated that the Aggregatibacter actinomycetemcomitans Cdt induces a pro-inflammatory response from human macrophages which involves activation of the NLRP3 inflammasome. We now demonstrate that in addition to activating caspase-1 (canonical inflammasome), Cdt treatment leads to caspase-4 activation and involvement of the noncanonical inflammasome. Cdt-treated cells exhibit pyroptosis characterised by cleavage of gasdermin-D (GSDMD), release of HMGB1 at 24 hr and LDH at 48 hr. Inhibition of either the canonical (caspase-1) or noncanonical (caspase-4) inflammasome blocks both Cdt-induced release of IL-1β and induction of pyroptosis. Analysis of upstream events indicates that Cdt induces Syk phosphorylation (activation); furthermore, blockade of Syk expression and inhibition of pSyk activity inhibit both Cdt-induced cytokine release and pyroptosis. Finally, we demonstrate that increases in pSyk are dependent upon Cdt-induced activation of GSK3β. These studies advance our understanding of Cdt function and provide new insight into the virulence potential of Cdt in mediating the pathogenesis of disease caused by Cdt-producing organisms such as A. actinomycetemcomitans.
Collapse
Affiliation(s)
- Bruce J Shenker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Lisa M Walker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Zeyed Zekavat
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, USA
| | - Pei-Rong Huang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Gomes MTR, Cerqueira DM, Guimarães ES, Campos PC, Oliveira SC. Guanylate-binding proteins at the crossroad of noncanonical inflammasome activation during bacterial infections. J Leukoc Biol 2019; 106:553-562. [PMID: 30897250 PMCID: PMC7516346 DOI: 10.1002/jlb.4mr0119-013r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/28/2019] [Accepted: 03/10/2019] [Indexed: 12/14/2022] Open
Abstract
The immune system is armed with a broad range of receptors to detect and initiate the elimination of bacterial pathogens. Inflammasomes are molecular platforms that sense a diverse range of microbial insults to develop appropriate host response. In that context, noncanonical inflammasome arose as a sensor for Gram-negative bacteria-derived LPS leading to the control of infections. This review describes the role of caspase-11/gasdermin-D-dependent immune response against Gram-negative bacteria and presents an overview of guanylate-binding proteins (GBPs) at the interface of noncanonical inflammasome activation. Indeed, caspase-11 acts as a receptor for LPS and this interaction elicits caspase-11 autoproteolysis that is required for its optimal catalytic activity. Gasdermin-D is cleaved by activated caspase-11 generating an N-terminal domain that is inserted into the plasmatic membrane to form pores that induce pyroptosis, a cell death program involved in intracellular bacteria elimination. This mechanism also promotes IL-1β release and potassium efflux that connects caspase-11 to NLRP3 activation. Furthermore, GBPs display many features to allow LPS recognition by caspase-11, initiating the noncanonical inflammasome response prompting the immune system to control bacterial infections. In this review, we discuss the recent findings and nuances related to this mechanism and its biological functions.
Collapse
Affiliation(s)
- Marco Túlio R Gomes
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daiane M Cerqueira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Erika S Guimarães
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Priscila C Campos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
31
|
Immunological Pathways Triggered by Porphyromonas gingivalis and Fusobacterium nucleatum: Therapeutic Possibilities? Mediators Inflamm 2019; 2019:7241312. [PMID: 31341421 PMCID: PMC6612971 DOI: 10.1155/2019/7241312] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/28/2019] [Accepted: 05/19/2019] [Indexed: 02/06/2023] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum) are Gram-negative anaerobic bacteria possessing several virulence factors that make them potential pathogens associated with periodontal disease. Periodontal diseases are chronic inflammatory diseases of the oral cavity, including gingivitis and periodontitis. Periodontitis can lead to tooth loss and is considered one of the most prevalent diseases worldwide. P. gingivalis and F. nucleatum possess virulence factors that allow them to survive in hostile environments by selectively modulating the host's immune-inflammatory response, thereby creating major challenges to host cell survival. Studies have demonstrated that bacterial infection and the host immune responses are involved in the induction of periodontitis. The NLRP3 inflammasome and its effector molecules (IL-1β and caspase-1) play roles in the development of periodontitis. We and others have reported that the purinergic P2X7 receptor plays a role in the modulation of periodontal disease and intracellular pathogen control. Caspase-4/5 (in humans) and caspase-11 (in mice) are important effectors for combating bacterial pathogens via mediation of cell death and IL-1β release. The exact molecular events of the host's response to these bacteria are not fully understood. Here, we review innate and adaptive immune responses induced by P. gingivalis and F. nucleatum infections and discuss the possibility of manipulations of the immune response as therapeutic strategies. Given the global burden of periodontitis, it is important to develop therapeutic targets for the prophylaxis of periodontopathogen infections.
Collapse
|
32
|
Kumar V. The complement system, toll-like receptors and inflammasomes in host defense: three musketeers’ one target. Int Rev Immunol 2019; 38:131-156. [PMID: 31066339 DOI: 10.1080/08830185.2019.1609962] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vijay Kumar
- Children’s Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, QLD, Australia
| |
Collapse
|
33
|
Rathinam VAK, Zhao Y, Shao F. Innate immunity to intracellular LPS. Nat Immunol 2019; 20:527-533. [PMID: 30962589 PMCID: PMC7668400 DOI: 10.1038/s41590-019-0368-3] [Citation(s) in RCA: 422] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/04/2019] [Indexed: 12/20/2022]
Abstract
Monitoring of the cytosolic compartment by the innate immune system for pathogen-encoded products or pathogen activities often enables the activation of a subset of caspases. In most cases, the cytosolic surveillance pathways are coupled to activation of caspase-1 via canonical inflammasome complexes. A related set of caspases, caspase-11 in rodents and caspase-4 and caspase-5 in humans, monitors the cytosol for bacterial lipopolysaccharide (LPS). Direct activation of caspase-11, caspase-4 and caspase-5 by intracellular LPS elicits the lytic cell death called 'pyroptosis', which occurs in multiple cell types. The pyroptosis is executed by the pore-forming protein GSDMD, which is activated by cleavage mediated by caspase-11, caspase-4 or caspase-5. In monocytes, formation of GSDMD pores can induce activation of the NLRP3 inflammasome for maturation of the cytokines IL-1β and IL-18. Caspase-11-mediated pyroptosis in response to cytosolic LPS is critical for antibacterial defense and septic shock. Here we review the emerging literature on the sensing of cytosolic LPS and its regulation and pathophysiological functions.
Collapse
Affiliation(s)
- Vijay A K Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA.
| | - Yue Zhao
- National Institute of Biological Sciences, Beijing, China
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, China.
| |
Collapse
|
34
|
Pfalzgraff A, Weindl G. Intracellular Lipopolysaccharide Sensing as a Potential Therapeutic Target for Sepsis. Trends Pharmacol Sci 2019; 40:187-197. [DOI: 10.1016/j.tips.2019.01.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 12/22/2022]
|
35
|
Di Lorenzo F, De Castro C, Silipo A, Molinaro A. Lipopolysaccharide structures of Gram-negative populations in the gut microbiota and effects on host interactions. FEMS Microbiol Rev 2019; 43:257-272. [DOI: 10.1093/femsre/fuz002] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
| | - Cristina De Castro
- Task Force on Microbiome Studies, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
- Department of Agricultural Sciences, University of Naples Federico II, via Università 100, 80055 Portici, Italy
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, via Cinthia 4, 80126 Naples, Italy
| |
Collapse
|
36
|
The Inflammatory Response to Enterotoxigenic E. coli and Probiotic E. faecium in a Coculture Model of Porcine Intestinal Epithelial and Dendritic Cells. Mediators Inflamm 2018; 2018:9368295. [PMID: 30670931 PMCID: PMC6317115 DOI: 10.1155/2018/9368295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/23/2018] [Indexed: 12/31/2022] Open
Abstract
The gut epithelium constitutes an interface between the intestinal contents and the underlying gut-associated lymphoid tissue (GALT) including dendritic cells (DC). Interactions of intestinal epithelial cells (IEC) and resident DC are characterized by bidirectional crosstalk mediated by various factors, such as transforming growth factor-β (TGF-β) and thymic stromal lymphopoietin (TSLP). In the present study, we aimed (1) to model the interplay of both cell types in a porcine in vitro coculture consisting of IEC (cell line IPEC-J2) and monocyte-derived DC (MoDC) and (2) to assess whether immune responses to bacteria are altered because of the interplay between IPEC-J2 cells and MoDC. With regard to the latter, we focused on the inflammasome pathway. Here, we propose caspase-13 as a promising candidate for the noncanonical inflammasome activation in pigs. We conducted challenge experiments with enterotoxigenic Escherichia coli (ETEC) and probiotic Enterococcus faecium (E. faecium) NCIMB 10415. As potential mediators of IEC/DC interactions, TGF-β and TSLP were selected for analyses. Cocultured MoDC showed attenuated ETEC-induced inflammasome-related and proinflammatory interleukin (IL)-8 reactions compared with MoDC monocultures. Caspase-13 was more strongly expressed in IPEC-J2 cells cocultured with MoDC and upon ETEC incubation. We found that IPEC-J2 cells and MoDC were capable of releasing TSLP. The latter cells secreted greater amounts of TSLP when cocultured with IPEC-J2 cells. TGF-β was not modulated under the present experimental conditions in either cell types. We conclude that, in the presence of IPEC-J2 cells, porcine MoDC exhibited a more tolerogenic phenotype, which might be partially regulated by autocrine TSLP production. Noncanonical inflammasome signaling appeared to be modulated in IPEC-J2 cells. Our results indicate that the reciprocal interplay of the intestinal epithelium and GALT is essential for promoting balanced immune responses.
Collapse
|
37
|
Xia X, Wang X, Zheng Y, Jiang J, Hu J. What role does pyroptosis play in microbial infection? J Cell Physiol 2018; 234:7885-7892. [PMID: 30537070 DOI: 10.1002/jcp.27909] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/25/2018] [Indexed: 12/18/2022]
Abstract
Pyroptosis, a type of programmed cell death mediated by gasdermin, is characterized by the swelling and rupture of cells, release of cellular contents and a strong inflammatory response, which is critical for controlling microbial infection. Pattern recognition receptors recognize the intracellular and extracellular pathogenic microbial components and stimulate the organism's inflammatory response by activating the pyroptosis signaling pathway and releasing interleukin-1β (IL-1β), IL-18, and other inflammatory factors to promote pathogen clearance and prevent infection. In the process of continuous evolution, pathogens have developed multiple strategies to modulate the occurrence of pyroptosis and thus enhance their ability to induce disease; that is, the competition between host cells and pathogens controls the occurrence of pyroptosis. Competition can directly affect tissue inflammation outbreaks and even alter cell survival. Studies have shown that various bacterial infections, including Shigella flexneri, Salmonella, Listeria monocytogenes, and Legionella pneumophila, can lead to pyroptosis. Pyroptosis is associated with the occurrence and development of various diseases caused by microbial infection, and the identification of molecules related to the pyroptosis signaling pathway may provide new drug targets for the treatment of related diseases. This study reviews the molecular mechanisms of pyroptosis and the role of pyroptosis in microbial infection-related diseases.
Collapse
Affiliation(s)
- Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.,Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China.,Postdoctoral Research Station, Henan Agriculture University, Zhengzhou, China
| | - Xin Wang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Yi Zheng
- College of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jinqing Jiang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.,Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
38
|
Bertinaria M, Gastaldi S, Marini E, Giorgis M. Development of covalent NLRP3 inflammasome inhibitors: Chemistry and biological activity. Arch Biochem Biophys 2018; 670:116-139. [PMID: 30448387 DOI: 10.1016/j.abb.2018.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022]
Abstract
The NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is the best recognized and most widely implicated regulator of caspase-1 activation. It is a key regulator of innate immune response and is involved in many pathophysiological processes. Recent evidences for its inappropriate activation in autoinflammatory, autoimmune, as well as in neurodegenerative diseases attract a growing interest toward the development of small molecules NLRP3 inhibitors. Based on the knowledge of biochemical and structural aspects of NLRP3 activation, one successful strategy in the identification of NLRP3 inhibitors relies on the development of covalent irreversible inhibitors. Covalent inhibitors are reactive electrophilic molecules able to alkylate nucleophiles in the target protein. These inhibitors could ensure good efficacy and prolonged duration of action both in vitro and in vivo. In spite of these advantages, effects on other signalling pathways, prone to alkylation, may occur. In this review, we will illustrate the chemistry and the biological action of the most studied covalent NLRP3 inhibitors developed so far. A description of what we know about their mechanism of action will address the reader toward a critical understanding of NLRP3 inhibition by electrophilic compounds.
Collapse
Affiliation(s)
- Massimo Bertinaria
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125, Torino, Italy.
| | - Simone Gastaldi
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125, Torino, Italy
| | - Elisabetta Marini
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125, Torino, Italy
| | - Marta Giorgis
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125, Torino, Italy
| |
Collapse
|
39
|
Rüter C, Lubos ML, Norkowski S, Schmidt MA. All in—Multiple parallel strategies for intracellular delivery by bacterial pathogens. Int J Med Microbiol 2018; 308:872-881. [PMID: 29936031 DOI: 10.1016/j.ijmm.2018.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/01/2018] [Accepted: 06/16/2018] [Indexed: 02/06/2023] Open
|
40
|
Abstract
In humans and other mammals, recognition of endotoxins—abundant surface lipopolysaccharides (LPS) of Gram-negative bacteria—provides a potent stimulus for induction of inflammation and mobilization of host defenses. The structurally unique lipid A region of LPS is the principal determinant of this pro-inflammatory activity. This region of LPS is normally buried within the bacterial outer membrane and aggregates of purified LPS, making even more remarkable its picomolar potency and the ability of discrete variations in lipid A structure to markedly alter the pro-inflammatory activity of LPS. Two recognition systems—MD-2/TLR4 and “LPS-sensing” cytosolic caspases—together confer LPS responsiveness at the host cell surface, within endosomes, and at sites physically accessible to the cytosol. Understanding how the lipid A of LPS is delivered and recognized at these diverse sites is crucial to understanding how the magnitude and character of the inflammatory responses are regulated.
Collapse
Affiliation(s)
- Jerrold Weiss
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Jason Barker
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| |
Collapse
|
41
|
Schroder K, Kanneganti TD, Shao F, Broz P. Mechanisms and Consequences of Inflammasome Activation. J Mol Biol 2017; 430:131-132. [PMID: 29247654 DOI: 10.1016/j.jmb.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kate Schroder
- Institute for Molecular Bioscience, Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia 4072, Australia.
| | | | - Feng Shao
- National Institute of Biological Sciences, #7 Science Park Rd, Beijing, China.
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland.
| |
Collapse
|