51
|
Toraman A, Sağlam E, Savran L, Sağlam M, Köseoğlu S. Salivary levels of NLRC4 inflammasome in different periodontal clinical status. Oral Dis 2023; 29:2765-2771. [PMID: 36327138 DOI: 10.1111/odi.14424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/20/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Nucleotide-binding and oligomerization domain (NOD)-like receptor family CARD domain-containing protein 4 (NLRC4) has a critical role in the regulation of interleukin-1β (IL-1β), an important cytokine in the pathogenesis of the periodontal diseases. In this study, we aimed to evaluate levels of salivary NLRC4 inflammasomes in different periodontal clinical statuses. METHODS The individuals with 20 periodontally healthy (healthy), 20 gingivitis, and 20 periodontitis were periodontally examined. Saliva samples were collected, after the clinical measurements (plaque index, gingival index, gingival bleeding index, probing depth, and clinical attachment level). The levels of salivary NLRC4, IL-1β, and interleukin 10 (IL-10) were examined by enzyme-linked immunosorbent assay. RESULTS The results demonstrated that levels of salivary NLRC4 (p < 0.01), and IL-1β (p < 0.001) were significantly higher in gingivitis and periodontitis than in the healthy group. No significant difference was salivary IL-10 levels between the groups (p > 0.05). Positive significant correlations among NLRC4 and IL-1β salivary levels and clinical parameters were detected (p < 0.05). CONCLUSION The findings of this study suggest that the NLRC4 is elevated in periodontal disease. Larger randomized controlled clinical studies are needed to use salivary NLRC4 levels as a potential marker for detecting the presence and/or severity of the periodontal disease.
Collapse
Affiliation(s)
- Ayşe Toraman
- Department of Periodontology, Hamidiye Faculty of Dentistry, Health Sciences University, İstanbul, Turkey
| | - Ebru Sağlam
- Department of Periodontology, Hamidiye Faculty of Dentistry, Health Sciences University, İstanbul, Turkey
| | - Levent Savran
- Department of Periodontology, Faculty of Dentistry, İzmir Katip Çelebi University, İzmir, Turkey
| | - Mehmet Sağlam
- Department of Periodontology, Faculty of Dentistry, İzmir Katip Çelebi University, İzmir, Turkey
| | - Serhat Köseoğlu
- Department of Periodontology, Faculty of Dentistry, İstanbul Medeniyet University, İstanbul, Turkey
| |
Collapse
|
52
|
An Y, Zhai Z, Wang X, Ding Y, He L, Li L, Mo Q, Mu C, Xie R, Liu T, Zhong W, Wang B, Cao H. Targeting Desulfovibrio vulgaris flagellin-induced NAIP/NLRC4 inflammasome activation in macrophages attenuates ulcerative colitis. J Adv Res 2023; 52:219-232. [PMID: 37586642 PMCID: PMC10555950 DOI: 10.1016/j.jare.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION The perturbations of gut microbiota could interact with excessively activated immune responses and play key roles in the etiopathogenesis of ulcerative colitis (UC). Desulfovibrio, the most predominant sulfate reducing bacteria (SRB) resided in the human gut, was observed to overgrow in patients with UC. The interactions between specific gut microbiota and drugs and their impacts on UC treatment have not been demonstrated well. OBJECTIVES This study aimed to elucidate whether Desulfovibrio vulgaris (D. vulgaris, DSV) and its flagellin could activate nucleotide-binding oligomerization domain-like receptors (NLR) family of apoptosis inhibitory proteins (NAIP) / NLR family caspase activation and recruitment domain-containing protein 4 (NLRC4) inflammasome and promote colitis, and further evaluate the efficacy of eugeniin targeting the interaction interface of D. vulgaris flagellin (DVF) and NAIP to attenuate UC. METHODS The abundance of DSV and the occurrence of macrophage pyroptosis in human UC tissues were investigated. Colitis in mice was established by dextran sulfate sodium (DSS) and gavaged with DSV or its purified flagellin. NAIP/NLRC4 inflammasome activation and macrophage pyroptosis were evaluated in vivo and in vitro. The effects of eugeniin on blocking the interaction of DVF and NAIP/NLRC4 and relieving colitis were also assessed. RESULTS The abundance of DSV increased in the feces of patients with UC and was found to be associated with disease activity. DSV and its flagellin facilitated DSS-induced colitis in mice. Mechanistically, RNA sequencing showed that gene expression associated with inflammasome complex and pyroptosis was upregulated after DVF treatment in macrophages. DVF was further demonstrated to induce significant macrophage pyroptosis in vitro, depending on NAIP/NLRC4 inflammasome activation. Furthermore, eugeniin was screened as an inhibitor of the interface between DVF and NAIP and successfully alleviated the proinflammatory effect of DVF in colitis. CONCLUSION Targeting DVF-induced NAIP/NLRC4 inflammasome activation and macrophage pyroptosis ameliorates UC. This finding is of great significance for exploring the gut microbiota-host interactions in UC development and providing new insights for precise treatment.
Collapse
Affiliation(s)
- Yaping An
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Zihan Zhai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yiyun Ding
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Linlin He
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Lingfeng Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Qi Mo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Chenlu Mu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
53
|
Sim J, Park J, Moon JS, Lim J. Dysregulation of inflammasome activation in glioma. Cell Commun Signal 2023; 21:239. [PMID: 37723542 PMCID: PMC10506313 DOI: 10.1186/s12964-023-01255-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/01/2023] [Indexed: 09/20/2023] Open
Abstract
Gliomas are the most common brain tumors characterized by complicated heterogeneity. The genetic, molecular, and histological pathology of gliomas is characterized by high neuro-inflammation. The inflammatory microenvironment in the central nervous system (CNS) has been closely linked with inflammasomes that control the inflammatory response and coordinate innate host defenses. Dysregulation of the inflammasome causes an abnormal inflammatory response, leading to carcinogenesis in glioma. Because of the clinical importance of the various physiological properties of the inflammasome in glioma, the inflammasome has been suggested as a promising treatment target for glioma management. Here, we summarize the current knowledge on the contribution of the inflammasomes in glioma and therapeutic insights. Video Abstract.
Collapse
Affiliation(s)
- JeongMin Sim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - JeongMan Park
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Jaejoon Lim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea.
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea.
| |
Collapse
|
54
|
Okin D, Kagan JC. Inflammasomes as regulators of non-infectious disease. Semin Immunol 2023; 69:101815. [PMID: 37506489 PMCID: PMC10527946 DOI: 10.1016/j.smim.2023.101815] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Inflammasomes are cytoplasmic organelles that stimulate inflammation upon cellular detection of infectious or non-infectious stress. While much foundational work has focused on the infection-associated aspects of inflammasome activities, recent studies have highlighted the role of inflammasomes in non-infectious cellular and organismal functions. Herein, we discuss the evolution of inflammasome components and highlight characteristics that permit inflammasome regulation of physiologic processes. We focus on emerging data that highlight the importance of inflammasome proteins in the regulation of reproduction, development, and malignancy. A framework is proposed to contextualize these findings.
Collapse
Affiliation(s)
- Daniel Okin
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
55
|
Zou Z, Zhao M, Yang Y, Xie Y, Li Z, Zhou L, Shang R, Zhou P. The role of pyroptosis in hepatocellular carcinoma. Cell Oncol (Dordr) 2023; 46:811-823. [PMID: 36864264 DOI: 10.1007/s13402-023-00787-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the main histologic type of liver cancer. It accounts for the majority of all diagnoses and deaths due to liver cancer. The induction of tumor cell death is an effective strategy to control tumor development. Pyroptosis is an inflammatory programmed cell death caused by microbial infection, accompanied by activation of inflammasomes and release of pro-inflammatory cytokines, interleukin-1β (IL-1β), and interleukin-18 (IL-18). The cleavage of gasdermins (GSDMs) promotes the occurrence of pyroptosis leading to cell swelling, lysis, and death. Accumulating evidence has indicated that pyroptosis influences the progression of HCC by regulating immune-mediated tumor cell death. Currently, some researchers hold the view that inhibition of pyroptosis-related components may prevent the incidence of HCC, but more researchers have the view that activation of pyroptosis exerts a tumor-inhibitory effect. Growing evidence indicates that pyroptosis can prevent or promote tumor development depending on the type of tumor. In this review, pyroptosis pathways and pyroptosis-related components were discussed. Next, the role of pyroptosis and its components in HCC was described. Finally, the therapeutic significance of pyroptosis in HCC was discussed.
Collapse
Affiliation(s)
- Zhimiao Zou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Minghui Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Yang Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Yalong Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Zeyang Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Liang Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Runshi Shang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Ping Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education, Chinese Academy of Medical Sciences, 1095 Jiefang Road, Wuhan, Hubei, 430030, China.
| |
Collapse
|
56
|
Clénet ML, Keaney J, Gillet G, Valadas JS, Langlois J, Cardenas A, Gasser J, Kadiu I. Divergent functional outcomes of NLRP3 blockade downstream of multi-inflammasome activation: therapeutic implications for ALS. Front Immunol 2023; 14:1190219. [PMID: 37575265 PMCID: PMC10415077 DOI: 10.3389/fimmu.2023.1190219] [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: 03/20/2023] [Accepted: 06/26/2023] [Indexed: 08/15/2023] Open
Abstract
NOD-Like Receptor Family Pyrin Domain Containing 3 (NLRP3) inflammasome modulation has emerged as a potential therapeutic approach targeting inflammation amplified by pyroptotic innate immune cell death. In diseases characterized by non-cell autonomous neurodegeneration including amyotrophic lateral sclerosis (ALS), the activation of several inflammasomes has been reported. Since functional redundancy can exist among inflammasome pathways, here we investigate the effects of NLRP3 inhibition on NLRP3, NLR family CARD Domain Containing 4 (NLRC4) and non-canonical pathways to understand whether NLRP3 blockade alone can mitigate pro-inflammatory cytokine release and pyroptotic cell death in contexts where single or multiple inflammasome pathways independent of NLRP3 are activated. In this study we do not limit our insights into inflammasome biology by solely relying on the THP-1 monocytic line under the LPS/nigericin-mediated NLRP3 pathway activation paradigm. We assess therapeutic potential and limitations of NLRP3 inhibition in multi-inflammasome activation contexts utilizing various human cellular systems including cell lines expressing gain of function (GoF) mutations for several inflammasomes, primary human monocytes, macrophages, healthy and Amyotrophic Lateral Sclerosis (ALS) patient induced pluripotent stem cells (iPSC)-derived microglia (iMGL) stimulated for canonical and non-canonical inflammasome pathways. We demonstrate that NLRP3 inhibition can modulate the NLRC4 and non-canonical inflammasome pathways; however, these effects differ between immortalized, human primary innate immune cells, and iMGL. We extend our investigation in more complex systems characterized by activation of multiple inflammasomes such as the SOD1G93A mouse model. Through deep immune phenotyping by single-cell mass cytometry we demonstrate that acute NLRP3 inhibition does not ameliorate spinal cord inflammation in this model. Taken together, our data suggests that NLRP3 inhibition alone may not be sufficient to address dynamic and complex neuroinflammatory pathobiological mechanisms including dysregulation of multiple inflammasome pathways in neurodegenerative disease such as ALS.
Collapse
Affiliation(s)
- Marie-Laure Clénet
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - James Keaney
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Jorge S. Valadas
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Julie Langlois
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Alvaro Cardenas
- Development Science, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Julien Gasser
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| | - Irena Kadiu
- Neuroinflammation Focus Area, Neuroscience Research, Early Solutions, UCB Biopharma SRL, Braine l’Alleud, Belgium
| |
Collapse
|
57
|
Batta SM, Far NNE, Soliman NA, Nassar SO. A Study of NLRC4 in Patients with Leprosy. Indian J Dermatol 2023; 68:385-392. [PMID: 37822385 PMCID: PMC10564182 DOI: 10.4103/ijd.ijd_781_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Background Activation of the NLRC4 inflammasome appears to start many signalling processes inside the host, including caspase-1, the principal protease responsible for converting proIL-1β and IL-18 to active, secreted IL-1β and IL-18, resulting in pyroptosis. Aims To evaluate NLRC4 level in patient's blood serum to highlight its role in the pathogenesis of leprosy. Materials and Methods This prospective study was conducted on 40 patients with leprosy and 30 healthy individuals of matched ages and sexes. All patients were subjected to complete history taking, general and dermatological examination, laboratory investigations, slit skin smear with bacillary index, and clinical classification of the studied leprosy group patients regarding disability according to disability grading. And finally, measurement of serum NLRC4 level by ELISA. Results In the paucibacillary (PB) group, NLRC4 serum level ranged from 0.9 to 1.8 ng/ml with 1.43 ± 0.28 ng/ml, while in the multibacillary (MB) group, it ranged from 1.2 to 5.7 ng/ml with 2.83 ± 1.11 ng/ml. NLRC4 serum level had increased significantly in MB patients compared to PB patients (P < 0.05). There was a significant difference among the three studied groups, regarding the serum level of NLRC4 (P < 0.05). In leprosy patients, significant positive correlations were found between serum levels of NLRC4 and bacillary index and duration of leprosy. Conclusions Leprosy patients had considerably greater serum levels of NLRC4 than controls. It was much greater in MB patients than in PB patients.
Collapse
Affiliation(s)
- Salma Mosaad Batta
- From the Dermatology and Venereology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nashwa Naeem El Far
- From the Dermatology and Venereology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nema Ali Soliman
- From the Dermatology and Venereology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Samia Othman Nassar
- From the Dermatology and Venereology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
58
|
Chen S, Lei Q, Zou X, Ma D. The role and mechanisms of gram-negative bacterial outer membrane vesicles in inflammatory diseases. Front Immunol 2023; 14:1157813. [PMID: 37398647 PMCID: PMC10313905 DOI: 10.3389/fimmu.2023.1157813] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Outer membrane vesicles (OMVs) are spherical, bilayered, and nanosized membrane vesicles that are secreted from gram-negative bacteria. OMVs play a pivotal role in delivering lipopolysaccharide, proteins and other virulence factors to target cells. Multiple studies have found that OMVs participate in various inflammatory diseases, including periodontal disease, gastrointestinal inflammation, pulmonary inflammation and sepsis, by triggering pattern recognition receptors, activating inflammasomes and inducing mitochondrial dysfunction. OMVs also affect inflammation in distant organs or tissues via long-distance cargo transport in various diseases, including atherosclerosis and Alzheimer's disease. In this review, we primarily summarize the role of OMVs in inflammatory diseases, describe the mechanism through which OMVs participate in inflammatory signal cascades, and discuss the effects of OMVs on pathogenic processes in distant organs or tissues with the aim of providing novel insights into the role and mechanism of OMVs in inflammatory diseases and the prevention and treatment of OMV-mediated inflammatory diseases.
Collapse
|
59
|
Feng WD, Wang Y, Luo T, Jia X, Cheng CQ, Wang HJ, Zhang MQ, Li QQ, Wang XJ, Li YY, Wang JY, Huang GR, Wang T, Xu AL. Scoparone suppresses mitophagy-mediated NLRP3 inflammasome activation in inflammatory diseases. Acta Pharmacol Sin 2023; 44:1238-1251. [PMID: 36522512 PMCID: PMC10203299 DOI: 10.1038/s41401-022-01028-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/07/2022] [Indexed: 12/23/2022]
Abstract
Recent evidence shows that targeting NLRP3 inflammasome activation is an important means to treat inflammasome-driven diseases. Scoparone, a natural compound isolated from the Chinese herb Artemisia capillaris Thunb, has anti-inflammatory activity. In this study we investigated the effect of scoparone on NLRP3 inflammasome activation in inflammatory diseases. In LPS-primed, ATP or nigericin-stimulated mouse macrophage J774A.1 cells and bone marrow-derived macrophages (BMDMs), pretreatment with scoparone (50 μM) markedly restrained canonical and noncanonical NLRP3 inflammasome activation, evidenced by suppressed caspase-1 cleavage, GSDMD-mediated pyroptosis, mature IL-1β secretion and the formation of ASC specks. We then conducted a transcriptome analysis in scoparone-pretreated BMDMs, and found that the differentially expressed genes were significantly enriched in mitochondrial reactive oxygen species (ROS) metabolic process, mitochondrial translation and assembly process, as well as in inflammatory response. We demonstrated in J774A.1 cells and BMDMs that scoparone promoted mitophagy, a well-characterized mechanism to control mitochondrial quality and reduce ROS production and subsequent NLRP3 inflammasome activation. Mitophagy blockade by 3-methyladenine (3-MA, 5 mM) reversed the protective effects of scoparone on mitochondrial damage and inflammation in the murine macrophages. Moreover, administration of scoparone (50 mg/kg) exerted significant preventive effects via inhibition of NLRP3 activation in mouse models of bacterial enteritis and septic shock. Collectively, scoparone displays potent anti-inflammatory effects via blocking NLRP3 inflammasome activation through enhancing mitophagy, highlighting a potential action mechanism in treating inflammasome-related diseases for further clinical investigation.
Collapse
Affiliation(s)
- Wan-di Feng
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tong Luo
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin Jia
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cui-Qin Cheng
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hao-Jia Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mei-Qi Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qi-Qi Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Jiao Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi-Ying Li
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jin-Yong Wang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guang-Rui Huang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - An-Long Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
60
|
Franklin ME, Bennett C, Arboite M, Alvarez-Ciara A, Corrales N, Verdelus J, Dietrich WD, Keane RW, de Rivero Vaccari JP, Prasad A. Activation of inflammasomes and their effects on neuroinflammation at the microelectrode-tissue interface in intracortical implants. Biomaterials 2023; 297:122102. [PMID: 37015177 PMCID: PMC10614166 DOI: 10.1016/j.biomaterials.2023.122102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Invasive neuroprosthetics rely on microelectrodes (MEs) to record or stimulate the activity of large neuron assemblies. However, MEs are subjected to tissue reactivity in the central nervous system (CNS) due to the foreign body response (FBR) that contribute to chronic neuroinflammation and ultimately result in ME failure. An endogenous, acute set of mechanisms responsible for the recognition and targeting of foreign objects, called the innate immune response, immediately follows the ME implant-induced trauma. Inflammasomes are multiprotein structures that play a critical role in the initiation of an innate immune response following CNS injuries. The activation of inflammasomes facilitates a range of innate immune response cascades and results in neuroinflammation and programmed cell death. Despite our current understanding of inflammasomes, their roles in the context of neural device implantation remain unknown. In this study, we implanted a non-functional Utah electrode array (UEA) into the rat somatosensory cortex and studied the inflammasome signaling and the corresponding downstream effects on inflammatory cytokine expression and the inflammasome-mediated cell death mechanism of pyroptosis. Our results not only demonstrate the continuous activation of inflammasomes and their contribution to neuroinflammation at the electrode-tissue interface but also reveal the therapeutic potential of targeting inflammasomes to attenuate the FBR in invasive neuroprosthetics.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Maelle Arboite
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jennifer Verdelus
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
| | - Robert W Keane
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
61
|
Zhou W, Zhao L, Wang H, Liu X, Liu Y, Xu K, Yu H, Suda K, He Y. Pyroptosis: A promising target for lung cancer therapy. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:94-101. [PMID: 39170826 PMCID: PMC11332860 DOI: 10.1016/j.pccm.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Indexed: 08/23/2024]
Abstract
Pyroptosis is a type of programed cell death that differs from apoptosis, ferroptosis, or necrosis. Numerous studies have reported that it plays a critical role in tumorigenesis and modification of the tumor microenvironment in multiple tumors. In this review, we briefly describe the canonical, non-canonical, and alternative mechanisms of pyroptotic cell death. We also summarize the potential roles of pyroptosis in oncogenesis, tumor development, and lung cancer treatment, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Pyroptosis has double-edged effects on the modulation of the tumor environment and lung cancer treatment. Further exploration of pyroptosis-based drugs could provide novel therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Wensheng Zhou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Xinyue Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Yujin Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Kandi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Yu
- Department of Medicine, Division of Medical Oncology and Department of Pathology, University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Kenichi Suda
- Department of Surgery, Division of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama 589-8511, Japan
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
62
|
Ma Q. Pharmacological Inhibition of the NLRP3 Inflammasome: Structure, Molecular Activation, and Inhibitor-NLRP3 Interaction. Pharmacol Rev 2023; 75:487-520. [PMID: 36669831 PMCID: PMC10121800 DOI: 10.1124/pharmrev.122.000629] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023] Open
Abstract
The nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a multiprotein complex that combines sensing, regulation, and effector functions to regulate inflammation in health and disease. NLRP3 is activated by a diverse range of inflammation-instigating signals known as pathogen associated molecular patterns and danger associated molecular patterns. Upon activation, NLRP3 oligomerizes and recruits partner proteins to form a supramolecular platform to process the maturation and release of interleukin (IL)-1β, IL-18, and gasdermin D, major mediators of inflammation and inflammatory cell death termed pyroptosis. The NLRP3 inflammasome has been implicated in the pathogenesis of a wide range of disease conditions, including chronic inflammatory disease that are associated with lifestyle and dietary changes, aging, and environmental exposures, and have become the leading cause of death worldwide. Pharmacological targeting of NLRP3 and signaling demonstrated promising efficacy in ameliorating a list of disease conditions in animal models. These findings underscore the potential and importance of NLRP3 as a druggable target for treating a range of diseases. In this review, recent progress in understanding the structure and mechanism of action of the NLRP3 inflammasome is discussed with focus on pharmacological inhibition of NLRP3 by small molecule inhibitors. New structural and mechanistic insights into NLRP3 activation and inhibitor-NLRP3 interactions would aid in the rational design and pharmacological evaluation of NLRP3 inhibitors for treatment of human disease. SIGNIFICANCE STATEMENT: The NLRP3 inflammasome plays central role in innate immune sensing and control of inflammation. Pharmacological inhibition of NLRP3 demonstrated promising efficacy in a range of diseases in animal models. Recent elucidation of the structure and inhibitor binding of NLRP3 generated new insights into its mode of action and inhibitor-NLRP3 interaction at molecular levels, providing new framework for developing small chemical inhibitors of NLRP3 with improved efficacy and specificity against chronic disease that has become major health concerns worldwide.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| |
Collapse
|
63
|
Gao Y, Yang Y, Wei J, Yue J, Wang Y, Zhang Q, Jin M, Wang R, Yang X, Zhang J, Liu X, Liu L, Zhang Y, Yang R. LNCGM1082-mediated NLRC4 activation drives resistance to bacterial infection. Cell Mol Immunol 2023; 20:475-488. [PMID: 36941318 PMCID: PMC10203293 DOI: 10.1038/s41423-023-00995-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 02/23/2023] [Indexed: 03/23/2023] Open
Abstract
The activation of NLRC4 is a major host response against intracellular bacteria infection. However, NLRC4 activation after a host senses diverse stimuli is difficult to understand. Here, we found that the lncRNA LNCGM1082 plays a critical role in the activation of NLRC4. LNCGM1082 in macrophages affects the maturation of interleukin (IL)-1β and pyroptotic cell death only after exposure to an NLRC4 ligand. Similar to NLRC4-/- mice, LNCGM1082-/- mice were highly sensitive to Salmonella Typhimurium (S. T) infection. LNCGM1082 deficiency in mouse or human macrophages inhibited IL-1β maturation and pyroptosis. Mechanistically, LNCGM1082 induced the binding of PKCδ with NLRC4 in both mice and humans. In contrast, NLRC4 did not bind PKCδ in LNCGM1082-/- macrophages. The activity of the lncRNA LNCGM1082 induced by S. T may be mediated through TLR5 in the macrophages of both mice and humans. In summary, our data indicate that TLR5-mediated LNCGM1082 activity can promote the binding of PKCδ with NLRC4 to activate NLRC4 and induce resistance to bacterial infection.
Collapse
Affiliation(s)
- Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yazheng Yang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Jianmei Wei
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Jianmei Yue
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Ya Wang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Qianjing Zhang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Mengli Jin
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Rong Wang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Xiaorong Yang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Junqi Zhang
- College of Life Science, Nankai University, Tianjin, 300121, China
| | - Xinqi Liu
- College of Life Science, Nankai University, Tianjin, 300121, China
| | - Lin Liu
- China National Center for Bioinformation & Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine and Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
64
|
Saad N, Duroux-Richard I, Touitou I, Jeziorski E, Apparailly F. MicroRNAs in inflammasomopathies. Immunol Lett 2023; 256-257:48-54. [PMID: 37023968 DOI: 10.1016/j.imlet.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
microRNAs (miRNAs) are small non-coding RNA sequences that negatively regulate the expression of protein-encoding genes at the post-transcriptional level. They play a role in the regulation of inflammatory responses by controlling the proliferation and activation of immune cells and their expression is disrupted in several immune-mediated inflammatory disorders. Among these, autoinflammatory diseases (AID) are a group of rare hereditary disorders caused by abnormal activation of the innate immune system and characterized by recurrent fevers. Major groups of AID are inflammasomopathies, which are associated with hereditary defects in the activation of inflammasomes, cytosolic multiprotein signaling complexes regulating IL-1 family cytokine maturation and pyroptosis. The study of the role of miRNAs in AID is only recently emerging and remains scarce in inflammasomopathies. In this review, we describe the AID and inflammasomopathies, and the current knowledge on the role of miRNAs in disease processes.
Collapse
Affiliation(s)
- Norma Saad
- Institute for Regenerative Medicine and Biotherapy, INSERM, U1183, University of Montpellier, Montpellier, France
| | - Isabelle Duroux-Richard
- Institute for Regenerative Medicine and Biotherapy, INSERM, U1183, University of Montpellier, Montpellier, France
| | - Isabelle Touitou
- Institute for Regenerative Medicine and Biotherapy, INSERM, U1183, University of Montpellier, Montpellier, France; Department of Molecular genetics, Medical Genetics of Rare and Autoinflammatory disease unit, Montpellier University Hospital, Montpellier, France; Centre de référence des maladies autoinflammatoires et des amyloses d'origine inflammatoire, CeRéMAIA, Montpellier University Hospital, Montpellier, France
| | - Eric Jeziorski
- Centre de référence des maladies autoinflammatoires et des amyloses d'origine inflammatoire, CeRéMAIA, Montpellier University Hospital, Montpellier, France; Department of Paediatric Emergency and Post-Emergency, Team of General Paediatrics, Infectious Diseases and Clinical Immunology, Montpellier University Hospital, Montpellier, France
| | - Florence Apparailly
- Institute for Regenerative Medicine and Biotherapy, INSERM, U1183, University of Montpellier, Montpellier, France; Clinical Department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier, France.
| |
Collapse
|
65
|
Lu S, Li Y, Qian Z, Zhao T, Feng Z, Weng X, Yu L. Role of the inflammasome in insulin resistance and type 2 diabetes mellitus. Front Immunol 2023; 14:1052756. [PMID: 36993972 PMCID: PMC10040598 DOI: 10.3389/fimmu.2023.1052756] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
The inflammasome is a protein complex composed of a variety of proteins in cells and which participates in the innate immune response of the body. It can be activated by upstream signal regulation and plays an important role in pyroptosis, apoptosis, inflammation, tumor regulation, etc. In recent years, the number of metabolic syndrome patients with insulin resistance (IR) has increased year by year, and the inflammasome is closely related to the occurrence and development of metabolic diseases. The inflammasome can directly or indirectly affect conduction of the insulin signaling pathway, involvement the occurrence of IR and type 2 diabetes mellitus (T2DM). Moreover, various therapeutic agents also work through the inflammasome to treat with diabetes. This review focuses on the role of inflammasome on IR and T2DM, pointing out the association and utility value. Briefly, we have discussed the main inflammasomes, including NLRP1, NLRP3, NLRC4, NLRP6 and AIM2, as well as their structure, activation and regulation in IR were described in detail. Finally, we discussed the current therapeutic options-associated with inflammasome for the treatment of T2DM. Specially, the NLRP3-related therapeutic agents and options are widely developed. In summary, this article reviews the role of and research progress on the inflammasome in IR and T2DM.
Collapse
Affiliation(s)
- Shen Lu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhaojun Qian
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tiesuo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaogang Weng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| | - Lili Yu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| |
Collapse
|
66
|
Li W, Lv X, Ma Y, Cai Y, Zhu S. Prognostic significance of serum NLRC4 in patients with acute supratentorial intracerebral hemorrhage: A prospective longitudinal cohort study. Front Neurol 2023; 14:1125674. [PMID: 36970543 PMCID: PMC10036848 DOI: 10.3389/fneur.2023.1125674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
ObjectiveCaspase activation and recruitment domain-containing protein 4 (NLRC4) is implicated in neuroinflammation. The aim of the study was to discern the potential ability of serum NLRC4 in assessment of prognosis after intracerebral hemorrhage (ICH).MethodsIn this prospective, observational study, serum NLRC4 levels were quantified in 148 acute supratentorial ICH patients and 148 controls. Severity was evaluated using the National Institutes of Health Stroke Scale (NIHSS) and hematoma volume, and poststroke 6-month functional outcome was estimated according to the modified Rankin Scale (mRS). Early neurologic deterioration (END) and 6-month poor outcome (mRS 3–6) were deemed as the two prognostic parameters. Multivariate models were established for investigating associations, and receiver operating characteristic (ROC) curves were configured to indicate predictive capability.ResultsPatients had substantially higher serum NLRC4 levels than controls (median, 363.2 pg/ml vs. 74.7 pg/ml). Serum NLRC4 levels had independent correlation with NIHSS scores [β, 0.308; 95% confidence interval (CI), 0.088–0.520], hematoma volume (β, 0.527; 95% CI, 0.385–0.675), serum C-reactive protein levels (β, 0.288; 95% CI, 0.109–0.341) and 6-month mRS scores (β, 0.239; 95% CI, 0.100–0.474). Serum NLRC4 levels above 363.2 pg/ml were independently predictive of END (odds ratio, 3.148; 95% CI, 1.278–7.752) and 6-month poor outcome (odds ratio, 2.468; 95% CI, 1.036–5.878). Serum NLRC4 levels significantly distinguished END risk [area under ROC curve (AUC), 0.765; 95% CI, 0.685–0.846] and 6-month poor outcome (AUC, 0.795; 95% CI, 0.721–0.870). In terms of predictive ability for 6-month poor outcome, serum NLRC4 levels combined with NIHSS scores and hematoma volume was superior to NIHSS scores combined with hematoma volume, NIHSS scores and hematoma volume (AUC, 0.913 vs. 0.870, 0.864 and 0.835; all P < 0.05). Nomograms were built to reflect prognosis and END risk of combination models, where serum NLRC4, NIHSS scores and hematoma volume were enforced. Calibration curves confirmed stability of combination models.ConclusionsMarkedly raised serum NLRC4 levels following ICH, in close relation to illness severity, are independently associated with poor prognosis. Such results are indicative of the notion that determination of serum NLRC4 may aid in severity assessment and prediction of functional outcome of ICH patients.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, First People's Hospital of Linping District, Hangzhou, China
- Department of Neurosurgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Lv
- Department of Neurosurgery, First People's Hospital of Linping District, Hangzhou, China
- Department of Neurosurgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yijun Ma
- Department of Neurosurgery, First People's Hospital of Linping District, Hangzhou, China
- Department of Neurosurgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Cai
- Department of Neurosurgery, First People's Hospital of Linping District, Hangzhou, China
- Department of Neurosurgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Suijun Zhu
- Department of Neurosurgery, First People's Hospital of Linping District, Hangzhou, China
- Department of Neurosurgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Suijun Zhu
| |
Collapse
|
67
|
Dong CC, Zhang QH, Zhang Y, Zhang Y, Ruan H, Qin T, Zhao JH, Wu G, Zhu Z, Yang JR. Comprehensive landscape of the IPAF inflammasomes in pan-cancer: A bulk omics research and single-cell sequencing validation. Comput Biol Med 2023; 155:106622. [PMID: 36780800 DOI: 10.1016/j.compbiomed.2023.106622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/28/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND IPAF (ICE-protease Activating Factor) is a nucleotide-binding/leucine-rich repeat (NLR) protein known as the cysteine-associated recruitment domain 12 (CARD12). Previous studies only discuss the role of IPAF inflammasomes in specific tumors. The role of IPAF inflammasomes in pan-cancer is still unclear. Therefore, we performed a comprehensive analysis of IPAF inflammasome in 33 tumors. METHODS We used databases like The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) from the UCSC XENA (http://xena.ucsc.edu/) to retrieve and analyze gene expression. The influence of IPAF inflammasome on the prognosis of tumor patients was analyzed using univariate Cox regression analysis and Kaplan-Meier survival analysis. Furthermore, we conducted the following analysis: Single-sample gene set enrichment analysis, single-cell level functional state analysis, single-cell sequencing, immune cell infiltration analysis, and tumor immune dysfunction and exclusion (TIDE) score. RESULTS First, the differential expression of IPAF inflammasome-related genes (IPAF-RGs) in 33 tumors were analyzed. The results revealed that IPAF-RGs were significantly and differentially expressed in eight tumors. The prognostic significance of IPAF inflammasome scores was different in different tumors. A positive correlation was observed between IPAF inflammasomes scores and CD8+ T cells in most tumors. Further analysis revealed that IPAF inflammasome might affect tumor immunity mainly by mediating effector T cell recruitment via the expression of chemokines such as CXCL9, CXCL10, and CCL5. The analysis of TIDE and IPAF inflammasome scores revealed a significant negative correlation between IPAF inflammasome and TIDE scores in 11 tumors. CONCLUSION A pan-cancer analysis of IPAF inflammasome in various tumors was performed. The results highlight the potential value of IPAF inflammasome in response to immunotherapy in patients and provide a new direction for future immunotherapy.
Collapse
Affiliation(s)
- Chen-Cheng Dong
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Qiu-Huan Zhang
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Yan Zhang
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Yujie Zhang
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Hanyi Ruan
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Tianyu Qin
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Jie-Hua Zhao
- Department of Breast and Thyroid Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Guo Wu
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Zhou Zhu
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China.
| | - Jian-Rong Yang
- Department of Breast and Thyroid Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China.
| |
Collapse
|
68
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
69
|
Sun P, Wang X, Zhong J, Yu D, Xuan H, Xu T, Song D, Yang C, Wang P, Liu Y, Meng X, Cai J. Development and validation of a pyroptosis-related genes signature for risk stratification in gliomas. Front Genet 2023; 14:1087563. [PMID: 36861130 PMCID: PMC9968976 DOI: 10.3389/fgene.2023.1087563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Glioma is a highly heterogeneous disease, causing the prognostic prediction a challenge. Pyroptosis, a programmed cell death mediated by gasdermin (GSDM), is characterized by cell swelling and the release of inflammatory factors. Pyroptosis occurs in several types of tumor cells, including gliomas. However, the value of pyroptosis-related genes (PRGs) in the prognosis of glioma remains to be further clarified. Methods: In this study, mRNA expression profiles and clinical data of glioma patients were acquired from TCGA and CGGA databases, and one hundred and eighteen PRGs were obtained from the Molecular Signatures Database and GeneCards. Then, consensus clustering analysis was performed to cluster glioma patients. The least absolute shrinkage and selection operator (LASSO) Cox regression model was used to establish a polygenic signature. Functional verification of the pyroptosis-related gene GSDMD was achieved by gene knockdown and western blotting. Moreover, the immune infiltration status between two different risk groups were analyzed through the "gsva" R package. Results: Our results demonstrated that the majority of PRGs (82.2%) were differentially expressed between lower-grade gliomas (LGG) and glioblastoma (GBM) in the TCGA cohort. In univariate Cox regression analysis, eighty-three PRGs were shown to be associated with overall survival (OS). A five-gene signature was constructed to divide patients into two risk groups. Compared with patients in the low-risk group, patients in the high-risk group had obviously shorter OS (p < 0.001). Also, we found that the high-risk group showed a higher infiltrating score of immune cells and immune-related functions. Risk score was an independent predictor of OS (HR > 1, p < 0.001). Furthermore, knockdown of GSDMD decreased the expression of IL-1β and cleaved caspase-1. Conclusion: Our study constructed a new PRGs signature, which can be used to predict the prognosis of glioma patients. Targeting pyroptosis might serve as a potential therapeutic strategy for glioma.
Collapse
Affiliation(s)
| | | | - Junzhe Zhong
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Daohan Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanwen Xuan
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianye Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changxiao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pandeng Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuxiang Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Jinquan Cai
- *Correspondence: Jinquan Cai, ; Xiangqi Meng,
| |
Collapse
|
70
|
Zhou J, Qiu J, Song Y, Liang T, Liu S, Ren C, Song X, Cui L, Sun Y. Pyroptosis and degenerative diseases of the elderly. Cell Death Dis 2023; 14:94. [PMID: 36755014 PMCID: PMC9908978 DOI: 10.1038/s41419-023-05634-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
Pyroptosis is a recently described mechanism of programmed cell death mediated by proteins of the gasdermin family. Widely recognized signaling cascades include the classical, non-classical, caspase-3-dependent gasdermin E and caspase-8-dependent gasdermin D pathways. Additional pyroptotic pathways have been subsequently reported. With the rising prevalence of advanced age, the role of pyroptosis in the degenerative diseases of the elderly has attracted increased research attention. This article reviews the primary mechanisms of pyroptosis and summarizes progress in the research of degenerative diseases of the elderly such as presbycusis, age-related macular degeneration, Alzheimer's disease, intervertebral disc degeneration, and osteoarthritis.
Collapse
Affiliation(s)
- Jiamin Zhou
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Jingjing Qiu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Yuwan Song
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Tiantian Liang
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Sha Liu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Chao Ren
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China
| | - Limei Cui
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China.
| | - Yan Sun
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, PR China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, PR China.
| |
Collapse
|
71
|
Paidimuddala B, Cao J, Nash G, Xie Q, Wu H, Zhang L. Mechanism of NAIP-NLRC4 inflammasome activation revealed by cryo-EM structure of unliganded NAIP5. Nat Struct Mol Biol 2023; 30:159-166. [PMID: 36604500 PMCID: PMC10576962 DOI: 10.1038/s41594-022-00889-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 11/03/2022] [Indexed: 01/07/2023]
Abstract
The nucleotide-binding domain (NBD), leucine rich repeat (LRR) domain containing protein family (NLR family) apoptosis inhibitory proteins (NAIPs) are cytosolic receptors that play critical roles in the host defense against bacterial infection. NAIPs interact with conserved bacterial ligands and activate the NLR family caspase recruitment domain containing protein 4 (NLRC4) to initiate the NAIP-NLRC4 inflammasome pathway. Here we found the process of NAIP activation is completely different from NLRC4. Our cryo-EM structure of unliganded mouse NAIP5 adopts an unprecedented wide-open conformation, with the nucleating surface fully exposed and accessible to recruit inactive NLRC4. Upon ligand binding, the winged helix domain (WHD) of NAIP5 undergoes roughly 20° rotation to form a steric clash with the inactive NLRC4, which triggers the conformational change of NLRC4 from inactive to active state. We also show the rotation of WHD places the 17-18 loop at a position that directly bind the active NLRC4 and stabilize the NAIP5-NLRC4 complex. Overall, these data provide structural mechanisms of inactive NAIP5, the process of NAIP5 activation and NAIP-dependent NLRC4 activation.
Collapse
Affiliation(s)
- Bhaskar Paidimuddala
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jianhao Cao
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Grady Nash
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Qing Xie
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Liman Zhang
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University (OHSU), Portland, OR, USA.
| |
Collapse
|
72
|
Benameur T, Frota Gaban SV, Giacomucci G, Filannino FM, Trotta T, Polito R, Messina G, Porro C, Panaro MA. The Effects of Curcumin on Inflammasome: Latest Update. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020742. [PMID: 36677800 PMCID: PMC9866118 DOI: 10.3390/molecules28020742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Curcumin, a traditional Chinese medicine extracted from natural plant rhizomes, has become a candidate drug for the treatment of different diseases due to its anti-inflammatory, anticancer, antioxidant, and antibacterial activities. Curcumin is generally beneficial to improve human health with anti-inflammatory and antioxidative properties as well as antitumor and immunoregulatory properties. Inflammasomes are NLR family, pyrin domain-containing 3 (NLRP3) proteins that are activated in response to a variety of stress signals and that promote the proteolytic conversion of pro-interleukin-1β and pro-interleukin-18 into active forms, which are central mediators of the inflammatory response; inflammasomes can also induce pyroptosis, a type of cell death. The NLRP3 protein is involved in a variety of inflammatory pathologies, including neurological and autoimmune disorders, lung diseases, atherosclerosis, myocardial infarction, and many others. Different functional foods may have preventive and therapeutic effects in a wide range of pathologies in which inflammasome proteins are activated. In this review, we have focused on curcumin and evidenced its therapeutic potential in inflammatory diseases such as neurodegenerative diseases, respiratory diseases, and arthritis by acting on the inflammasome.
Collapse
Affiliation(s)
- Tarek Benameur
- College of Medicine, Department of Biomedical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Socorro Vanesca Frota Gaban
- Department of Food Engineering, Federal University of Ceara, Campus do Pici., Fortaleza CEP60356-000, Brazil
| | - Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy
| | | | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
- Correspondence:
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| |
Collapse
|
73
|
Patil P, Doshi G. Deciphering the Role of Pyroptosis Impact on Cardiovascular Diseases. Curr Drug Targets 2023; 24:1166-1183. [PMID: 38164730 DOI: 10.2174/0113894501267496231102114410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 01/03/2024]
Abstract
Pyroptosis has become a noteworthy area of focus in recent years due to its association with inflammatory diseases. Pyroptosis is a type of programmed cell death accompanied by an inflammatory response, and the discovery of the gasdermin family has expanded the study of pyroptosis. The primary characteristics of pyroptosis include cell expansion, membrane penetration, and the ejection of cell contents. In healthy physiology, pyroptosis is an essential part of the host's defence against pathogen infection. Excessive Pyroptosis, however, can lead to unchecked and persistent inflammatory responses, including the emergence of inflammatory diseases. More precisely, gasdermin family members have a role in the creation of membrane holes during pyroptosis, which leads to cell lysis. It is also related to how pro-inflammatory intracellular substances, including IL-1, IL-18, and High mobility group box 1 (HMGB1), are used. Two different signalling pathways, one of which is regulated by caspase-1 and the other by caspase-4/5/11, are the primary causes of pyroptosis. Cardiovascular diseases are often associated with cell death and acute or chronic inflammation, making this area of research particularly relevant. In this review, we first systematically summarize recent findings related to Pyroptosis, exploring its characteristics and the signalling pathway mechanisms, as well as various treatment strategies based on its modulation that has emerged from the studies. Some of these strategies are currently undergoing clinical trials. Additionally, the article elaborates on the scientific evidence indicating the role of Pyroptosis in various cardiovascular diseases. As a whole, this should shed insight into future paths and present innovative ideas for employing Pyroptosis as a strong disease-fighting weapon.
Collapse
Affiliation(s)
- Poonam Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VLM Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VLM Road, Vile Parle (w), Mumbai, 400056, India
| |
Collapse
|
74
|
Chang MX. Emerging mechanisms and functions of inflammasome complexes in teleost fish. Front Immunol 2023; 14:1065181. [PMID: 36875130 PMCID: PMC9978379 DOI: 10.3389/fimmu.2023.1065181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Inflammasomes are multiprotein complexes, which are assembled in response to a diverse range of exogenous pathogens and endogenous danger signals, leading to produce pro-inflammatory cytokines and induce pyroptotic cell death. Inflammasome components have been identified in teleost fish. Previous reviews have highlighted the conservation of inflammasome components in evolution, inflammasome function in zebrafish infectious and non-infectious models, and the mechanism that induce pyroptosis in fish. The activation of inflammasome involves the canonical and noncanonical pathways, which can play critical roles in the control of various inflammatory and metabolic diseases. The canonical inflammasomes activate caspase-1, and their signaling is initiated by cytosolic pattern recognition receptors. However the noncanonical inflammasomes activate inflammatory caspase upon sensing of cytosolic lipopolysaccharide from Gram-negative bacteria. In this review, we summarize the mechanisms of activation of canonical and noncanonical inflammasomes in teleost fish, with a particular focus on inflammasome complexes in response to bacterial infection. Furthermore, the functions of inflammasome-associated effectors, specific regulatory mechanisms of teleost inflammasomes and functional roles of inflammasomes in innate immune responses are also reviewed. The knowledge of inflammasome activation and pathogen clearance in teleost fish will shed new light on new molecular targets for treatment of inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of InSciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
75
|
Tang B, Zhong Z, Wu J, Ma J, Li L, Zhong X, Lin D, Hu J, Yu P. Evaluation of Serum NLRC4 as a Potential Prognostic Biochemical Marker in Humans with Severe Traumatic Brain Injury: A Prospective Cohort Study. Risk Manag Healthc Policy 2023; 16:439-454. [PMID: 36994425 PMCID: PMC10042254 DOI: 10.2147/rmhp.s404877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Objective Involvement of NLR CARD domain containing 4 (NLRC4) in neuroinflammation has been demonstrated. The aim of this study was to ascertain the prognostic role of serum NLRC4 in severe traumatic brain injury (sTBI). Methods In this prospective cohort study including 140 sTBI patients and 140 controls, serum NLRC4 levels were quantified. Follow-up time was 180 days after trauma and poor prognosis was designated as extended Glasgow outcome scale (GOSE) scores of 1-4. Severity correlations and prognosis associations were determined under multivariate models. Results Enhanced serum NLRC4 levels after sTBI, in comparison to controls (median, 0.8 ng/mL versus 0.1 ng/mL; P < 0.001), were independently correlated with Glasgow coma scale (GCS) scores (β, -0.091; 95% confidence interval (CI), -0.161-0.021; P = 0.011), Rotterdam computed tomography (CT) scores (β, 0.136; 95% CI, 0.024-0.248; P = 0.018), serum C-reactive protein levels (β, 0.016; 95% CI, 0.002-0.030; P = 0.025) and 180-day GOSE scores (β, -0.906; 95% CI, -1.632-0.180; P = 0.015); and were independently predictive of 180-day death (odds ratio, 4.307; 95% CI, 1.706-10.879; P = 0.014)), overall survival (hazard ratio, 2.360; 95% CI, 1.118-4.981; P = 0.040) and poor prognosis (odds ratio, 6.705; 95% CI, 2.889-15.561; P = 0.016). Under receiver operating characteristic curve, combination of serum NLRC4 levels, GCS scores and Rotterdam CT scores had significantly higher death predictive ability than Rotterdam CT scores (P = 0.040), but not than GCS scores (P = 0.070); and exhibited substantially higher predictive capability for poor prognosis than Rotterdam CT scores (P < 0.001) and GCS scores alone (P = 0.023). Conclusion There is a dramatical elevation of serum NLRC4 levels after sTBI, which has strong correlation with severity and inflammation, and is significantly associated with long-term death and poor outcome, substantializing serum NLRC4 as an inflammatory, prognostic biomarker in sTBI.
Collapse
Affiliation(s)
- Bei Tang
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Ze Zhong
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
- Correspondence: Ze Zhong, Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China, Tel/Fax +86 571 64096607, Email
| | - Jinping Wu
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Jianping Ma
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Li Li
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Xuzheng Zhong
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Dongmei Lin
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Jiayuan Hu
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| | - Pingan Yu
- Department of Critical Care Medicine, The First People’s Hospital of Jiande City, Jiande, People’s Republic of China
| |
Collapse
|
76
|
Kan W, Li Q, Li P, Ren L, Mu W, Lin L, Wen J, Ge F, Hou M, Hui S, He P, Liang L, Xu Y, Li X, Xu G, Xiao X, Bai Z. Glycyrrhiza uralensis polysaccharides ameliorate acute lung injury by inhibiting the activation of multiple inflammasomes. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
77
|
Anderson MJ, den Hartigh AB, Fink SL. Molecular Mechanisms of Pyroptosis. Methods Mol Biol 2023; 2641:1-16. [PMID: 37074637 DOI: 10.1007/978-1-0716-3040-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Pyroptosis is a regulated form of cell death that leads to inflammation and plays a role in many different diseases. Pyroptosis was initially defined by the dependence on caspase-1, a protease which is activated by innate immune signaling complexes called inflammasomes. Caspase-1 cleaves the protein gasdermin D, releasing the N-terminal pore-forming domain, which inserts into the plasma membrane. Recent studies have revealed that other gasdermin family members form plasma membrane pores, leading to lytic cell death, and the definition of pyroptosis was revised to gasdermin-dependent cell death. In this review, we discuss how the use of the term pyroptosis has changed over time, as well as currently understood molecular mechanisms leading to pyroptosis and functional consequences of this form of regulated cell death.
Collapse
Affiliation(s)
- Marisa J Anderson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Andreas B den Hartigh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
78
|
Planas R, Felber M, Vavassori S, Pachlopnik Schmid J. The hyperinflammatory spectrum: from defects in cytotoxicity to cytokine control. Front Immunol 2023; 14:1163316. [PMID: 37187762 PMCID: PMC10175623 DOI: 10.3389/fimmu.2023.1163316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Cytotoxic lymphocytes kill target cells through polarized release of the content of cytotoxic granules towards the target cell. The importance of this cytotoxic pathway in immune regulation is evidenced by the severe and often fatal condition, known as hemophagocytic lymphohistiocytosis (HLH) that occurs in mice and humans with inborn errors of lymphocyte cytotoxic function. The clinical and preclinical data indicate that the damage seen in severe, virally triggered HLH is due to an overwhelming immune system reaction and not the direct effects of the virus per se. The main HLH-disease mechanism, which links impaired cytotoxicity to excessive release of pro-inflammatory cytokines is a prolongation of the synapse time between the cytotoxic effector cell and the target cell, which prompts the former to secrete larger amounts of cytokines (including interferon gamma) that activate macrophages. We and others have identified novel genetic HLH spectrum disorders. In the present update, we position these newly reported molecular causes, including CD48-haploinsufficiency and ZNFX1-deficiency, within the pathogenic pathways that lead to HLH. These genetic defects have consequences on the cellular level on a gradient model ranging from impaired lymphocyte cytotoxicity to intrinsic activation of macrophages and virally infected cells. Altogether, it is clear that target cells and macrophages may play an independent role and are not passive bystanders in the pathogenesis of HLH. Understanding these processes which lead to immune dysregulation may pave the way to novel ideas for medical intervention in HLH and virally triggered hypercytokinemia.
Collapse
Affiliation(s)
- Raquel Planas
- Division of Immunology, University Children’s Hospital Zurich, Zurich, Switzerland
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Matthias Felber
- Division of Immunology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Stefano Vavassori
- Division of Immunology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children’s Hospital Zurich, Zurich, Switzerland
- Pediatric Immunology, University of Zurich, Zurich, Switzerland
- *Correspondence: Jana Pachlopnik Schmid,
| |
Collapse
|
79
|
Paerewijck O, Lamkanfi M. The human inflammasomes. Mol Aspects Med 2022; 88:101100. [PMID: 35696786 DOI: 10.1016/j.mam.2022.101100] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/25/2022] [Accepted: 06/01/2022] [Indexed: 12/14/2022]
Abstract
Two decades of inflammasome research has led to a vast body of knowledge on the complex regulatory mechanisms and pathological roles of canonical and non-canonical inflammasome activation in a plethora of research models of primarily rodent origin. More recently, the field has made notable progress in characterizing human-specific inflammasomes and their regulation mechanisms, including an expansion of inflammasome biology to adaptive immune cells. These exciting developments in basic research have been accompanied by potentially transformative results from large clinical trials and translational efforts to develop inflammasome-targeted small molecule inhibitors for therapeutic use. Here, we will discuss recent findings in the field with a specific emphasis on activation mechanisms of human inflammasomes and their potential role in auto-inflammatory, metabolic and neoplastic diseases.
Collapse
Affiliation(s)
- Oonagh Paerewijck
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, B-9000, Belgium
| | - Mohamed Lamkanfi
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, B-9000, Belgium.
| |
Collapse
|
80
|
Chuphal B, Rai U, Roy B. Teleost NOD-like receptors and their downstream signaling pathways: A brief review. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2022; 3:100056. [DOI: 10.1016/j.fsirep.2022.100056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 02/08/2023] Open
|
81
|
Zhang RN, Sun ZJ, Zhang L. Pyroptosis in inflammatory bone diseases: Molecular insights and targeting strategies. FASEB J 2022; 36:e22670. [PMID: 36412502 DOI: 10.1096/fj.202201229r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Inflammatory bone diseases include osteoarthritis (OA) and rheumatoid arthritis (RA), which can cause severe bone damage in a chronic inflammation state, putting tremendous pressure on the patients' families and government agencies regarding medical costs. In addition, the complexity of osteoimmunology makes research on these diseases difficult. Hence, it is urgent to determine the potential mechanisms and find effective drugs to target inflammatory bone diseases to reduce the negative effects of these diseases. Recently, pyroptosis, a gasdermin-induced necrotic cell death featuring secretion of pro-inflammatory cytokines and lysis, has become widely known. Based on the effect of pyroptosis on immunity, this process has gradually emerged as a vital component in the etiopathogenesis of inflammatory bone diseases. Herein, we review the characteristics and mechanisms of pyroptosis and then focus on its clinical significance in inflammatory bone diseases. In addition, we summarize the current research progress of drugs targeting pyroptosis to enhance the therapeutic efficacy of inflammatory bone diseases and provide new insights for future directions.
Collapse
Affiliation(s)
- Ruo-Nan Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
82
|
Liu Y, Guo ZW, Li J, Li AH, Huo TG. Insight into the regulation of NLRP3 inflammasome activation by mitochondria in liver injury and the protective role of natural products. Biomed Pharmacother 2022; 156:113968. [DOI: 10.1016/j.biopha.2022.113968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|
83
|
Liang Y, Gu T, Peng S, Lin Y, Liu J, Wang X, Huang X, Zhang X, Zhu J, Zhao L, Fan C, Wang G, Gu X, Lin J. p16 INK4a Plays Critical Role in Exacerbating Inflammaging in High Fat Diet Induced Skin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3415528. [PMID: 36457728 PMCID: PMC9706253 DOI: 10.1155/2022/3415528] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 09/02/2023]
Abstract
BACKGROUND Long term high fat diets (HFD) promote skin aging pathogenesis, but detailed mechanisms remain unclear especially for inflammaging, which has recently emerged as a pathway correlating aging and age-related disease with inflammation. p16INK4a (hereafter termed p16) inhibits the cell cycle, with p16 deletion significantly inhibiting inflammaging. We observed that HFD-induced p16 overexpression in the skin. Therefore, we investigated if p16 exacerbated inflammaging in HFD-induced skin and also if p16 deletion exerted protective effects against this process. METHODS Eight-week-old double knockout (KO) ApoE-/-p16-/- mice and ApoE-/- littermates were fed HFD for 12 weeks and their skin phenotypes were analyzed. We measured skin fibrosis, senescence-associated secretory phenotype (SASP) levels, and integrin-inflammasome pathway activation using histopathological, RNA-sequencing (RNA-seq), bioinformatics analysis, and molecular techniques. RESULTS We found that HFD contributed to inflammaging in the skin by activating the NLRP3 inflammasome pathway, increasing inflammatory infiltration, and promoting apoptosis by balancing expression between proapoptotic and antiapoptotic molecules. p16 knockout, when compared with the ApoE-/- phenotype, inhibited skin fibrosis by ameliorating inflammatory infiltration and proinflammatory factor expression (Interleukin-1β (IL-1β), Interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)), and also alleviated inflammaging skin progress induced by HFD in the ApoE-/- mouse model. RNA-seq showed that p16 KO mice inhibited both integrin-inflammasome and NF-κB proinflammatory pathway activation. CONCLUSIONS p16 deletion or p16 positive cell clearance could be a novel strategy preventing long term HFD-induced skin aging.
Collapse
Affiliation(s)
- Yan Liang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tianya Gu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Su Peng
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yi Lin
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - JiaBao Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Xin Huang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Jun Zhu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Lin Zhao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Changyan Fan
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Guangyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Xin Gu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - JinDe Lin
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
84
|
[NLRC4 plays a regulatory role in F. nucleatum-induced pyroptosis in macrophages]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1560-1565. [PMID: 36329592 PMCID: PMC9637494 DOI: 10.12122/j.issn.1673-4254.2022.10.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To explore the mechanism of F.nucleatum-induced pyroptosis in macrophages and the regulatory role of inflammasomes. METHODS Lactate dehydrogenase (LDH) cytotoxicity assay and Hoechst 33342/PI double fluorescence staining were used to analyze cytolysis in F.nucleatum-infected macrophage RAW264.7 cells.The expressions of pyroptosis-related proteins caspase-1, GSDMD and IL-1β were determined using Western blotting.Inflammasome activation in the cells was analyzed by detecting the mRNA expressions of NLRP3, NLRC4, AIM2, and NLRP1 with qRT-PCR.RNA interference technique was used to knock down the key molecules involved in pyroptosis regulation in the macrophages, and the pyroptosis and necrosis rates of the cells following F.nucleatum infection were examined. RESULTS The results of LDH cytotoxicity assay and double-fluorescence staining showed that F.nucleatum infection caused swelling and lytic cell death in RAW264.7 cells.F.nucleatum infection resulted in the activation of caspase-1 and GSDMD and upregulated IL-1β expression in a multiplicity of infection (MOI)-and time-dependent manner (P < 0.05).qRT-PCR revealed significantly increased expression of NLRC4 mRNA in the macrophages after F.nucleatum infection (P < 0.05).NLRC4 silencing by siRNA strongly inhibited the activation of caspase-1/GSDMD pathway and reduced cell death (P < 0.05) and IL-1β expression in F.nucleatum-infected cells. CONCLUSION NLRC4 inflammasome drives caspase-1/GSDMD-mediated pyroptosis and inflammatory signaling in F.nucleatum-infected macrophages, suggesting the potential of NLRC4 inflammasome as a therapeutic target for F.nucleatum infections.
Collapse
|
85
|
Chebly H, Marvaud JC, Safa L, Elkak AK, Kobeissy PH, Kansau I, Larrazet C. Clostridioides difficile Flagellin Activates the Intracellular NLRC4 Inflammasome. Int J Mol Sci 2022; 23:ijms232012366. [PMID: 36293218 PMCID: PMC9604438 DOI: 10.3390/ijms232012366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Clostridioides difficile (C. difficile), is a major cause of nosocomial diarrhea and colitis. C. difficile flagellin FliC contributes toxins to gut inflammation by interacting with the immune Toll-like receptor 5 (TLR5) to activate nuclear factor-kappa B (NF-kB) and mitogen-activated protein kinase (MAPK) signaling pathways. Flagella of intracellular pathogens can activate the NLR family CARD domain-containing protein 4 (NLRC4) inflammasome pathway. In this study, we assessed whether flagellin of the extracellular bacterium C. difficile internalizes into epithelial cells and activates the NLRC4 inflammasome. Confocal microscopy showed internalization of recombinant green fluorescent protein (GFP)-FliC into intestinal Caco-2/TC7 cell line. Full-length GFP-FliC activates NLRC4 in Caco-2/TC7 cells in contrast to truncated GFP-FliC lacking the C-terminal region recognized by the inflammasome. FliC induced cleavage of pro-caspase-1 into two subunits, p20 and p10 as well as gasdermin D (GSDMD), suggesting the caspase-1 and NLRC4 inflammasome activation. In addition, colocalization of GFP-FliC and pro-caspase-1 was observed, indicating the FliC-dependent NLRC4 inflammasome activation. Overexpression of the inflammasome-related interleukin (interleukin (IL)-1β, IL-18, and IL-33) encoding genes as well as increasing of the IL-18 synthesis was detected after cell stimulation. Inhibition of I-kappa-B kinase alpha (IKK-α) decreased the FliC-dependent inflammasome interleukin gene expression suggesting a role of the NF-κB pathway in regulating inflammasome. Altogether, these results suggest that FliC internalizes into the Caco-2/TC7 cells and activates the intracellular NLRC4 inflammasome thus contributing to the inflammatory process of C. difficile infection.
Collapse
Affiliation(s)
- Hiba Chebly
- Institut Micalis, Université Paris-Saclay, INRAE, AgroParisTech, 91400 Orsay, France
- Health Resources and Products Valorization Laboratory, Faculty of Pharmacy, Lebanese University, Beirut 1102-2801, Lebanon
| | | | - Layale Safa
- Health Resources and Products Valorization Laboratory, Faculty of Pharmacy, Lebanese University, Beirut 1102-2801, Lebanon
| | - Assem Khalil Elkak
- Health Resources and Products Valorization Laboratory, Faculty of Pharmacy, Lebanese University, Beirut 1102-2801, Lebanon
| | - Philippe Hussein Kobeissy
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Imad Kansau
- Institut Micalis, Université Paris-Saclay, INRAE, AgroParisTech, 91400 Orsay, France
| | - Cécile Larrazet
- Institut Micalis, Université Paris-Saclay, INRAE, AgroParisTech, 91400 Orsay, France
- Correspondence:
| |
Collapse
|
86
|
Somm E, Jornayvaz FR. Interleukin-18 in metabolism: From mice physiology to human diseases. Front Endocrinol (Lausanne) 2022; 13:971745. [PMID: 36313762 PMCID: PMC9596921 DOI: 10.3389/fendo.2022.971745] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin-18 (IL-18) is a classical member of the IL-1 superfamily of cytokines. As IL-1β, IL-18 precursor is processed by inflammasome/caspase-1 into a mature and biologically active form. IL-18 binds to its specific receptor composed of two chains (IL-18Rα and IL-18Rβ) to trigger a similar intracellular signaling pathway as IL-1, ultimately leading to activation of NF-κB and inflammatory processes. Independently of this IL-1-like signaling, IL-18 also specifically induces IFN-γ production, driving the Th1 immune response. In circulation, IL-18 binds to the IL-18 binding protein (IL-18BP) with high affinity, letting only a small fraction of free IL-18 able to trigger receptor-mediated signaling. In contrast to other IL-1 family members, IL-18 is produced constitutively by different cell types, suggesting implications in normal physiology. If the roles of IL-18 in inflammatory processes and infectious diseases are well described, recent experimental studies in mice have highlighted the action of IL-18 signaling in the control of energy homeostasis, pancreatic islet immunity and liver integrity during nutritional stress. At the same time, clinical observations implicate IL-18 in various metabolic diseases including obesity, type 1 and 2 diabetes and nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). In the present review, we summarize and discuss both the physiological actions of IL-18 in metabolism and its potential roles in pathophysiological mechanisms leading to the most common human metabolic disorders, such as obesity, diabetes and NAFLD/NASH.
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - François R. Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
87
|
Wan J, Liu D, Pan S, Zhou S, Liu Z. NLRP3-mediated pyroptosis in diabetic nephropathy. Front Pharmacol 2022; 13:998574. [PMID: 36304156 PMCID: PMC9593054 DOI: 10.3389/fphar.2022.998574] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic nephropathy (DN) is the main cause of end-stage renal disease (ESRD), which is characterized by a series of abnormal changes such as glomerulosclerosis, podocyte loss, renal tubular atrophy and excessive deposition of extracellular matrix. Simultaneously, the occurrence of inflammatory reaction can promote the aggravation of DN-induced kidney injury. The most important processes in the canonical inflammasome pathway are inflammasome activation and membrane pore formation mediated by gasdermin family. Converging studies shows that pyroptosis can occur in renal intrinsic cells and participate in the development of DN, and its activation mechanism involves a variety of signaling pathways. Meanwhile, the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome can not only lead to the occurrence of inflammatory response, but also induce pyroptosis. In addition, a number of drugs targeting pyroptosis-associated proteins have been shown to have potential for treating DN. Consequently, the pathogenesis of pyroptosis and several possible activation pathways of NLRP3 inflammasome were reviewed, and the potential drugs used to treat pyroptosis in DN were summarized in this review. Although relevant studies are still not thorough and comprehensive, these findings still have certain reference value for the understanding, treatment and prognosis of DN.
Collapse
Affiliation(s)
- Jiayi Wan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Sijie Zhou
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- *Correspondence: Sijie Zhou, ; Zhangsuo Liu,
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- *Correspondence: Sijie Zhou, ; Zhangsuo Liu,
| |
Collapse
|
88
|
Wang L, Zhang J, Zhang L, Hu L, Tian J. Significant difference of differential expression pyroptosis-related genes and their correlations with infiltrated immune cells in sepsis. Front Cell Infect Microbiol 2022; 12:1005392. [PMID: 36250055 PMCID: PMC9556990 DOI: 10.3389/fcimb.2022.1005392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundSepsis is regarded as a life-threatening organ dysfunction syndrome that responds to infection. Pyroptosis, a unique form of programmed cell death, is characterized by inflammatory cytokine secretion. Recently, an increasing number of studies have investigated the relationship between sepsis and pyroptosis. Appropriate pyroptosis can help to control infection during sepsis, but an immoderate one may cause immune disorders. The present study aimed to identify pyroptosis-related gene biomarkers and their relationship with the immune microenvironment using the genome-wide technique.MethodsThe training dataset GSE154918 and the validation dataset GSE185263 were downloaded for bioinformatics analysis. Differentially expressed pyroptosis-related genes (DEPRGs) were identified between sepsis (including septic shock) and healthy samples. Gene Set Enrichment Analysis (GSEA) was performed to explore gene function. CIBERSORT tools were applied to quantify infiltrating immune cells, and the correlation between differentially infiltrating immune cells and DEPRG expression was investigated. Furthermore, based on multivariable Cox regression, the study also utilized a random forest (RF) model to screen biomarkers.ResultsIn total, 12 DEPRGs were identified. The expression level of PLCG1 was continuously significantly decreased, while the expression level of NLRC4 was elevated from control to sepsis and then to septic shock. GSEA found that one DEPRG (PLCG1) was involved in the T-cell receptor signaling pathway and that many T cell-related immunologic signature gene sets were enriched. The proportions of plasma cells, T cells CD4 memory activated, and some innate cells in the sepsis group were significantly higher than those in the healthy group, while the proportions of T cells CD8, T cells CD4 memory resting, T cells regulatory (Tregs), and NK cells were lower. Additionally, CASP4 was positively correlated with Neutrophils and negatively correlated with T cells CD4 memory resting and Tregs. Lastly, two biomarkers (CASP4 and PLCG1) were identified, and a nomogram model was constructed for diagnosis with area under the curve (AUC) values of 0.998.ConclusionThis study identified two potential pyroptosis-related diagnostic genes, CASP4 and PLCG1, and explored the correlation between DEPRGs and the immune microenvironment. Also, our study indicated that some DEPRGs were satisfactorily correlated with several representative immune cells that can regulate pyroptosis.
Collapse
Affiliation(s)
- Li Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning Commission, Shanghai, China
- *Correspondence: Li Wang, ; Lingli Hu, ; Jianhui Tian,
| | - Jiting Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Zhang
- Huashan Hospital, Fudan University, Shanghai, China
| | - Lingli Hu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Li Wang, ; Lingli Hu, ; Jianhui Tian,
| | - Jianhui Tian
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Li Wang, ; Lingli Hu, ; Jianhui Tian,
| |
Collapse
|
89
|
Puleo MG, Miceli S, Di Chiara T, Pizzo GM, Della Corte V, Simonetta I, Pinto A, Tuttolomondo A. Molecular Mechanisms of Inflammasome in Ischemic Stroke Pathogenesis. Pharmaceuticals (Basel) 2022; 15:1168. [PMID: 36297283 PMCID: PMC9612213 DOI: 10.3390/ph15101168] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke (also called cerebral ischemia) is one of the leading causes of death and severe disability worldwide. NLR inflammasomes play a crucial role in sensing cell damage in response to a harmful stimuli and modulating the inflammatory response, promoting the release of pro-inflammatory cytokines such as IL-18 and IL-1β following ischemic injury. Therefore, a neuroprotective effect is achieved by inhibiting the expression, assembly, and secretion of inflammasomes, thus limiting the extent of brain detriment and neurological sequelae. This review aims to illustrate the molecular characteristics, expression levels, and assembly of NLRP3 (nucleotide-binding oligomerization domain-like receptor [NLR] family pyrin-domain-containing 3) inflammasome, the most studied in the literature, in order to discover promising therapeutic implications. In addition, we provide some information regarding the contribution of NLRP1, NLRP2, and NLRC4 inflammasomes to ischemic stroke pathogenesis, highlighting potential therapeutic strategies that require further study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Antonino Tuttolomondo
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties, “G. D’Alessandro”, University of Palermo, Piazza delle Cliniche n.2, 90127 Palermo, Italy
| |
Collapse
|
90
|
Pan Y, Cai W, Huang J, Cheng A, Wang M, Yin Z, Jia R. Pyroptosis in development, inflammation and disease. Front Immunol 2022; 13:991044. [PMID: 36189207 PMCID: PMC9522910 DOI: 10.3389/fimmu.2022.991044] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022] Open
Abstract
In the early 2000s, caspase-1, an important molecule that has been shown to be involved in the regulation of inflammation, cell survival and diseases, was given a new function: regulating a new mode of cell death that was later defined as pyroptosis. Since then, the inflammasome, the inflammatory caspases (caspase-4/5/11) and their substrate gasdermins (gasdermin A, B, C, D, E and DFNB59) has also been reported to be involved in the pyroptotic pathway, and this pathway is closely related to the development of various diseases. In addition, important apoptotic effectors caspase-3/8 and granzymes have also been reported to b involved in the induction of pyroptosis. In our article, we summarize findings that help define the roles of inflammasomes, inflammatory caspases, gasdermins, and other mediators of pyroptosis, and how they determine cell fate and regulate disease progression.
Collapse
Affiliation(s)
- Yuhong Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Wenjun Cai
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- *Correspondence: Anchun Cheng, ; Renyong Jia,
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- *Correspondence: Anchun Cheng, ; Renyong Jia,
| |
Collapse
|
91
|
Chen X, Zhang Z, Sun N, Li J, Ma Z, Rao Z, Sun X, Zeng Q, Wu Y, Li J, Zhang J, Chen Y. Vitamin D receptor enhances
NLRC4
inflammasome activation by promoting
NAIPs–NLRC4
association. EMBO Rep 2022; 23:e54611. [DOI: 10.15252/embr.202254611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Xin Chen
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Zaikui Zhang
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Naishuang Sun
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Jinzhou Li
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Zemeng Ma
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Zebing Rao
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Xiaomeng Sun
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Qiang Zeng
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Yuxuan Wu
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
| | - Jiahuang Li
- School of Biopharmacy China Pharmaceutical University Nanjing China
| | - Jing Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing China
| | - Yunzi Chen
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology Nanjing Medical University Nanjing China
- Medical Centre for Digestive Diseases Second Affiliated Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
92
|
Zhou F, Zhang G, Wu Y, Xiong Y. Inflammasome Complexes: Crucial mediators in osteoimmunology and bone diseases. Int Immunopharmacol 2022; 110:109072. [DOI: 10.1016/j.intimp.2022.109072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/15/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022]
|
93
|
Wang N, Chu F, Zhang L, Fei C, Yu C, Xue S, Wang Y, Fang L, Peng D, Duan X, Chen W. Taohong siwu decoction attenuates AIM2 and NLRC4 inflammasomes by ameliorates deoxyribonucleic acid damage after ischemic stroke. Front Pharmacol 2022; 13:954867. [PMID: 36034843 PMCID: PMC9411787 DOI: 10.3389/fphar.2022.954867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Taohong siwu decoction (THSWD) has been shown to have a therapeutic effect on ischemic strokes (IS). However, it is not clear to us whether THSWD reduces deoxyribonucleic acid (DNA) damage after stroke and reduces the inflammatory response caused by the damage. Therefore, we constructed an IS model (I/R) in rats and performed oxygen-glucose deprivation/reoxygenation (OGD/R) on BV2 cells. Then ELISA, immunofluorescence staining, immunohistochemistry staining, and RT-qPCR were performed to detect the expressions of absent in melanoma 2 (AIM2), NLRC4, and Caspase-1 inflammasomes and other inflammatory factors. Experimental stroke causes DNA damage, and we found that the aforementioned inflammasomes as well as inflammatory factors were significantly inhibited after treatment with THSWD by comparing the model group with the model administration group. In addition, we examined the expression of AIM2, NLRC4, and Caspase-1 in BV2 cells of OGD/R and found that the expression of the aforementioned inflammasomes was significantly decreased in OGD/R by administration of THSWD-containing serum. Our data suggest that THSWD can reduced DNA damage after stroke as well as the inflammatory response caused by the damage.
Collapse
Affiliation(s)
- Ni Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Furui Chu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Lijuan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Changyi Fei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Chao Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Sujun Xue
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yongzhong Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Ling Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
- *Correspondence: Xianchun Duan, ; Weidong Chen,
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- *Correspondence: Xianchun Duan, ; Weidong Chen,
| |
Collapse
|
94
|
Qing F, Xie T, Xie L, Guo T, Liu Z. How Gut Microbiota Are Shaped by Pattern Recognition Receptors in Colitis and Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14153821. [PMID: 35954484 PMCID: PMC9367250 DOI: 10.3390/cancers14153821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The pathogenesis of intestinal inflammatory disorders such as colitis and colorectal cancer is complicated and dysregulation of gut microbiota is considered an important contributing factor. Inflammation is often initiated by the activation of pattern recognition receptors. However, the relationship between these innate immune receptors and gut microbiota is not fully understood. Here, we show that pattern recognition receptors not only recognize pathogens and initiate inflammatory signal transduction to induce immune responses, but also influence the composition of intestinal microorganisms, thus affecting the development of intestinal inflammation and cancer through various mechanisms. This suggests that the modification of innate immune receptors and relevant molecules could be therapeutic targets for the treatment of colitis and colorectal cancer by regulating gut microbiota. Abstract Disorders of gut microbiota have been closely linked to the occurrence of various intestinal diseases including colitis and colorectal cancer (CRC). Specifically, the production of beneficial bacteria and intestinal metabolites may slow the development of some intestinal diseases. Recently, it has been proposed that pattern recognition receptors (PRRs) not only recognize pathogens and initiate inflammatory signal transduction to induce immune responses but also influence the composition of intestinal microorganisms. However, the mechanisms through which PRRs regulate gut microbiota in the setting of colitis and CRC have rarely been systematically reviewed. Therefore, in this paper, we summarize recent advances in our understanding of how PRRs shape gut microbiota and how this influences the development of colitis and CRC.
Collapse
Affiliation(s)
- Furong Qing
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- School of Graduate, Gannan Medical University, Ganzhou 341000, China
| | - Tao Xie
- Center for Scientific Research, Gannan Medical University, Ganzhou 341000, China
| | - Lu Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Tianfu Guo
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Correspondence: (T.G.); (Z.L.)
| | - Zhiping Liu
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Center for Scientific Research, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Correspondence: (T.G.); (Z.L.)
| |
Collapse
|
95
|
Kienes I, Johnston EL, Bitto NJ, Kaparakis-Liaskos M, Kufer TA. Bacterial subversion of NLR-mediated immune responses. Front Immunol 2022; 13:930882. [PMID: 35967403 PMCID: PMC9367220 DOI: 10.3389/fimmu.2022.930882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Members of the mammalian Nod-like receptor (NLR) protein family are important intracellular sensors for bacteria. Bacteria have evolved under the pressure of detection by host immune sensing systems, leading to adaptive subversion strategies to dampen immune responses for their benefits. These include modification of microbe-associated molecular patterns (MAMPs), interception of innate immune pathways by secreted effector proteins and sophisticated instruction of anti-inflammatory adaptive immune responses. Here, we summarise our current understanding of subversion strategies used by bacterial pathogens to manipulate NLR-mediated responses, focusing on the well-studied members NOD1/2, and the inflammasome forming NLRs NLRC4, and NLRP3. We discuss how bacterial pathogens and their products activate these NLRs to promote inflammation and disease and the range of mechanisms used by bacterial pathogens to evade detection by NLRs and to block or dampen NLR activation to ultimately interfere with the generation of host immunity. Moreover, we discuss how bacteria utilise NLRs to facilitate immunotolerance and persistence in the host and outline how various mechanisms used to attenuate innate immune responses towards bacterial pathogens can also aid the host by reducing immunopathologies. Finally, we describe the therapeutic potential of harnessing immune subversion strategies used by bacteria to treat chronic inflammatory conditions.
Collapse
Affiliation(s)
- Ioannis Kienes
- Department of Immunology, University of Hohenheim, Stuttgart, Germany
| | - Ella L. Johnston
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Natalie J. Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Thomas A. Kufer
- Department of Immunology, University of Hohenheim, Stuttgart, Germany
- *Correspondence: Thomas A. Kufer,
| |
Collapse
|
96
|
Activation and Pharmacological Regulation of Inflammasomes. Biomolecules 2022; 12:biom12071005. [PMID: 35883561 PMCID: PMC9313256 DOI: 10.3390/biom12071005] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammasomes are intracellular signaling complexes of the innate immune system, which is part of the response to exogenous pathogens or physiological aberration. The multiprotein complexes mainly consist of sensor proteins, adaptors, and pro-caspase-1. The assembly of the inflammasome upon extracellular and intracellular cues drives the activation of caspase-1, which processes pro-inflammatory cytokines IL-1β and IL-18 to maturation and gasdermin-D for pore formation, leading to pyroptosis and cytokine release. Inflammasome signaling functions in numerous infectious or sterile inflammatory diseases, including inherited autoinflammatory diseases, metabolic disorders, cardiovascular diseases, cancers, neurodegenerative disorders, and COVID-19. In this review, we summarized current ideas on the organization and activation of inflammasomes, with details on the molecular mechanisms, regulations, and interventions. The recent developments of pharmacological strategies targeting inflammasomes as disease therapeutics were also covered.
Collapse
|
97
|
Liu J, Liu Q, Shen H, Liu Y, Wang Y, Wang G, Du J. Identification and Validation of a Three Pyroptosis-Related lncRNA Signature for Prognosis Prediction in Lung Adenocarcinoma. Front Genet 2022; 13:838624. [PMID: 35928454 PMCID: PMC9345371 DOI: 10.3389/fgene.2022.838624] [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: 12/18/2021] [Accepted: 06/15/2022] [Indexed: 12/03/2022] Open
Abstract
Pyroptosis, defined as programmed cell death, results in the release of inflammatory mediators. Recent studies have revealed that pyroptosis plays essential roles in antitumor immunity and immunotherapy efficacy. Long noncoding RNAs (lncRNAs) are involved in a variety of biological behaviors in tumor cells, although the roles and mechanisms of lncRNAs in pyroptosis are rarely studied. Our study aimed to establish a novel pyroptosis-related lncRNA signature as a forecasting tool for predicting prognosis and ascertaining immune value. Based on lung adenocarcinoma (LUAD) patients from The Cancer Genome Atlas (TCGA), we performed Pearson’s correlation analysis to identify pyroptosis-related lncRNAs. After differentially expressed gene analysis and univariate Cox regression analysis, we selected prognosis-related and differentially expressed lncRNAs. Finally, we performed multivariate Cox regression analysis to establish the three pyroptosis-related lncRNA signature. Kaplan–Meier (KM) survival analyses and receiver operating characteristic (ROC) curves indicated the excellent performance for predicting the prognosis of LUAD patients. At the same time, we applied multidimensional approaches to further explore the functional enrichment, tumor microenvironment (TME) landscape, and immunotherapy efficacy among the different risk groups. A nomogram was constructed by integrating risk scores and clinical characteristics, which was validated using calibrations and ROC curves. Three lncRNAs, namely, AC090559.1, AC034102.8, and AC026355.2, were involved in this signature and used to classify LUAD patients into low- and high-risk groups. Overall survival time (OS) was higher in the low-risk group than in the high-risk group, which was also validated in our LUAD cohort from Shandong Provincial Hospital. TME landscape analyses revealed that a higher abundance of infiltrating immune cells and a greater prevalence of immune-related events existed in the low-risk group. Meanwhile, higher expression of immune checkpoint (ICP) genes, higher immunophenoscore (IPSs), and greater T cell dysfunction in the low-risk group demonstrated a better response to immunotherapy than the high-risk group. Combined with predictions from the Tumor Immune Dysfunction and Exclusion (TIDE) website, we found that LUAD patients in the low-risk group significantly benefited from programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte–associated protein 4 (CTLA4) immune checkpoint blockade (ICB) therapy compared with those in the high-risk group. Furthermore, drug susceptibility analysis identified potential sensitive chemotherapeutic drugs for each risk group. In this study, a novel three pyroptosis-related lncRNA signature was constructed, which could accurately predict the immunotherapy efficacy and prognosis in LUAD patients.
Collapse
Affiliation(s)
- Jichang Liu
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiang Liu
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongchang Shen
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yong Liu
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yadong Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guanghui Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Jiajun Du,
| |
Collapse
|
98
|
Lara-Reyna S, Caseley EA, Topping J, Rodrigues F, Jimenez Macias J, Lawler SE, McDermott MF. Inflammasome activation: from molecular mechanisms to autoinflammation. Clin Transl Immunology 2022; 11:e1404. [PMID: 35832835 PMCID: PMC9262628 DOI: 10.1002/cti2.1404] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammasomes are assembled by innate immune sensors that cells employ to detect a range of danger signals and respond with pro-inflammatory signalling. Inflammasomes activate inflammatory caspases, which trigger a cascade of molecular events with the potential to compromise cellular integrity and release the IL-1β and IL-18 pro-inflammatory cytokines. Several molecular mechanisms, working in concert, ensure that inflammasome activation is tightly regulated; these include NLRP3 post-translational modifications, ubiquitination and phosphorylation, as well as single-domain proteins that competitively bind to key inflammasome components, such as the CARD-only proteins (COPs) and PYD-only proteins (POPs). These diverse regulatory systems ensure that a suitable level of inflammation is initiated to counteract any cellular insult, while simultaneously preserving tissue architecture. When inflammasomes are aberrantly activated can drive excessive production of pro-inflammatory cytokines and cell death, leading to tissue damage. In several autoinflammatory conditions, inflammasomes are aberrantly activated with subsequent development of clinical features that reflect the degree of underlying tissue and organ damage. Several of the resulting disease complications may be successfully controlled by anti-inflammatory drugs and/or specific cytokine inhibitors, in addition to more recently developed small-molecule inhibitors. In this review, we will explore the molecular processes underlying the activation of several inflammasomes and highlight their role during health and disease. We also describe the detrimental effects of these inflammasome complexes, in some pathological conditions, and review current therapeutic approaches as well as future prospective treatments.
Collapse
Affiliation(s)
- Samuel Lara-Reyna
- Institute of Microbiology and Infection University of Birmingham Birmingham UK
| | - Emily A Caseley
- School of Biomedical Sciences, Faculty of Biological Sciences University of Leeds Leeds UK
| | - Joanne Topping
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital University of Leeds Leeds UK
| | - François Rodrigues
- AP-HP, Hôpital Tenon, Sorbonne Université, Service de Médecine interne Centre de Référence des Maladies Auto-inflammatoires et des Amyloses d'origine inflammatoire (CEREMAIA) Paris France
| | - Jorge Jimenez Macias
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA.,Brown Cancer Centre, Department of Pathology and Laboratory Medicine Brown University Providence Rhode Island USA
| | - Sean E Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA.,Brown Cancer Centre, Department of Pathology and Laboratory Medicine Brown University Providence Rhode Island USA
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital University of Leeds Leeds UK
| |
Collapse
|
99
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
100
|
de Araújo FM, Cuenca-Bermejo L, Fernández-Villalba E, Costa SL, Silva VDA, Herrero MT. Role of Microgliosis and NLRP3 Inflammasome in Parkinson's Disease Pathogenesis and Therapy. Cell Mol Neurobiol 2022; 42:1283-1300. [PMID: 33387119 PMCID: PMC11421755 DOI: 10.1007/s10571-020-01027-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked primarily by motor symptoms such as rigidity, bradykinesia, postural instability and resting tremor associated with dopaminergic neuronal loss in the Substantia Nigra pars compacta (SNpc) and deficit of dopamine in the basal ganglia. These motor symptoms can be preceded by pre-motor symptoms whose recognition can be useful to apply different strategies to evaluate risk, early diagnosis and prevention of PD progression. Although clinical characteristics of PD are well defined, its pathogenesis is still not completely known, what makes discoveries of therapies capable of curing patients difficult to be reached. Several theories about the cause of idiopathic PD have been investigated and among them, the key role of inflammation, microglia and the inflammasome in the pathogenesis of PD has been considered. In this review, we describe the role and relation of both the inflammasome and microglial activation with the pathogenesis, symptoms, progression and the possibilities for new therapeutic strategies in PD.
Collapse
Affiliation(s)
- Fillipe M de Araújo
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - Lorena Cuenca-Bermejo
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Emiliano Fernández-Villalba
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Silvia L Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.
| | - Victor Diogenes A Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.
| | - Maria Trinidad Herrero
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain.
| |
Collapse
|