1
|
Yi YS. Regulatory roles of noncanonical inflammasomes in kidney diseases. Int Immunopharmacol 2025; 157:114787. [PMID: 40319748 DOI: 10.1016/j.intimp.2025.114787] [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: 03/12/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Inflammation is a body's immune defense against infection and injury. The inflammatory response has two main phases: the priming and triggering phases. The triggering phase involves the activation of inflammasomes, cytoplasmic platforms for initiating inflammation, and Inflammasomes are classified into two categories: canonical and noncanonical. Kidney disease, or renal disease, refers to damage or illness affecting the kidneys, leading to a progressive decline in their function. Although the roles of canonical inflammasomes in kidney diseases are well-documented, recent research highlights novel roles of noncanonical inflammasomes in managing various inflammatory conditions. In particular, emerging studies emphasize the regulatory functions of noncanonical inflammasomes in a range of kidney diseases. This review explores recent discoveries regarding the regulatory functions of noncanonical inflammasomes, including human caspase-4/5 and murine caspase-11, in the progression of kidney diseases. It also examines the potential of targeting these inflammasomes as a novel therapeutic approach.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Biological Sciences, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
2
|
Tang Y, Tong W, Peng Y, Sun S. Targeting cholesterol-driven pyroptosis: a promising strategy for the prevention and treatment of atherosclerosis. Mol Biol Rep 2025; 52:459. [PMID: 40372511 DOI: 10.1007/s11033-025-10554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Funding Pyroptosis is a type of programmed cell death (PCD) pathway distinguished by inflammation. It is activated by specific inflammasomes. Once activated, it causes the physical breakdown of the cell, along with the discharge of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and interleukin-18 (IL-18). Abundant evidence has demonstrated the existence of pyroptotic cell death within atherosclerotic plaques, which has significance for the development of atherosclerosis (AS). As a result, pyroptosis has become a new and important topic in cardiovascular disease (CVD) research. Cholesterol, it is recognized to have a connection with inflammation, exerts a crucial function in the development process of AS, and has been linked to the initiation of pyroptosis. This review aims to briefly summarize the fundamental aspects of pyroptosis and the influence of cholesterol-related inflammation in AS. Additionally, this review will explore potential therapeutic approaches based on pyroptosis that could be utilized for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Yujiao Peng
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
3
|
Yi YS. Functional interplay between non-canonical inflammasomes and autophagy in inflammatory responses and diseases. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:129-138. [PMID: 39539180 PMCID: PMC11842290 DOI: 10.4196/kjpp.24.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024]
Abstract
The inflammasome is a cytosolic multiprotein platform that plays a key role in the inflammatory response, an essential innate immune response that protects the body from pathogens and cellular danger signals. Autophagy is a fundamental cellular mechanism that maintains homeostasis through the elimination and recycling of dysfunctional molecules and subcellular elements. Many previous studies have demonstrated a functional interplay between canonical inflammasomes that were earlier discovered and autophagy in inflammatory responses and diseases. Given the increasing evidence that non-canonical inflammasomes are unique and key factors in inflammatory responses, the functional interplay between non-canonical inflammasomes and autophagy is noteworthy. Recent studies have demonstrated that non-canonical inflammasomes and autophagy are functionally correlated with inflammatory responses and diseases. This review comprehensively discusses recent studies that have investigated the functional interplay of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4, with autophagy and autophagy-related proteins in inflammatory responses and diseases and provides insight into the development of novel anti-inflammatory therapeutics by modulating the functional interplay between non-canonical inflammasomes and autophagy.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Korea
| |
Collapse
|
4
|
Kuc-Ciepluch D, Ciepluch K, Augustyniak D, Guła G, Maciejewska B, Kowalik A, Jop E, Drulis-Kawa Z, Arabski M. Exploiting gasdermin-mediated pyroptosis for enhanced antimicrobial activity of phage endolysin against Pseudomonas aeruginosa. mSystems 2025; 10:e0110624. [PMID: 39714210 PMCID: PMC11748493 DOI: 10.1128/msystems.01106-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024] Open
Abstract
Pyroptosis is an inflammatory immune response of eukaryotic cells to bacterial lipopolysaccharide (LPS) and other pathological stimuli, leading to the activation of the gasdermin D (GSDMD) and secretion of pore-forming domain GSDMDNterm, facilitating the release of cytokines. Additionally, GSDMDNterm exhibits antibacterial properties through interactions with bacterial outer membranes (OM). We explored alternative antimicrobial strategy to determine whether inducing natural pyroptosis via GSDMD activation by P. aeruginosa LPS could enhance the effectiveness of recombinant phage endopeptidase KP27 (peptidoglycan-degrading enzyme) against P. aeruginosa, enabling penetration through OM and bacterial killing synergistically. Our findings demonstrated that recombinant GSDMD alone exhibited antibacterial effects against wild-type P. aeruginosa with smooth LPS, while recombinant GSDMDNterm efficiently permeabilized both smooth LPS-bearing and O-chain-deficient P. aeruginosa potentially synergizing with endolysin KP27. Transcriptomic analyses revealed the activation of the immune system pathways in response to LPS, mainly in monocytic cells, in contrast to epithelial A549 or HeLa cell lines. LPS-induced pyroptosis in monocytes led to GSDMD cleavage and the release of interleukins, regardless of the nature/origin of the LPS used. However, the pyroptosis stimulation by LPS in the antibacterial assay was not effective enough for bacterial OM permeabilization and enhancement of endolysin activity. We assume that leveraging pyroptosis induction in monocytic cells to augment the bactericidal activity of endolysins may be limited. IMPORTANCE Recombinant GSDMDNterm protein was able to efficiently permeabilize P. aeruginosa outer membranes and increase endolysin activity against bacteria, producing either long LPS O-chains or lack them entirely. The obtained results suggest the limited possibility of using the natural process of pyroptosis occurring in monocytic cells to enhance the bactericidal effect of recombinant phage endolysins against Gram-negative bacteria infection.
Collapse
Affiliation(s)
- Dorota Kuc-Ciepluch
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Karol Ciepluch
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Daria Augustyniak
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Grzegorz Guła
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Barbara Maciejewska
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Artur Kowalik
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland
| | - Ewelina Jop
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Michał Arabski
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| |
Collapse
|
5
|
Yi YS. Regulatory Roles of Noncanonical Inflammasomes in Inflammatory Lung Diseases. Int J Mol Sci 2024; 26:27. [PMID: 39795884 PMCID: PMC11719999 DOI: 10.3390/ijms26010027] [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: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
The inflammatory response consists of two stages: priming and triggering. The triggering stage is marked by the activation of inflammasomes, which are cytosolic protein complexes acting as platforms for inflammation. Inflammasomes are divided into canonical and noncanonical categories. Inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), inflammatory lung injury, and pulmonary fibrosis arise from lung inflammation and damage. While the role of canonical inflammasomes in these diseases is well demonstrated, recent findings emphasize the critical roles of noncanonical inflammasomes in regulating inflammation and various inflammatory conditions. Particularly, new studies highlight their involvement in inflammatory lung diseases. This review delves into recent research on the regulatory roles of noncanonical inflammasomes, such as human caspase-4 and murine caspase-11, in lung inflammation and the development of inflammatory lung diseases, as well as the potential for targeting these inflammasomes for new treatments.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
6
|
Kim E, Choi DH, Yi YS. Quercetin Ameliorates Acute Lethal Sepsis in Mice by Inhibiting Caspase-11 Noncanonical Inflammasome in Macrophages. Molecules 2024; 29:5900. [PMID: 39769989 PMCID: PMC11678081 DOI: 10.3390/molecules29245900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Quercetin is a natural polyphenolic flavonoid widely found in plants, fruits, and vegetables, and has been reported to play pharmacological roles in numerous pathogenic conditions. The anti-inflammatory effects of quercetin in various inflammatory conditions and diseases have been well-documented. However, its regulatory role in noncanonical inflammasome activation has not yet been demonstrated. This study investigated the anti-inflammatory effects of quercetin in caspase-11 noncanonical inflammasome-activated inflammatory responses in macrophages and a mouse model of acute lethal sepsis. Quercetin protected J774A.1 macrophages from lipopolysaccharide (LPS)-induced cell death and caspase-11 noncanonical inflammasome-induced pyroptosis. It significantly decreased the production and mRNA expression of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-18, and IL-6, but not tumor necrosis factor (TNF)-α, and inflammatory molecules, such as nitric oxide (NO) and inducible NO synthase in caspase-11 noncanonical inflammasome-activated J774A.1 cells. Mechanistically, quercetin strongly suppressed the autoproteolysis and secretion of caspase-11 and the proteolysis of gasdermin D in caspase-11 noncanonical inflammasome-activated J774A.1 cells. However, quercetin did not inhibit the direct binding of caspase-11 to LPS. In vivo, the study revealed that quercetin increased the survival rate of mice with acute lethal sepsis and decreased serum levels of pro-inflammatory cytokines without causing significant toxicity. In conclusion, this study highlights quercetin-mediated anti-inflammatory action in inflammatory responses and acute lethal sepsis through a novel mechanism that targets the caspase-11 noncanonical inflammasome in macrophages, suggesting quercetin as a promising anti-inflammatory agent in natural medicine.
Collapse
Affiliation(s)
| | | | - Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea; (E.K.); (D.-H.C.)
| |
Collapse
|
7
|
Verma A, Azhar G, Patyal P, Zhang W, Zhang X, Wei JY. Proteomic analysis of P. gingivalis-Lipopolysaccharide induced neuroinflammation in SH-SY5Y and HMC3 cells. GeroScience 2024; 46:4315-4332. [PMID: 38507186 PMCID: PMC11336124 DOI: 10.1007/s11357-024-01117-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Chronic periodontitis and its keystone pathogen, Porphyromonas gingivalis, have increasingly been linked with Alzheimer's disease (AD). However, P.gingivalis-lipopolysaccharide (LPS) mediated release of neuroinflammatory proteins contributes to AD remains underexplored. In this study, we utilized data-independent acquisition mass spectrometry to characterize P.gingivalis-LPS induced profile of differentially expressed proteins associated with the neuroinflammatory response in human neuroblastoma (SH-SY5Y) and human microglial (HMC3) cells. We reported a set of 124 proteins in SH-SY5Y cells and 96 proteins in HMC3 cells whose levels were significantly upregulated or downregulated by exposure to P. gingivalis-LPS. Our findings demonstrate that P. gingivalis-LPS contributed to the elevated expressions of dementia biomarkers and pro-inflammatory cytokines that include APP, Aβ1-42, Aβ1-40, T-Tau, p-Tau, VEGF, TGF-β, IL-1β, IL-6 and TNF-α through 2 distinct pathways of extracellular sensing by cell surface receptors and intracellular cytosolic receptors. Interestingly, intracellular signaling proteins activated with P. gingivalis-LPS transfection using Lipofectamine™ 2000 had significantly higher fold change protein expression compared to the extracellular signaling with P. gingivalis-LPS treatment. Additionally, we also explored P. gingivalis-LPS mediated activation of caspase-4 dependent non canonical inflammasome pathway in both SH-SY5Y and HMC3 cells. In summary, P. gingivalis-LPS induced neuroinflammatory protein expression in SH-SY5Y and HMC3 cells, provided insights into the specific inflammatory pathways underlying the potential link between P. gingivalis-LPS infection and the pathogenesis of Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Ambika Verma
- Department of Geriatrics, Donald W. Reynolds Institute On Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR, 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, Donald W. Reynolds Institute On Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR, 72205, USA
| | - Pankaj Patyal
- Department of Geriatrics, Donald W. Reynolds Institute On Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR, 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Xiaomin Zhang
- Department of Geriatrics, Donald W. Reynolds Institute On Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR, 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, Donald W. Reynolds Institute On Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR, 72205, USA.
| |
Collapse
|
8
|
Sánchez-Gil MA, Fraile-Martinez O, García-Montero C, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Camacho-Alcázar A, De León-Luis JA, Bravo C, Díaz-Pedrero R, López-Gonzalez L, Bujan J, Cancelo MJ, Álvarez-Mon M, García-Honduvilla N, Saez MA, Ortega MA. Exacerbated Activation of the NLRP3 Inflammasome in the Placentas from Women Who Developed Chronic Venous Disease during Pregnancy. Int J Mol Sci 2024; 25:5528. [PMID: 38791563 PMCID: PMC11122606 DOI: 10.3390/ijms25105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic venous disease (CVD) comprises a spectrum of morphofunctional disorders affecting the venous system, affecting approximately 1 in 3 women during gestation. Emerging evidence highlights diverse maternofetal implications stemming from CVD, particularly impacting the placenta. While systemic inflammation has been associated with pregnancy-related CVD, preliminary findings suggest a potential link between this condition and exacerbated inflammation in the placental tissue. Inflammasomes are major orchestrators of immune responses and inflammation in different organs and systems. Notwithstanding the relevance of inflammasomes, specifically the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3)- which has been demonstrated in the placentas of women with different obstetric complications, the precise involvement of this component in the placentas of women with CVD remains to be explored. This study employs immunohistochemistry and real-time PCR (RT-qPCR) to examine the gene and protein expression of key components in both canonical and non-canonical pathways of the NLRP3 inflammasome (NLRP3, ASC-apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain-caspase 1, caspase 5, caspase 8, and interleukin 1β) within the placental tissue of women affected by CVD. Our findings reveal a substantial upregulation of these components in CVD-affected placentas, indicating a potential pathophysiological role of the NLRP3 inflammasome in the development of this condition. Subsequent investigations should focus on assessing translational interventions addressing this dysregulation in affected patient populations.
Collapse
Affiliation(s)
- María Asunción Sánchez-Gil
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- University Defense Center of Madrid (CUD), 28047 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Adrían Camacho-Alcázar
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
| | - Juan A. De León-Luis
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain; (J.A.D.L.-L.); (C.B.)
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain; (J.A.D.L.-L.); (C.B.)
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Raúl Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Laura López-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - María J. Cancelo
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
- Department of Obstetrics and Gynecology, University Hospital of Guadalajara, 19002 Guadalajara, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.S.-G.); (O.F.-M.); (C.G.-M.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (A.C.-A.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| |
Collapse
|
9
|
Joon Lee D, Yeol Lee S, Yi YS. Maclurin inhibits caspase-11 non-canonical inflammasome in macrophages and ameliorates acute lethal sepsis in mice. Int Immunopharmacol 2024; 129:111615. [PMID: 38330799 DOI: 10.1016/j.intimp.2024.111615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
Maclurin is a natural phenolic compound isolated from Morus alba(white mulberry) andGarcinia mangostana (purple mangosteen) and has been reported to regulate cancer progression, oxidative stress, and melanogenesis. The regulatory role of maclurin, however, has never been demonstrated. This study investigated in vitro and in vivo anti-inflammatory roles of maclurin and the underlying mechanism in caspase-11 non-canonical inflammasome-stimulated inflammatory responses in macrophages and an animal model of acute lethal sepsis. Maclurin protected J774A.1 macrophages from LPS-induced cytotoxicity and suppressed caspase-11 non-canonical inflammasome-stimulated pyroptosis. Maclurin decreased the secretion and mRNA expression of pro-inflammatory cytokines and inflammatory mediators, such as IL-1β, IL-18, TNF-α, IL-6, nitric oxide (NO), and inducible NO synthase (iNOS) in caspase-11 non-canonical inflammasome-stimulated J774A.1 macrophages. Mechanistic studies revealed that maclurin markedly suppressed the proteolytic activation of caspase-11 and gasdermin D (GSDMD) in caspase-11 non-canonical inflammasome-stimulated J774A.1 macrophages, while it did not inhibit caspase-11-mediated direct sensing of LPS. In vivo study revealed that maclurin ameliorated acute lethal sepsis in mice by increasing the survival rate and decreasing the serum levels of IL-1β and IL-18 without significant toxicity. In conclusion, this study suggests that maclurin is a novel anti-inflammatory agent in inflammatory responses and against acute lethal sepsis via the inhibition of the caspase-11 non-canonical inflammasome in macrophages, which justifies its potential as an anti-inflammatory therapeutic agent in traditional medicine.
Collapse
Affiliation(s)
- Dong Joon Lee
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| | - Sang Yeol Lee
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea.
| | - Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
10
|
Yi YS. MicroRNA-mediated epigenetic regulation of inflammasomes in inflammatory responses and immunopathologies. Semin Cell Dev Biol 2024; 154:227-238. [PMID: 36437174 DOI: 10.1016/j.semcdb.2022.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
Inflammation represents the first-line defense mechanism of the host against pathogens and cellular stress. One of the most critical inflammatory responses is characterized by the activation of inflammasomes, intracellular multiprotein complexes that induce inflammatory signaling pathways in response to various pathogen-associated molecular patterns or danger-associated molecular patterns under physiological and pathological conditions. Inflammasomes are tightly regulated in normal cells, and dysregulation of these complexes is observed in various pathological conditions, especially inflammatory diseases and cancers. Epigenetic regulation has been suggested as a key mechanism in modulating inflammasome activity, and microRNAs (miRNAs) have been implicated in the post-transcriptional regulation of inflammasomes. Therefore, miRNA-mediated epigenetic regulation of inflammasomes in pathological conditions has received considerable attention, and current strategies for targeting inflammasomes have been shown to be effective in the treatment of diseases associated with inflammasome activation. This review summarizes recent studies suggesting the roles of miRNAs in the epigenetic control of inflammasomes and highlights the potential of miRNAs as a therapeutic tool for treating human diseases.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, South Korea.
| |
Collapse
|
11
|
Yi YS. Roles of the Caspase-11 Non-Canonical Inflammasome in Rheumatic Diseases. Int J Mol Sci 2024; 25:2091. [PMID: 38396768 PMCID: PMC10888639 DOI: 10.3390/ijms25042091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammasomes are intracellular multiprotein complexes that activate inflammatory signaling pathways. Inflammasomes comprise two major classes: canonical inflammasomes, which were discovered first and are activated in response to a variety of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), and non-canonical inflammasomes, which were discovered recently and are only activated in response to intracellular lipopolysaccharide (LPS). Although a larger number of studies have successfully demonstrated that canonical inflammasomes, particularly the NLRP3 inflammasome, play roles in various rheumatic diseases, including rheumatoid arthritis (RA), infectious arthritis (IR), gouty arthritis (GA), osteoarthritis (OA), systemic lupus erythematosus (SLE), psoriatic arthritis (PA), ankylosing spondylitis (AS), and Sjögren's syndrome (SjS), the regulatory roles of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 non-canonical inflammasomes, in these diseases are still largely unknown. Interestingly, an increasing number of studies have reported possible roles for non-canonical inflammasomes in the pathogenesis of various mouse models of rheumatic disease. This review comprehensively summarizes and discusses recent emerging studies demonstrating the regulatory roles of non-canonical inflammasomes, particularly focusing on the caspase-11 non-canonical inflammasome, in the pathogenesis and progression of various types of rheumatic diseases and provides new insights into strategies for developing potential therapeutics to prevent and treat rheumatic diseases as well as associated diseases by targeting non-canonical inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
12
|
Zhang S, Liu C, Sun J, Li Y, Lu J, Xiong X, Hu L, Zhao H, Zhou H. Bridging the Gap: Investigating the Link between Inflammasomes and Postoperative Cognitive Dysfunction. Aging Dis 2023; 14:1981-2002. [PMID: 37450925 PMCID: PMC10676784 DOI: 10.14336/ad.2023.0501] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 05/01/2023] [Indexed: 07/18/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a cluster of cognitive problems that may arise after surgery. POCD symptoms include memory loss, focus inattention, and communication difficulties. Inflammasomes, intracellular multiprotein complexes that control inflammation, may have a significant role in the development of POCD. It has been postulated that the NLRP3 inflammasome promotes cognitive impairment by triggering the inflammatory response in the brain. Nevertheless, there are many gaps in the current literature to understand the underlying pathophysiological mechanisms and develop future therapy. This review article underlines the limits of our current knowledge about the NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome and POCD. We first discuss inflammasomes and their types, structures, and functions, then summarize recent evidence of the NLRP3 inflammasome's involvement in POCD. Next, we propose a hypothesis that suggests the involvement of inflammasomes in multiple organs, including local surgical sites, blood circulation, and other peripheral organs, leading to systemic inflammation and subsequent neuronal dysfunction in the brain, resulting in POCD. Research directions are then discussed, including analyses of inflammasomes in more clinical POCD animal models and clinical trials, studies of inflammasome types that are involved in POCD, and investigations into whether inflammasomes occur at the surgical site, in circulating blood, and in peripheral organs. Finally, we discuss the potential benefits of using new technologies and approaches to study inflammasomes in POCD. A thorough investigation of inflammasomes in POCD might substantially affect clinical practice.
Collapse
Affiliation(s)
- Siyu Zhang
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Jintao Sun
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Yang Li
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Jian Lu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Heng Zhao
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Hongmei Zhou
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| |
Collapse
|
13
|
Garcia-Puente LM, Fraile-Martinez O, García-Montero C, Bujan J, De León-Luis JA, Bravo C, Rodríguez-Benitez P, Pintado P, Ruiz-Labarta FJ, Álvarez-Mon M, García-Honduvilla N, Cancelo MJ, Saez MA, Ortega MA. Placentas from Women with Late-Onset Preeclampsia Exhibit Increased Expression of the NLRP3 Inflammasome Machinery. Biomolecules 2023; 13:1644. [PMID: 38002326 PMCID: PMC10669618 DOI: 10.3390/biom13111644] [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] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Pre-eclampsia is a harmful and potentially lethal medical condition during pregnancy clinically diagnosed by hypertension and commonly accompanied by proteinuria and multiorgan affections. According to the time of diagnosis, it is differentiated between early-onset (EO-PE) and late-onset preeclampsia (LO-PE). Despite being less dangerous and presenting distinct pathophysiological signatures, LO-PE has a greater prevalence than EO-PE, both having significant consequences on the placenta. Previous works have evidenced that exacerbated inflammation in this organ might play a potential pathogenic role in the development of pre-eclampsia, and there is some preliminary evidence that the hyperactivation of inflammasomes can be related to the altered immunoinflammatory responses observed in the placentas of these patients. However, the precise role of inflammasomes in the placentas of women with LO-PE remains to be fully understood. In this work, we have studied the gene and protein expression of the main components related to the canonical and non-canonical pathways of the inflammasome NLRP3 (NLRP3, ASC, caspase 1, caspase 5, caspase 8, interleukin 1β, and interleukin 18) in the placental tissue of women with LO-PE. Our results show a marked increase in all these components in the placentas of women who have undergone LO-PE, suggesting that NLRP3 inflammasome plays a potentially pathophysiological role in the development of this entity. Future works should aim to evaluate possible translational approaches to this dysregulation in these patients.
Collapse
Affiliation(s)
- Luis M Garcia-Puente
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Juan A De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Patrocinio Rodríguez-Benitez
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
- Department of Nephrology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
| | - Pilar Pintado
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Francisco Javier Ruiz-Labarta
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - María J Cancelo
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Department of Obstetrics and Gynecology, University Hospital of Guadalajara, 19002 Guadalajara, Spain
| | - Miguel A Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcalá de Henares, Spain
| | - Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| |
Collapse
|
14
|
Cho HJ, Lee DJ, Yi YS. Anti-inflammatory activity of calmodulin-lysine N-methyltransferase through suppressing the caspase-11 non-canonical inflammasome. Immunobiology 2023; 228:152758. [PMID: 37948850 DOI: 10.1016/j.imbio.2023.152758] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Calmodulin (CaM)-lysine N-methyltransferase (CAMKMT) is a novel methyltransferase that catalyzes lysine trimethylation in CaM. However, its specific roles in inflammatory responses and diseases remain unclear. In this study, we investigated the effects of CAMKMT on caspase-11 non-canonical inflammasomes. CAMKMT expression levels were significantly decreased during inflammatory responses activated by caspase-11 non-canonical inflammasome in macrophages. Moreover, CaM lysine trimethylation was markedly inhibited, but no change was observed in CaM expression during these inflammatory responses in macrophages. Activation of the CaM downstream effectors, CaM-dependent proteinkinase kinase 2 and CaM-dependent proteinkinase type IV, was also inhibited during inflammatory responses activated by caspase-11 non-canonical inflammasome in macrophages. Notably, forced expression of CAMKMT restrained caspase-11 non-canonical inflammasome activation via inhibiting proteolytic activation of caspase-11 and gasdermin D (GSDMD), which in turn suppressed pyroptosis and the release of interleukin (IL)-1β and IL-18 in macrophages. Finally, an in vivo study revealed that CAMKMT ameliorated lipopolysaccharide (LPS)-stimulated acute lethal sepsis in mice by increasing the survival rate and reducing the serum levels of IL-1 β. These findings suggest CAMKMT as a novel methyltransferase that plays an anti-inflammatory role through restraining caspase-11 non-canonical inflammasome in macrophages.
Collapse
Affiliation(s)
- Hui-Jin Cho
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| | - Dong Joon Lee
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| | - Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
15
|
Tan J, Zhuo Z, Si Y. Application of pyroptosis in tumor research (Review). Oncol Lett 2023; 26:376. [PMID: 37559585 PMCID: PMC10407856 DOI: 10.3892/ol.2023.13962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/27/2023] [Indexed: 08/11/2023] Open
Abstract
As a potent clinical strategy, cancer therapy has sparked an academic boom over the past few years. Immune checkpoint inhibitors (ICIs) have been demonstrated to be highly successful. These achievements have progressed cancer treatment and have made an indelible mark on cancer. However, the inherent complexity of cancer means that only part of the population can benefit from this treatment. Pyroptosis is a new suicidal cellular mechanism that induces inflammation by releasing immunogenic cellular components. Inflammatory signaling cascades mediated by pyroptosis commonly inspire numerous cell lysis in immune diseases. Contrariwise, this consequence may be a promising target in cancer research. Therefore, the present study briefly described programmed cell death processes and their potential roles in cancer. Because of the rapid development of bioengineering in cancer, the present study also examined the associated scaffolding available for cancer, highlighting advances in tumor engineering approaches. Ultimately, an improved understanding of pyroptosis and tumor scaffolding might shed light on a combination that can be manipulated for therapeutic purposes.
Collapse
Affiliation(s)
- Jianing Tan
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Suzhou, Jiangsu 215500, P.R. China
| | - Ziliang Zhuo
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Suzhou, Jiangsu 215500, P.R. China
| | - Yu Si
- Basic Research Laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
16
|
Yi YS. Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases. Int J Mol Sci 2023; 24:10402. [PMID: 37373549 DOI: 10.3390/ijms241210402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Inflammasomes are multiprotein complexes that activate inflammatory responses by inducing pyroptosis and secretion of pro-inflammatory cytokines. Along with many previous studies on inflammatory responses and diseases induced by canonical inflammasomes, an increasing number of studies have demonstrated that non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 inflammasomes, are emerging key players in inflammatory responses and various diseases. Flavonoids are natural bioactive compounds found in plants, fruits, vegetables, and teas and have pharmacological properties in a wide range of human diseases. Many studies have successfully demonstrated that flavonoids play an anti-inflammatory role and ameliorate many inflammatory diseases by inhibiting canonical inflammasomes. Others have demonstrated the anti-inflammatory roles of flavonoids in inflammatory responses and various diseases, with a new mechanism by which flavonoids inhibit non-canonical inflammasomes. This review discusses recent studies that have investigated the anti-inflammatory roles and pharmacological properties of flavonoids in inflammatory responses and diseases induced by non-canonical inflammasomes and further provides insight into developing flavonoid-based therapeutics as potential nutraceuticals against human inflammatory diseases.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
17
|
Kim YB, Cho HJ, Yi YS. Anti-inflammatory role of Artemisia argyi methanol extract by targeting the caspase-11 non-canonical inflammasome in macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116231. [PMID: 36754190 DOI: 10.1016/j.jep.2023.116231] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia argyi possesses pharmacological activities against various immunopathological conditions associated with inflammation. AIM OF THE STUDY This study explored the inhibitory role of Artemisia argyi methanol extract (Aa-ME) in inflammatory responses and the underlying mechanism in macrophages. MATERIALS AND METHODS Caspase-11 non-canonical inflammasome was activated in J774A.1 macrophage by Pam3CSK4 treatment and lipopolysaccharide (LPS) transfection. Aa-ME-mediated in vitro anti-inflammatory action was examined using MTT assay, lactate dehydrogenase (LDH) activity assay, enzyme-linked immunosorbent assay (ELISA), nitric oxide (NO) generation assay, and quantitative real-time polymerase chain reaction (qPCR). Aa-ME-mediated in vivo anti-inflammatory action was examined in LPS-stimulated lethal septic mice. RESULTS Aa-ME inhibited caspase-11 non-canonical inflammasome-stimulated pyroptosis and the secretion of IL-1β and IL-18 in J774A.1 macrophages. Aa-ME also inhibited NO generation by downregulating inducible NO synthase (iNOS) expression in LPS-primed and caspase-11 non-canonical inflammasome-triggered J774A.1 cells. The mechanism study revealed Aa-ME suppressed the auto-proteolytic activation of caspase-11 and gasdermin D (GSDMD) in J774A.1 cells and also interfered with caspase-11-mediated direct recognition of LPS. Moreover, Aa-ME alleviated LPS-induced lethal sepsis in mice by increasing their survival rate without significant toxicity. CONCLUSION These results suggest a novel mechanism by which Aa-ME alleviates inflammatory responses by deactivating caspase-11 non-canonical inflammasome in macrophages.
Collapse
Affiliation(s)
- Young Bin Kim
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| | - Hui-Jin Cho
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| | - Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| |
Collapse
|
18
|
Cho HJ, Kim E, Yi YS. Korean Red Ginseng Saponins Play an Anti-Inflammatory Role by Targeting Caspase-11 Non-Canonical Inflammasome in Macrophages. Int J Mol Sci 2023; 24:ijms24021077. [PMID: 36674594 PMCID: PMC9861816 DOI: 10.3390/ijms24021077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
We previously reported that Korean red ginseng (KRG) exerts an anti-inflammatory role through inhibiting caspase-11 non-canonical inflammasome in macrophages; however, the components responsible for the anti-inflammatory role remained unclear. This study explored the anti-inflammatory activity of the KRG saponin fraction (KRGSF) in caspase-11 non-canonical inflammasome-activated macrophages. KRGSF inhibited pyroptosis, pro-inflammatory cytokine secretion, and inflammatory mediator production in caspase-11 non-canonical inflammasome-activated J774A.1 cells. A mechanism study revealed that KRGSF-induced anti-inflammatory action was mediated via suppressing the proteolytic activation of caspase-11 and gasdermin D (GSDMD) in caspase-11 non-canonical inflammasome-activated J774A.1 cells. Moreover, KRGSF increased the survival of lethal septic mice. Taken together, these results reveal KRGSF-mediated anti-inflammatory action with a novel mechanism, by inhibiting caspase-11 non-canonical inflammasome in macrophages.
Collapse
|
19
|
Yi YS, Kim HG, Kim JH, Yang WS, Kim E, Park JG, Aziz N, Parameswaran N, Cho JY. Syk promotes phagocytosis by inducing reactive oxygen species generation and suppressing SOCS1 in macrophage-mediated inflammatory responses. Int J Immunopathol Pharmacol 2022; 36:3946320221133018. [PMID: 36214175 PMCID: PMC9548688 DOI: 10.1177/03946320221133018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Inflammation, a vital innate immune response against infection and injury, is mediated by macrophages. Spleen tyrosine kinase (Syk) regulates inflammatory responses in macrophages; however, its role and underlying mechanisms are uncertain. MATERIALS AND METHODS In this study, overexpression and knockout (KO) cell preparations, phagocytosis analysis, confocal microscopy, reactive oxygen species (ROS) determination, mRNA analysis, and immunoprecipitation/western blotting analyses were used to investigate the role of Syk in phagocytosis and its underlying mechanisms in macrophages during inflammatory responses. RESULTS Syk inhibition by Syk KO, Syk-specific small interfering RNA (siSyk), and a selective Syk inhibitor (piceatannol) significantly reduced the phagocytic activity of RAW264.7 cells. Syk inhibition also decreased cytochrome c generation by inhibiting ROS-generating enzymes in lipopolysaccharide (LPS)-stimulated RAW264.7 cells, and ROS scavenging suppressed the phagocytic activity of RAW264.7 cells. LPS induced the tyrosine nitration (N-Tyr) of suppressor of cytokine signaling 1 (SOCS1) through Syk-induced ROS generation in RAW264.7 cells. On the other hand, ROS scavenging suppressed the N-Tyr of SOCS1 and phagocytosis. Moreover, SOCS1 overexpression decreased phagocytic activity, and SOCS1 inhibition increased the phagocytic activity of RAW264.7 cells. CONCLUSION These results suggest that Syk plays a critical role in the phagocytic activity of macrophages by inducing ROS generation and suppressing SOCS1 through SOCS1 nitration during inflammatory responses.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea,Department of Life Sciences, Kyonggi University, Suwon, Korea,Young-Su Yi, Department of Life Sciences, Kyonggi University,154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, Gyeonggi-do 16227, Korea. Jae Youl Cho, Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon Gyeonggi-do 16419, Korea.
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Narayanan Parameswaran
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, MI, USA
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea,Young-Su Yi, Department of Life Sciences, Kyonggi University,154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, Gyeonggi-do 16227, Korea. Jae Youl Cho, Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon Gyeonggi-do 16419, Korea.
| |
Collapse
|
20
|
Yi YS. Regulatory Roles of Caspase-11 Non-Canonical Inflammasome in Inflammatory Liver Diseases. Int J Mol Sci 2022; 23:4986. [PMID: 35563377 PMCID: PMC9104167 DOI: 10.3390/ijms23094986] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/11/2022] Open
Abstract
An inflammatory response consists of two consecutive steps: priming and triggering, to prepare and activate inflammatory responses, respectively. The cardinal feature of the triggering step is the activation of intracellular protein complexes called inflammasomes, which provide a platform for the activation of inflammatory signaling pathways. Despite many studies demonstrating the regulatory roles of canonical inflammasomes in inflammatory liver diseases, the roles of newly discovered non-canonical inflammasomes in inflammatory liver diseases are still largely unknown. Recent studies have reported the regulatory roles of the caspase-11 non-canonical inflammasome in inflammatory liver diseases, providing strong evidence that the caspase-11 non-canonical inflammasome may play key roles in the pathogenesis of inflammatory liver diseases. This review comprehensively discusses the emerging roles of the caspase-11 non-canonical inflammasome in the pathogenesis of inflammatory liver diseases, focusing on non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and inflammatory liver injuries and its underlying mechanisms. This review highlights the current knowledge on the regulatory roles of the caspase-11 non-canonical inflammasome in inflammatory liver diseases, providing new insights into the development of potential therapeutics to prevent and treat inflammatory liver diseases by targeting the caspase-11 non-canonical inflammasome.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Korea
| |
Collapse
|
21
|
A Fluorescence-Polarization-Based Lipopolysaccharide-Caspase-4 Interaction Assay for the Development of Inhibitors. Molecules 2022; 27:molecules27082458. [PMID: 35458656 PMCID: PMC9032125 DOI: 10.3390/molecules27082458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 02/01/2023] Open
Abstract
Recognition of intracellular lipopolysaccharide (LPS) by Caspase-4 (Casp-4) is critical for host defense against Gram-negative pathogens. LPS binds to the N-terminal caspase activation and recruitment domain (CARD) of procaspase-4, leading to auto-proteolytic activation followed by pro-inflammatory cytokine release and pyroptotic cell death. Aberrant hyper-activation of Casp-4 leads to amplification of the inflammatory response linked to sepsis. While the active site of a caspase has been targeted with peptide inhibitors, inhibition of LPS–Casp-4 interaction is an emerging strategy for the development of selective inhibitors with a new mode of action for treating infectious diseases and sepsis induced by LPS. In this study, a high-throughput screening (HTS) system based on fluorescence polarization (FP) was devised to identify inhibitors of the LPS and Casp-4 interaction. Using HTS and IC50 determination and subsequently showing inhibited Casp-4 activity, we demonstrated that the LPS–Casp-4 interaction is a druggable target for Casp-4 inhibition and possibly a non-canonical inflammatory pathway.
Collapse
|
22
|
Yi YS. Potential benefits of ginseng against COVID-19 by targeting inflammasomes. J Ginseng Res 2022; 46:722-730. [PMID: 35399195 PMCID: PMC8979607 DOI: 10.1016/j.jgr.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogenic virus that causes coronavirus disease 2019 (COVID-19), with major symptoms including hyper-inflammation and cytokine storm, which consequently impairs the respiratory system and multiple organs, or even cause death. SARS-CoV-2 activates inflammasomes and inflammasome-mediated inflammatory signaling pathways, which are key determinants of hyperinflammation and cytokine storm in COVID-19 patients. Additionally, SARS-CoV-2 inhibits inflammasome activation to evade the host's antiviral immunity. Therefore, regulating inflammasome initiation has received increasing attention as a preventive measure in COVID-19 patients. Ginseng and its major active constituents, ginsenosides and saponins, improve the immune system and exert anti-inflammatory effects by targeting inflammasome stimulation. Therefore, this review discussed the potential preventive and therapeutic roles of ginseng in COVID-19 based on its regulatory role in inflammasome initiation and the host's antiviral immunity.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, 16227, Republic of Korea.
| |
Collapse
|
23
|
Yi YS. Dual roles of the caspase-11 non-canonical inflammasome in inflammatory bowel disease. Int Immunopharmacol 2022; 108:108739. [PMID: 35366642 DOI: 10.1016/j.intimp.2022.108739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/17/2022] [Accepted: 03/27/2022] [Indexed: 12/29/2022]
Abstract
Inflammation is a two-step process comprising the first priming step that prepares inflammatory responses and the second triggering step that activates inflammatory responses. The key feature of the triggering step is the activation of inflammasomes and intracellular inflammatory protein complexes that provide molecular platforms to activate inflammatory signal transduction cascades. Although canonical inflammasomes have been well demonstrated to be actively involved in numerous human diseases, the roles of the recently identified non-canonical inflammasomes are largely unknown. However, recent studies have demonstrated the emerging roles of the caspase-11 non-canonical inflammasome in various human inflammatory diseases, ultimately providing strong evidence that the caspase-11 non-canonical inflammasome is a key player in the pathogenesis of various human diseases. Here, we comprehensively reviewed the regulatory roles of the caspase-11 non-canonical inflammasome in the pathogenesis of inflammatory bowel disease (IBD) and its underlying mechanisms. Overall, this review highlights the current understanding of the regulatory roles of the caspase-11 non-canonical inflammasome in IBD and may provide insight into new strategies for preventing and treating IBD and caspase-11 non-canonical inflammasome-driven diseases.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Korea.
| |
Collapse
|
24
|
Bahatyrevich-Kharitonik B, Medina-Guzman R, Flores-Cortes A, García-Cruzado M, Kavanagh E, Burguillos MA. Cell Death Related Proteins Beyond Apoptosis in the CNS. Front Cell Dev Biol 2022; 9:825747. [PMID: 35096845 PMCID: PMC8794922 DOI: 10.3389/fcell.2021.825747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
Cell death related (CDR) proteins are a diverse group of proteins whose original function was ascribed to apoptotic cell death signaling. Recently, descriptions of non-apoptotic functions for CDR proteins have increased. In this minireview, we comment on recent studies of CDR proteins outside the field of apoptosis in the CNS, encompassing areas such as the inflammasome and non-apoptotic cell death, cytoskeleton reorganization, synaptic plasticity, mitophagy, neurodegeneration and calcium signaling among others. Furthermore, we discuss the evolution of proteomic techniques used to predict caspase substrates that could potentially explain their non-apoptotic roles. Finally, we address new concepts in the field of non-apoptotic functions of CDR proteins that require further research such the effect of sexual dimorphism on non-apoptotic CDR protein function and the emergence of zymogen-specific caspase functions.
Collapse
Affiliation(s)
- Bazhena Bahatyrevich-Kharitonik
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Rafael Medina-Guzman
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Alicia Flores-Cortes
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Marta García-Cruzado
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Edel Kavanagh
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Miguel Angel Burguillos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| |
Collapse
|
25
|
Yi YS, Kim HG, Kim JH, Yang WS, Kim E, Jeong D, Park JG, Aziz N, Kim S, Parameswaran N, Cho JY. Syk-MyD88 Axis Is a Critical Determinant of Inflammatory-Response in Activated Macrophages. Front Immunol 2022; 12:767366. [PMID: 35003083 PMCID: PMC8733199 DOI: 10.3389/fimmu.2021.767366] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023] Open
Abstract
Background Inflammation, a vital immune response to infection and injury, is mediated by macrophage activation. While spleen tyrosine kinase (Syk) and myeloid differentiation primary response 88 (MyD88) are reportedly involved in inflammatory responses in macrophages, their roles and underlying mechanisms are largely unknown. Methods Here, the role of the MyD88-Syk axis and the mechanism by which Syk and MyD88 cooperate during macrophage-mediated inflammatory responses are explored using knockout conditions of these proteins and mutation strategy as well as flowcytometric and immunoblotting analyses. Results Syk rapidly activates the nuclear factor-kappa B (NF-κB) signaling pathway in lipopolysaccharide (LPS)-stimulated RAW264.7 cells, and the activation of the NF-κB signaling pathway is abolished in Syk−/− RAW264.7 cells. MyD88 activates Syk and Syk-induced activation of NF-κB signaling pathway in LPS-stimulated RAW264.7 cells but Syk-induced inflammatory responses are significantly inhibited in MyD88−/− RAW264.7 cells. MyD88 interacts with Syk through the tyrosine 58 residue (Y58) in the hemi-immunoreceptor tyrosine-based activation motif (ITAM) of MyD88, leading to Syk activation and Syk-induced activation of the NF-κB signaling pathway. Src activates MyD88 by phosphorylation at Y58 via the Src kinase domain. In addition, Ras-related C3 botulinum toxin substrate 1 (Rac1) activation and Rac1-induced formation of filamentous actin (F actin) activate Src in LPS-stimulated RAW264.7 cells. Conclusions These results suggest that the MyD88-Syk axis is a critical player in macrophage-mediated inflammatory responses, and its function is promoted by an upstream Src kinase activated by Rac1-generated filamentous actin (F-actin).
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea.,Department of Life Sciences, Kyonggi University, Suwon, South Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Deok Jeong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Suk Kim
- Institute of Animal Science, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Narayanan Parameswaran
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, MI, United States
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
26
|
Ismail N, Sharma A, Soong L, Walker DH. Review: Protective Immunity and Immunopathology of Ehrlichiosis. ZOONOSES (BURLINGTON, MASS.) 2022; 2:10.15212/zoonoses-2022-0009. [PMID: 35876763 PMCID: PMC9300479 DOI: 10.15212/zoonoses-2022-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Human monocytic ehrlichiosis, a tick transmitted infection, ranges in severity from apparently subclinical to a fatal toxic shock-like fatal disease. Models in immunocompetent mice range from an abortive infection to uniformly lethal depending on the infecting Ehrlichia species, dose of inoculum, and route of inoculation. Effective immunity is mediated by CD4+ T lymphocytes and gamma interferon. Lethal infection occurs with early overproduction of proinflammatory cytokines and overproduction of TNF alpha and IL-10 by CD8+ T lymphocytes. Furthermore, fatal ehrlichiosis is associated with signaling via TLR 9/MyD88 with upregulation of several inflammasome complexes and secretion of IL-1 beta, IL-1 alpha, and IL-18 by hepatic mononuclear cells, suggesting activation of canonical and noncanonical inflammasome pathways, a deleterious role for IL-18, and the protective role for caspase 1. Autophagy promotes ehrlichial infection, and MyD88 signaling hinders ehrlichial infection by inhibiting autophagy induction and flux. Activation of caspase 11 during infection of hepatocytes by the lethal ehrlichial species after interferon alpha receptor signaling results in the production of inflammasome-dependent IL-1 beta, extracellular secretion of HMGB1, and pyroptosis. The high level of HMGB1 in lethal ehrlichiosis suggests a role in toxic shock. Studies of primary bone marrow-derived macrophages infected by highly avirulent or mildly avirulent ehrlichiae reveal divergent M1 and M2 macrophage polarization that links with generation of pathogenic CD8 T cells, neutrophils, and excessive inflammation or with strong expansion of protective Th1 and NKT cells, resolution of inflammation and clearance of infection, respectively.
Collapse
Affiliation(s)
- Nahed Ismail
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Aditya Sharma
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - David H. Walker
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
27
|
Wu Y, Pan B, Zhang Z, Li X, Leng Y, Ji Y, Sun K, Chen AF. Caspase-4/11-Mediated Pulmonary Artery Endothelial Cell Pyroptosis Contributes to Pulmonary Arterial Hypertension. Hypertension 2022; 79:536-548. [PMID: 34984912 DOI: 10.1161/hypertensionaha.121.17868] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial dysfunction enhances vascular inflammation, which initiates pulmonary arterial hypertension (PAH) pathogenesis, further induces vascular remodeling and right ventricular failure. Activation of inflammatory caspases is an important initial event at the onset of pyroptosis. Studies have shown that caspase-1-mediated pyroptosis has played a crucial role in the pathogenesis of PAH. However, the role of caspase-11, another inflammatory caspase, remains to be elucidated. Therefore, the purpose of this study was to clarify the role of caspase-11 in the development of PAH and its mechanism on endothelial cell function. METHODS The role of caspase-11 in the progression of PAH and vascular remodeling was assessed in vivo. In vitro, the effect of caspase-4 silencing on the human pulmonary arterial endothelial cells pyroptosis was determined. RESULTS We confirmed that caspase-11 and its human homolog caspase-4 were activated in PAH animal models and TNF (tumor necrosis factor)-α-induced human pulmonary arterial endothelial cells. Caspase-11-/- relieved right ventricular systolic pressure, right ventricle hypertrophy, and vascular remodeling in Sugen-5416 combined with chronic hypoxia mice model. Meanwhile, pharmacological inhibition of caspase-11 with wedelolactone exhibited alleviated development of PAH on the monocrotaline-induced rat model. Moreover, knockdown of caspase-4 repressed the onset of TNF-α-induced pyroptosis in human pulmonary arterial endothelial cells and inhibited the activation of pyroptosis effector GSDMD (gasdermin D) and GSDME (gasdermin E). CONCLUSIONS These observations identified the critical role of caspase-4/11 in the pyroptosis pathway to modulate pulmonary vascular dysfunction and accelerate the progression of PAH. Our findings provide a potential diagnostic and therapeutic target in PAH.
Collapse
Affiliation(s)
- Yusi Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China (Y.W., X.L.).,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.W., Z.Z., Y.L., A.F.C.)
| | - Bingjie Pan
- Liuzhou Maternal and Child Healthcare Hospital, Liuzhou, Guangxi, China (B.P.)
| | - Zhen Zhang
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.W., Z.Z., Y.L., A.F.C.)
| | - Xiaohui Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China (Y.W., X.L.)
| | - Yiping Leng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.W., Z.Z., Y.L., A.F.C.).,The Affiliated Changsha Central Hospital, Research Center for Phase I Clinical Trials, Hengyang Medical School, University of South China, Changsha, Hunan, China (Y.L.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Y.J.)
| | - Kun Sun
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (K.S., A.F.C.)
| | - Alex F Chen
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.W., Z.Z., Y.L., A.F.C.).,Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (K.S., A.F.C.)
| |
Collapse
|
28
|
Lyu Z, Li Q, Yu Z, Chan Y, Fu L, Li Y, Zhang C. Yi-Zhi-Fang-Dai Formula Exerts Neuroprotective Effects Against Pyroptosis and Blood-Brain Barrier-Glymphatic Dysfunctions to Prevent Amyloid-Beta Acute Accumulation After Cerebral Ischemia and Reperfusion in Rats. Front Pharmacol 2022; 12:791059. [PMID: 34975487 PMCID: PMC8714930 DOI: 10.3389/fphar.2021.791059] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The dysfunctional blood–brain barrier (BBB)–glymphatic system is responsible for triggering intracerebral amyloid-beta peptide (Aβ) accumulation and acts as the key link between ischemic stroke and dementia dominated by Alzheimer’s disease (AD). Recently, pyroptosis in cerebral ischemia and reperfusion (I/R) injury is demonstrated as a considerable mechanism causing BBB–glymphatic dysfunctions and Aβ acute accumulation in the brain. Targeting glial pyroptosis to protect BBB–glymphatic functions after cerebral I/R could offer a new viewpoint to prevent Aβ accumulation and poststroke dementia. Yi-Zhi-Fang-Dai formula (YZFDF) is an herbal prescription used to cure dementia with multiple effects of regulating inflammatory responses and protecting the BBB against toxic Aβ-induced damage. Hence, YZFDF potentially possesses neuroprotective effects against cerebral I/R injury and the early pathology of poststroke dementia, which evokes our current study. Objectives: The present study was designed to confirm the potential efficacy of YZFDF against cerebral I/R injury and explore the possible mechanism associated with alleviating Aβ acute accumulation. Methods: The models of cerebral I/R injury in rats were built by the method of middle cerebral artery occlusion/reperfusion (MCAO/R). First, neurological function assessment and cerebral infarct measurement were used for confirming the efficacy of YZFDF on cerebral I/R injury, and the optimal dosage (YZFDF-H) was selected to conduct the experiments, which included Western blotting detections of pyroptosis, Aβ1-42 oligomers, and NeuN, immunofluorescence observations of glial pyroptosis, aquaporin-4 (AQP-4), and Aβ locations, brain water content measurement, SMI 71 (a specific marker for BBB)/AQP-4 immunohistochemistry, and Nissl staining to further evaluate BBB–glymphatic functions and neuronal damage. Results: YZFDF obviously alleviated neurological deficits and cerebral infarct after cerebral I/R in rats. Furthermore, YZFDF could inactivate pyroptosis signaling via inhibiting caspase-1/11 activation and gasdermin D cleavage, ameliorate glial pyroptosis and neuroinflammation, protect against BBB collapse and AQP-4 depolarization, prevent Aβ acute accumulation and Aβ1-42 oligomers formation, and reduce neuronal damage and increase neurons survival after reperfusion. Conclusion: Our study indicated that YZFDF could exert neuroprotective effects on cerebral I/R injury and prevent Aβ acute accumulation in the brain after cerebral I/R associated with inhibiting neuroinflammation-related pyroptosis and BBB–glymphatic dysfunctions.
Collapse
Affiliation(s)
- Zhongkuan Lyu
- Geriatrics Department of Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiyue Li
- Geriatrics Department of Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhonghai Yu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuanjin Chan
- Geriatrics Department of Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Lei Fu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yaming Li
- Geriatrics Department of Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Chunyan Zhang
- International Medical Center of Traditional Chinese Medicine, Haikou Hospital of Traditional Chinese Medicine, Haikou, China
| |
Collapse
|
29
|
Min JH, Cho HJ, Yi YS. A novel mechanism of Korean red ginseng-mediated anti-inflammatory action via targeting caspase-11 non-canonical inflammasome in macrophages. J Ginseng Res 2021; 46:675-682. [PMID: 36090677 PMCID: PMC9459075 DOI: 10.1016/j.jgr.2021.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 01/05/2023] Open
Abstract
Background Korean Red Ginseng (KRG) was reported to play an anti-inflammatory role, however, previous studies largely focused on the effects of KRG on priming step, the inflammation-preparing step, and the anti-inflammatory effect of KRG on triggering, the inflammation-activating step has been poorly understood. This study demonstrated anti-inflammatory role of KRG in caspase-11 non-canonical inflammasome activation in macrophages during triggering of inflammatory responses. Methods Caspase-11 non-canonical inflammasome-activated J774A.1 macrophages were established by priming with Pam3CSK4 and triggering with lipopolysaccharide (LPS). Cell viability and pyroptosis were examined by MTT and lactate dehydrogenase (LDH) assays. Nitric oxide (NO)-inhibitory effect of KRG was assessed using a NO production assay. Expression and proteolytic cleavage of proteins were examined by Western blotting analysis. In vivo anti-inflammatory action of KRG was evaluated with the LPS-injected sepsis model in mice. Results KRG reduced LPS-stimulated NO production in J774A.1 cells and suppressed pyroptosis and IL-1β secretion in caspase-11 non-canonical inflammasome-activated J774A.1 cells. Mechanistic studies demonstrated that KRG suppressed the direct interaction between LPS and caspase-11 and inhibited proteolytic processing of both caspase-11 and gasdermin D in caspase-11 non-canonical inflammasome-activated J774A.1 cells. Furthermore, KRG significantly ameliorated LPS-mediated lethal septic shock in mice. Conclusion The results demonstrate a novel mechanism of KRG-mediated anti-inflammatory action that operates through targeting the caspase-11 non-canonical inflammasome at triggering step of macrophage-mediated inflammatory response.
Collapse
|
30
|
The Antitriple Negative Breast cancer Efficacy of Spatholobus suberectus Dunn on ROS-Induced Noncanonical Inflammasome Pyroptotic Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5187569. [PMID: 34659633 PMCID: PMC8514942 DOI: 10.1155/2021/5187569] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer (BCa) is the leading cause of women's death worldwide; among them, triple-negative breast cancer (TNBC) is one of the most troublesome subtypes with easy recurrence and great aggressive properties. Spatholobus suberectus Dunn has been used in the clinic of Chinese society for hundreds of years. Shreds of evidence showed that Spatholobus suberectus Dunn has a favorable outcome in the management of cancer. However, the anti-TNBC efficacy of Spatholobus suberectus Dunn percolation extract (SSP) and its underlying mechanisms have not been fully elucidated. Hence, the present study is aimed at evaluating the anti-TNBC potential of SSP both in vitro and in vivo, through the cell viability, morphological analysis of MDA-MB-231, LDH release assay, ROS assay, and the tests of GSH aborted pyroptotic noninflammasome signaling pathway. Survival analysis using the KM Plotter and TNM plot database exhibited the inhibition of transcription levels of caspase-4 and 9 related to low relapse-free survival in patients with BCa. Based on the findings, SSP possesses anti-TNBC efficacy that relies on ROS-induced noncanonical inflammasome pyroptosis in cancer cells. In this study, our preclinical evidence is complementary to the preceding clinic of Chinese society; studies on the active principles of SPP remain underway in our laboratory.
Collapse
|
31
|
Yi YS. New mechanisms of ginseng saponin-mediated anti-inflammatory action via targeting canonical inflammasome signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114292. [PMID: 34089812 DOI: 10.1016/j.jep.2021.114292] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is an ethnopharmacological herbal plant in Asian countries, particularly in Korea, China, and Japan. Ginseng saponins, including ginsenosides, are major active components in ginseng and have been demonstrated to have numerous pharmacological effects on various human diseases. AIM OF THE REVIEW Many previous studies investigating the anti-inflammatory effect of ginseng saponins have mostly focused on the 'priming' step rather than the 'triggering' step. This review aims to discuss the studies investigating an inhibitory role of ginseng saponins in inflammasome activation of the triggering step. MATERIALS AND METHODS The literature was explored using the search strings, such as "ginseng saponins and inflammasomes" and "ginsenosides and inflammasomes" in several resources, such as PubMed, Google Scholar, and Scopus databases. RESULTS Various ginseng saponins of Panax ginseng, Panax japonicas, and Panax quinquefolius alleviated inflammatory responses and diseases by inhibiting the nucleotide-binding oligomerization domain-like receptor (NLR) P3 (NLRP3) inflammasome activation. Also, ginseng saponin, Rg1 of Panax ginseng alleviated neuroinflammation and diseases by inhibiting NLRP1 inflammasome activation. Finally, ginseng saponins, Rh1 and Rg3 in Korea red ginseng (KRG) of Panax ginseng ameliorated sepsis by inhibiting absent in melanoma 2 (AIM2) inflammasome activation. CONCLUSION The studies discussed in this review provide insight into the new paradigm of the ginseng saponins as the promising anti-inflammatory agents that could be ethnopharmacologically used to prevent and treat inflammatory and inflammation-induced disorders via targeting inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| |
Collapse
|
32
|
Functional Interplay between Methyltransferases and Inflammasomes in Inflammatory Responses and Diseases. Int J Mol Sci 2021; 22:ijms22147580. [PMID: 34299198 PMCID: PMC8306412 DOI: 10.3390/ijms22147580] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023] Open
Abstract
An inflammasome is an intracellular protein complex that is activated in response to a pathogenic infection and cellular damage. It triggers inflammatory responses by promoting inflammatory cell death (called pyroptosis) and the secretion of pro-inflammatory cytokines, interleukin (IL)-1β and IL-18. Many types of inflammasomes have been identified and demonstrated to play a central role in inducing inflammatory responses, leading to the onset and progression of numerous inflammatory diseases. Methylation is a biological process by which methyl groups are transferred from methyl donors to proteins, nucleic acids, and other cellular molecules. Methylation plays critical roles in various biological functions by modulating gene expression, protein activity, protein localization, and molecular stability, and aberrant regulation of methylation causes deleterious outcomes in various human diseases. Methylation is a key determinant of inflammatory responses and diseases. This review highlights the current understanding of the functional relationship between inflammasome regulation and methylation of cellular molecules in inflammatory responses and diseases.
Collapse
|
33
|
Kuc-Ciepluch D, Ciepluch K, Arabski M. Gasdermin family proteins as a permeabilization factor
of cell membrane in pyroptosis process. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.8985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The type of cell death, i.e. apoptosis, autophagy, necrosis or pyroptosis, depends on the inducing
factor and the phase of the cell cycle. The main role in immunological response to microorganisms
is played by a process called pyroptosis. Pyroptosis induces various types of inflammatory
factors in response to molecular patterns associated with pathogens, e.g., bacterial lipopolysaccharide
in the canonical or non-canonical pathway depending on the type of caspases involved.
In pyroptosis, the gasdermin D protein belonging to the gasdermin protein family (A, B, C, D, E
and DFNB59) plays an important role, which is characterized by specific tissue gene expression
mainly in epithelial cells, skin and the digestive system and is responsible for regulating the proliferation
and differentiation of cells and is responsible for inhibiting or developing cancers in
various organs. The GSDM family is responsible for the formation of pores in the cell membrane,
enabling the secretion of proinflammatory cytokines (IL-1β and IL-18) involved in initiating inflammatory
response pathways by recruiting and activating immune cells at the site of infection.
The gasdermin D protein plays an essential role in the non-canonical pyroptosis process, whose
N-terminal forming pores in the cell membrane leads to edema, osmotic lysis and, consequently,
to the death of the infected cell.
Collapse
Affiliation(s)
- Dorota Kuc-Ciepluch
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| | - Karol Ciepluch
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| | - Michał Arabski
- Zakład Biologii Medycznej, Instytut Biologii, Uniwersytet Jana Kochanowskiego w Kielcach
| |
Collapse
|
34
|
Ogger PP, Silva JD, Aghapour M, Mahmutovic Persson I, Tulen C, Jurkowska R, Ubags ND. Early Career Members at the ERS Lung Science Conference 2020: metabolic alterations in lung ageing and disease. Breathe (Sheff) 2021; 16:200063. [PMID: 33447269 PMCID: PMC7792764 DOI: 10.1183/20734735.0063-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Every year, the European Respiratory Society (ERS) organises the Lung Science Conference (LSC) in Estoril, Portugal, to discuss basic and translational science. The topic of the LSC 2020 was “Metabolic alterations in lung ageing and disease”. In addition to an outstanding scientific programme, the LSC provides excellent opportunities for career development and inclusion of Early Career Members (ECMs). All scientific and poster sessions are chaired by an ECM who is paired with a senior faculty member to allow ECMs to become acquainted with session chairing. In addition, 40 travel bursaries are made available to abstract authors and all bursary recipients are invited to take part in a mentorship lunch. Moreover, there is a session organised by the Early Career Members Committee (ECMC) dedicated to career development. Here, we describe the scientific highlights of LSC 2020 for those who could not attend. The Lung Science Conference 2020 brought together leading experts in the field to discuss the latest cutting-edge science, as well as various career development opportunities for early career membershttps://bit.ly/2XZ5YGQ
Collapse
Affiliation(s)
- Patricia P Ogger
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Johnatas Dutra Silva
- Wellcome-Wolfson Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Mahyar Aghapour
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Irma Mahmutovic Persson
- Institution of Medical Radiation Physics, Dept of Translational Medicine, Lund University, Malmö, Sweden
| | - Christy Tulen
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Dept of Pharmacology and Toxicology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Epalinges, Switzerland
| |
Collapse
|
35
|
Flavonoids: Nutraceuticals for Rheumatic Diseases via Targeting of Inflammasome Activation. Int J Mol Sci 2021; 22:ijms22020488. [PMID: 33418975 PMCID: PMC7825303 DOI: 10.3390/ijms22020488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation, an innate immune response that prevents cellular damage caused by pathogens, consists of two successive mechanisms, namely priming and triggering. While priming is an inflammation-preparation step, triggering is an inflammation-activation step, and the central feature of triggering is the activation of inflammasomes and intracellular inflammatory protein complexes. Flavonoids are natural phenolic compounds predominantly present in plants, fruits, and vegetables and are known to possess strong anti-inflammatory activities. The anti-inflammatory activity of flavonoids has long been demonstrated, with the main focus on the priming mechanisms, while increasing numbers of recent studies have redirected the research focus on the triggering step, and studies have reported that flavonoids inhibit inflammatory responses and diseases by targeting inflammasome activation. Rheumatic diseases are systemic inflammatory and autoimmune diseases that primarily affect joints and connective tissues, and they are associated with numerous deleterious effects. Here, we discuss the emerging literature on the ameliorative role of flavonoids targeting inflammasome activation in inflammatory rheumatic diseases.
Collapse
|
36
|
Yi YS. Caspase-11 Noncanonical Inflammasome: A Novel Key Player in Murine Models of Neuroinflammation and Multiple Sclerosis. Neuroimmunomodulation 2021; 28:195-203. [PMID: 34044393 DOI: 10.1159/000516064] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/18/2021] [Indexed: 11/19/2022] Open
Abstract
Inflammasomes are intracellular protein complexes consisting of the pattern recognition receptors and inflammatory molecules in the inflamed cells. In response to various ligands, inflammasomes play a pivotal role to execute the inflammatory responses by inducing the pyroptosis and the secretion of pro-inflammatory cytokines, interleukin (IL)-1β, and IL-18. Unlike canonical inflammasomes, including NOD-like receptor family inflammasomes, such as NLRP1, NLRP3, NLRC4, and absence in melanoma 2 inflammasomes, noncanonical inflammasomes, such as mouse caspase-11 and human caspase-4/5 were recently discovered, and their roles in the inflammatory responses have been poorly understood. However, emerging studies have been successfully demonstrating the regulatory roles of these noncanonical inflammasomes on inflammatory responses and the pathogenesis of inflammatory/autoimmune diseases. This review summarizes and discusses the recent studies investigating the regulatory roles of the caspase-11 noncanonical inflammasome in neuroinflammation and the pathogenesis of multiple sclerosis (MS), which provides the insight for the validation of caspase-11 noncanonical inflammasome to develop novel and promising therapeutics for MS.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, Republic of Korea
| |
Collapse
|
37
|
Tupik JD, Coutermarsh-Ott SL, Benton AH, King KA, Kiryluk HD, Caswell CC, Allen IC. ASC-Mediated Inflammation and Pyroptosis Attenuates Brucella abortus Pathogenesis Following the Recognition of gDNA. Pathogens 2020; 9:E1008. [PMID: 33266295 PMCID: PMC7760712 DOI: 10.3390/pathogens9121008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/25/2022] Open
Abstract
Brucella abortus is a zoonotic pathogen that causes brucellosis. Because of Brucella's unique LPS layer and intracellular localization predominately within macrophages, it can often evade immune detection. However, pattern recognition receptors are capable of sensing Brucella pathogen-associated molecular patterns (PAMPS). For example, NOD-like receptors (NLRs) can form a multi-protein inflammasome complex to attenuate Brucella pathogenesis. The inflammasome activates IL-1β and IL-18 to drive immune cell recruitment. Alternatively, inflammasome activation also initiates inflammatory cell death, termed pyroptosis, which augments bacteria clearance. In this report, we assess canonical and non-canonical inflammasome activation following B. abortus infection. We conducted in vivo studies using Asc-/- mice and observed decreased mouse survival, immune cell recruitment, and increased bacteria load. We also conducted studies with Caspase-11-/- mice and did not observe any significant impact on B. abortus pathogenesis. Through mechanistic studies using Asc-/- macrophages, our data suggests that the protective role of ASC may result from the induction of pyroptosis through a gasdermin D-dependent mechanism in macrophages. Additionally, we show that the recognition of Brucella is facilitated by sensing the PAMP gDNA rather than the less immunogenic LPS. Together, these results refine our understanding of the role that inflammasome activation and pyroptosis plays during brucellosis.
Collapse
Affiliation(s)
- Juselyn D. Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Sheryl L. Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Angela H. Benton
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Kellie A. King
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Hanna D. Kiryluk
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Clayton C. Caswell
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (J.D.T.); (S.L.C.-O.); (A.H.B.); (K.A.K.); (H.D.K.); (C.C.C.)
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
38
|
Peng L, Wen L, Shi QF, Gao F, Huang B, Meng J, Hu CP, Wang CM. Scutellarin ameliorates pulmonary fibrosis through inhibiting NF-κB/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Cell Death Dis 2020; 11:978. [PMID: 33188176 PMCID: PMC7666141 DOI: 10.1038/s41419-020-03178-2] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is featured with inflammation and extensive lung remodeling caused by overloaded deposition of extracellular matrix. Scutellarin is the major effective ingredient of breviscapine and its anti-inflammation efficacy has been reported before. Nevertheless, the impact of scutellarin on IPF and the downstream molecular mechanism remain unclear. In this study, scutellarin suppressed BLM-induced inflammation via NF-κB/NLRP3 pathway both in vivo and in vitro. BLM significantly elevated p-p65/p65 ratio, IκBα degradation, and levels of NLRP3, caspase-1, caspase-11, ASC, GSDMDNterm, IL-1β, and IL-18, while scutellarin reversed the above alterations except for that of caspase-11. Scutellarin inhibited BLM-induced epithelial-mesenchymal transition (EMT) process in vivo and in vitro. The expression levels of EMT-related markers, including fibronectin, vimentin, N-cadherin, matrix metalloproteinase 2 (MMP-2) and MMP-9, were increased in BLM group, and suppressed by scutellarin. The expression level of E-cadherin showed the opposite changes. However, overexpression of NLRP3 eliminated the anti-inflammation and anti-EMT functions of scutellarin in vitro. In conclusion, scutellarin suppressed inflammation and EMT in BLM-induced pulmonary fibrosis through NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Ling Peng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Li Wen
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Qing-Feng Shi
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Feng Gao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Bin Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Jie Meng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Cheng-Ping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China.
| | - Chang-Ming Wang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China.
| |
Collapse
|
39
|
Rodríguez-Ruiz L, Lozano-Gil JM, Lachaud C, Mesa-Del-Castillo P, Cayuela ML, García-Moreno D, Pérez-Oliva AB, Mulero V. Zebrafish Models to Study Inflammasome-Mediated Regulation of Hematopoiesis. Trends Immunol 2020; 41:1116-1127. [PMID: 33162327 DOI: 10.1016/j.it.2020.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
Hematopoiesis is a complex process through which immature bone marrow precursor cells mature into all types of blood cells. Although the association of hematopoietic lineage bias (including anemia and neutrophilia) with chronic inflammatory diseases has long been appreciated, the causes involved are obscure. Recently, cytosolic multiprotein inflammasome complexes were shown to activate inflammatory and immune responses, and directly regulate hematopoiesis in zebrafish models; this was deemed to occur via cleavage and inactivation of the master erythroid transcription factor GATA1. Herein summarized are the zebrafish models that are currently available to study this unappreciated role of inflammasome-mediated regulation of hematopoiesis. Novel putative therapeutic strategies, for the treatment of hematopoietic alterations associated with chronic inflammatory diseases in humans, are also proposed.
Collapse
Affiliation(s)
- Lola Rodríguez-Ruiz
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30100 Murcia, Spain
| | - Juan M Lozano-Gil
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30100 Murcia, Spain
| | - Christophe Lachaud
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Pablo Mesa-Del-Castillo
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30120 Murcia, Spain
| | - María L Cayuela
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30120 Murcia, Spain
| | - Diana García-Moreno
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30100 Murcia, Spain.
| | - Ana B Pérez-Oliva
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30100 Murcia, Spain.
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, IMIB-Arrixaca, Centro de Investigación Biomédica en Red de Enfermedades Raras, 30100 Murcia, Spain.
| |
Collapse
|
40
|
Ghoneim MES, Abdallah DM, Shebl AM, El-Abhar HS. The interrupted cross-talk of inflammatory and oxidative stress trajectories signifies the effect of artesunate against hepatic ischemia/reperfusion-induced inflammasomopathy. Toxicol Appl Pharmacol 2020; 409:115309. [PMID: 33130049 DOI: 10.1016/j.taap.2020.115309] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
The antimalarial drug artesunate (Art) has proven its beneficial effects against ischemia/reperfusion (I/R) injury in diverse organs, but its potential role against hepatic I/R is still obscure. This study, hence, examined whether treatment with Art alone or in combination with rapamycin (Rapa), an mTOR inhibitor, can ameliorate hepatic I/R injury via targeting the NLRP3 inflammasome signaling pathway. Rats were divided into hepatic sham- and I/R-operated rats. The latter were either left untreated (I/R group) or treated with Art, Rapa, or their combination. On the molecular level, all treatment regimens succeeded to hinder inflammasome assembly and activation, assessed as NLRP3, ASC, cleaved caspase-1, caspase-11, N-terminal cleaved gasdermin-D (GSDMD-N), IL-1β, and IL-18. This effect was associated by the inhibition in the harmful signaling pathways HMGB1/RAGE and TLR4/MyD88/TRAF6 to inactivate the transcription factor NF-κB and the production of its pro-inflammatory cytokines IL-1β, IL-18, IL-6, and TNF-α. Additionally, this effect entailed the inhibition of ICAM-1/MPO/ROS cascade, which in turn hampered cell demise induced by apoptosis, manifested as correction of the imbalanced Bcl2/Bax, as well as pyroptosis (LDH, cleaved caspase-1, caspase-11, GSDMD-N, IL-1β, and IL-18), and necrosis. The corrected pathways were reflected on the improved liver function (serum ALT, AST, and LDH) and microscopical hepatic architecture. Noteworthy, the effect of Art on all parameters exceeded significantly that of Rapa and even improved the effect of the latter in the combination group. In conclusion, our results suggest novel roles for Art in abating functional and structural I/R-induced hepatic abnormalities via several traversing cross-talking pathways that succeeded to abate NLRP3 inflammasome and cell death.
Collapse
Affiliation(s)
- Mai El-Sayed Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt.
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., 11562 Cairo, Egypt.
| | | | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), 11835 Cairo, Egypt
| |
Collapse
|
41
|
Sanz P, Garcia-Gimeno MA. Reactive Glia Inflammatory Signaling Pathways and Epilepsy. Int J Mol Sci 2020; 21:ijms21114096. [PMID: 32521797 PMCID: PMC7312833 DOI: 10.3390/ijms21114096] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation and epilepsy are interconnected. Brain inflammation promotes neuronal hyper-excitability and seizures, and dysregulation in the glia immune-inflammatory function is a common factor that predisposes or contributes to the generation of seizures. At the same time, acute seizures upregulate the production of pro-inflammatory cytokines in microglia and astrocytes, triggering a downstream cascade of inflammatory mediators. Therefore, epileptic seizures and inflammatory mediators form a vicious positive feedback loop, reinforcing each other. In this work, we have reviewed the main glial signaling pathways involved in neuroinflammation, how they are affected in epileptic conditions, and the therapeutic opportunities they offer to prevent these disorders.
Collapse
Affiliation(s)
- Pascual Sanz
- Instituto de Biomedicina de Valencia (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Jaime Roig 11, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-963391779; Fax: +34-963690800
| | - Maria Adelaida Garcia-Gimeno
- Department of Biotechnology, Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural (ETSIAMN), Universitat Politècnica de València, 46022 Valencia, Spain;
| |
Collapse
|
42
|
LOMIX, a Mixture of Flaxseed Linusorbs, Exerts Anti-Inflammatory Effects through Src and Syk in the NF-κB Pathway. Biomolecules 2020; 10:biom10060859. [PMID: 32512905 PMCID: PMC7356372 DOI: 10.3390/biom10060859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Although flax (Linum usitatissimum L.) has long been used as Ayurvedic medicine, its anti-inflammatory role is still unclear. Therefore, we aimed to investigate the anti-inflammatory role of a linusorb mixture (LOMIX) recovered from flaxseed oil. Effects of LOMIX on inflammation and its mechanism of action were examined using several in vitro assays (i.e., NO production, real-time PCR analysis, luciferase-reporter assay, Western blot analysis, and kinase assay) and in vivo analysis with animal inflammation models as well as acute toxicity test. Results: LOMIX inhibited NO production, cell shape change, and inflammatory gene expression in stimulated RAW264.7 cells through direct targeting of Src and Syk in the NF-κB pathway. In vivo study further showed that LOMIX alleviated symptoms of gastritis, colitis, and hepatitis in murine model systems. In accordance with in vitro results, the in vivo anti-inflammatory effects were mediated by inhibition of Src and Syk. LOMIX was neither cytotoxic nor did it cause acute toxicity in mice. In addition, it was found that LOB3, LOB2, and LOA2 are active components included in LOMIX, as assessed by NO assay. These in vitro and in vivo results suggest that LOMIX exerts an anti-inflammatory effect by inhibiting the inflammatory responses of macrophages and ameliorating symptoms of inflammatory diseases without acute toxicity and is a promising anti-inflammatory medication for inflammatory diseases.
Collapse
|
43
|
Feng YS, Tan ZX, Wang MM, Xing Y, Dong F, Zhang F. Inhibition of NLRP3 Inflammasome: A Prospective Target for the Treatment of Ischemic Stroke. Front Cell Neurosci 2020; 14:155. [PMID: 32581721 PMCID: PMC7283578 DOI: 10.3389/fncel.2020.00155] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the major devastating diseases with no effective medical therapeutics. Because of the high rate of disability and mortality among stroke patients, new treatments are urgently required to decrease brain damage following a stroke. In recent years, the inflammasome is a novel breakthrough point that plays an important role in the stroke, and the inhibition of inflammasome may be an effective method for stroke treatment. Briefly, inflammasome is a multi-protein complex that causes activation of caspase-1 and subsequent production of pro-inflammatory factors including interleukin (IL)-18 and IL-1β. Among them, the NLRP3 inflammasome is the most typical inflammasome, which can detect cell damage and mediate inflammatory response to tissue damage in ischemic stroke. The NLRP3 inflammasome has become a key mediator of post-ischemic inflammation, leading to a cascade of inflammatory reactions and cell death eventually. Thus, NLRP3 inflammasome is an ideal therapeutic target due to its important role in the inflammatory response after ischemic stroke. In this mini review article, we will summarize the structure, assembly, and regulation of NLRP3 inflammasome, the role of NLRP3 inflammasome in ischemic stroke, and several treatments targeting NLRP3 inflammasome in ischemic stroke. The further understanding of the mechanism of NLRP3 inflammasome in patients with ischemic stroke will provide novel targets for the treatment of cerebral ischemic stroke patients.
Collapse
Affiliation(s)
- Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Man-Man Wang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Provincial Orthopedic Biomechanics Key Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
44
|
Caspase-11 Non-Canonical Inflammasome: Emerging Activator and Regulator of Infection-Mediated Inflammatory Responses. Int J Mol Sci 2020; 21:ijms21082736. [PMID: 32326466 PMCID: PMC7215657 DOI: 10.3390/ijms21082736] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022] Open
Abstract
Inflammation is a body’s protective mechanism to eliminate invading pathogens and cellular damaging signals. The inflammatory response consists of two main consecutive steps—a priming step preparing the inflammatory responses and a triggering step boosting the inflammatory responses. The main feature of the triggering step is the activation of the inflammasome, an intracellular multiprotein complex facilitating the inflammatory responses. The regulatory roles of ‘canonical’ inflammasomes in the inflammatory responses and diseases have been largely investigated, so far. New types of inflammasomes have been recently discovered and named as ‘non-canonical’ inflammasomes since their roles to induce inflammatory responses are similar to those of canonical inflammasomes, however, the stimulating ligands and the underlying mechanisms are different. Therefore, a growing number of studies have actively investigated the novel roles of non-canonical inflammasomes in inflammatory responses and diseases. This review summarizes and discusses the recent studies exploring the regulatory roles of caspase-11 non-canonical inflammasome during the inflammatory responses and provides insight into the development of novel therapeutics for infectious and inflammatory diseases by targeting caspase-11 non-canonical inflammasome.
Collapse
|
45
|
Wada N, Yamada H, Motoyama S, Saburi M, Sugimoto T, Kubota H, Miyawaki D, Wakana N, Kami D, Ogata T, Matoba S. Maternal high-fat diet exaggerates diet-induced insulin resistance in adult offspring by enhancing inflammasome activation through noncanonical pathway of caspase-11. Mol Metab 2020; 37:100988. [PMID: 32272237 PMCID: PMC7210595 DOI: 10.1016/j.molmet.2020.100988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Maternal high-fat diet (HFD) has been shown to promote the development of insulin resistance (IR) in adult offspring; however, the underlying mechanisms remain unclear. Methods Eight-week-old female wild-type mice (C57BL/6) were fed either an HFD or a normal diet (ND), one week prior to mating, and the diet was continued throughout gestation and lactation. Eight-week-old male offspring of both groups were fed an HFD for 8 weeks. Results Offspring of HFD-fed dams (O-HFD) exhibited significantly impaired insulin sensitivity compared with the offspring of ND-fed dams (O-ND). The adipocyte size of the eWAT increased significantly in O-HFD and was accompanied by abundant crown-like structures (CLSs), as well as a higher concentration of interleukin 1β (IL-1β) in the eWAT. Treatment with an inflammasome inhibitor, MCC950, completely abrogated the enhanced IR in O-HFD. However, ex vivo caspase-1 activity in eWAT revealed no difference between the two groups. In contrast, noncanonical inflammasome activation of caspase-11 was significantly augmented in O-HFD compared with O-ND, suggesting that membrane pore formation, but not cleavage of pro-IL-1β by caspase-1, is augmented in O-HFD. To examine the membrane pore formation, we performed metabolic activation of bone marrow-derived macrophages (BMDMs). The percentage of pore formation assessed by ethidium bromide staining was significantly higher in BMDMs of O-HFD, accompanied by an enhanced active caspase-11 expression. Consistently, the concentration of IL-1β in culture supernatants was significantly higher in the BMDMs from O-HFD than those from O-ND. Conclusions These findings demonstrate that maternal HFD exaggerates diet-induced IR in adult offspring by enhancing noncanonical caspase-11-mediated inflammasome activation. Maternal HFD increases the vulnerability to HFD-induced IR in adult offspring. Maternal HFD augments inflammasome activation in eWAT in adult offspring. Treatment with an inflammasome inhibitor abrogates IR in offspring of HFD-fed dam. Maternal HFD augments the noncanonical inflammasome activation pathway of caspase-11. Maternal HFD augments IL-1β release from BMDMs by enhancing membrane pore formation.
Collapse
Affiliation(s)
- Naotoshi Wada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Yamada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Shinichiro Motoyama
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Makoto Saburi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Sugimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Kubota
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Miyawaki
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Wakana
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
46
|
Huang S, Che J, Chu Q, Zhang P. The Role of NLRP3 Inflammasome in Radiation-Induced Cardiovascular Injury. Front Cell Dev Biol 2020; 8:140. [PMID: 32226786 PMCID: PMC7080656 DOI: 10.3389/fcell.2020.00140] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The increasing risk of long-term adverse effects from radiotherapy on the cardiovascular structure is receiving increasing attention. However, the mechanisms underlying this increased risk remain poorly understood. Recently, the nucleotide-binding domain and leucine-rich-repeat-containing family pyrin 3 (NLRP3) inflammasome was suggested to play a critical role in radiation-induced cardiovascular injury. However, the relationship between ionizing radiation and the NLRP3 inflammasome in acute and chronic inflammation is complex. We reviewed literature detailing pathological changes and molecular mechanisms associated with radiation-induced damage to the cardiovascular structure, with a specific focus on NLRP3 inflammasome-related cardiovascular diseases. We also summarized possible therapeutic strategies for the prevention of radiation-induced heart disease (RIHD).
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Che
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Jo M, Yi YS, Cho JY. Archidendron lucidum Exerts Anti-Inflammatory Effects by Targeting PDK1 in the NF- κB Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:429-444. [PMID: 32160757 DOI: 10.1142/s0192415x20500226] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pharmacological activities of some Leguminosae family members were reported. Pharmacological activities of Archidendron lucidum, a Leguminosae family member have never been explored. Therefore, this study investigated anti-inflammatory effects of an Archidendron lucidum methanol extract (Al-ME). In this study, anti-inflammatory effects of Al-ME were investigated in LPS-stimulated RAW264.7 cells and HCl/EtOH-induced gastritis mice by MTT assay, nitric oxide (NO) production assay, semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), luciferase reporter assay, and Western blotting. High-performance liquid chromatography (HPLC) analysis identified ethnopharmacological compounds in Al-ME. Al-ME inhibited NO production without cytotoxicity in peritoneal macrophages and RAW264.7 cells stimulated with LPS or Pam3CSK4. Al-ME downregulated mRNA expression of inflammatory genes (inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2)) and pro-inflammatory cytokines (tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6). Al-ME exerted anti-inflammatory activity in LPS-stimulated RAW264.7 cells by inhibiting nuclear factor-kappa B (NF-κB) signaling pathway. HPLC analysis identified quercetin, luteolin, and kaempferol as major anti-inflammatory components in Al-ME. Al-ME ameliorated HCl/EtOH-induced gastritis symptoms in mice by suppressing iNOS and IL-6 mRNA expressions and IκBα phosphorylation. Therefore, these results suggest that Al-ME exhibited anti-inflammatory activity by targeting NF-κB signaling pathway, implying that Al-ME could be potent anti-inflammatory medications to prevent and treat inflammatory diseases.
Collapse
Affiliation(s)
- Minkyeong Jo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Young-Su Yi
- Department of Life Science, Kyonggi University, Suwon 16227, Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
48
|
Yin N, Gao Q, Tao W, Chen J, Bi J, Ding F, Wang Z. Paeoniflorin relieves LPS-induced inflammatory pain in mice by inhibiting NLRP3 inflammasome activation via transient receptor potential vanilloid 1. J Leukoc Biol 2020; 108:229-241. [PMID: 32083340 DOI: 10.1002/jlb.3ma0220-355r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
LPS has been widely used to induce inflammatory pain, attributing to production of inflammatory cytokines and sensitization of nociceptors. Paeoniflorin (PF) possesses anti-nociceptive property, but its effect on LPS-induced inflammatory pain has not been investigated. In this study, we aimed to investigate the analgesic effect of PF on an inflammatory pain mouse model and explore the underlying mechanisms. LPS-induced inflammatory pain model was established in C57BL/6J mice after PF treatment. Then, thermal hyperalgesia, neutrophil infiltration, inflammatory cytokine production, intracellular Ca2+ levels, PKC activity, transient receptor potential vanilloid 1 (TRPV-1) expression, NF-κB transcription, and NLPR3 inflammasome activation were assessed by thermal withdrawal latency, histopathology, ELISA, intracellular Ca2+ concentration, immunohistochemistry, and Western blot, separately. PF significantly relieved inflammatory pain and paw edema in mice with LPS-induced inflammatory pain. Additionally, PF inhibited neutrophil infiltration, inflammatory cytokine production (IL-1β, TNF-α, and IL-6), intracellular Ca2+ levels, and PKC activity as well as suppressed TRPV-1 expression, NF-κB transcription, and NLPR3 inflammasome activation in the footpad tissue samples. Importantly, capsaicin (TRPV-1 agonists) obviously reversed the pain-relieving effect of PF, suggesting the involvement of TRPV-1 in the analgesic activity of PF. Our results indicated PF ameliorated LPS-induced inflammation and pain in mice by inhibiting TRPV-1-mediated NLRP3 inflammasome activation. These findings suggest that PF can be as a potential pharmacological agent for inflammatory pain and thus deserves more attention and further investigation.
Collapse
Affiliation(s)
- Nina Yin
- Department of Anatomy, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Qinghua Gao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Wenting Tao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiaojiao Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Bi
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Fengmin Ding
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhigang Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
49
|
Yi Y. Functional crosstalk between non-canonical caspase-11 and canonical NLRP3 inflammasomes during infection-mediated inflammation. Immunology 2020; 159:142-155. [PMID: 31630388 PMCID: PMC6954705 DOI: 10.1111/imm.13134] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a part of the body's immune response for protection against pathogenic infections and other cellular damages; however, chronic inflammation is a major cause of various diseases. One key step in the inflammatory response is the activation of inflammasomes, intracellular protein complexes comprising pattern recognition receptors and other inflammatory molecules. The role of the NLRP3 inflammasome in inflammatory responses has been extensively investigated; however, the caspase-11 inflammasome has been recently identified and has been classified as a 'non-canonical' inflammasome, and emerging studies have highlighted its role in inflammatory responses. Because the ligands and the mechanisms for the activation of these two inflammasomes are different, studies to date have separately described their roles, although recent studies have reported the functional cooperation between these two inflammasomes during an inflammatory response. This review discusses the studies investigating the functional crosstalk between non-canonical caspase-11 and canonical NLRP3 inflammasomes in the context of inflammatory responses; moreover, it provides insight for the development of novel anti-inflammatory therapeutics to prevent and treat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Young‐Su Yi
- Department of Pharmaceutical and Biomedical EngineeringCheongju UniversityCheongjuKorea
| |
Collapse
|
50
|
Ko YJ, Lee JW, Yang EJ, Jang N, Park J, Jeon YK, Yu JW, Cho NH, Kim HS, Chan Kwon I. Non-invasive in vivo imaging of caspase-1 activation enables rapid and spatiotemporal detection of acute and chronic inflammatory disorders. Biomaterials 2020; 226:119543. [DOI: 10.1016/j.biomaterials.2019.119543] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022]
|