1
|
Li H, Pu J, Yang D, Liu L, Hu Y, Yang S, Wang B. GSDMD protects intestinal epithelial cells against bacterial infections through its N-terminal activity impacting intestinal immune homeostasis. J Biomed Res 2024; 38:1-12. [PMID: 38807373 PMCID: PMC11629157 DOI: 10.7555/jbr.38.20240041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The intestinal mucosal barrier serves as a vital guardian for gut health, maintaining a delicate equilibrium between gut microbiota and host immune homeostasis. Recent studies have found the intricate roles of Gasdermin D (GSDMD), a key executioner of pyroptosis downstream of the inflammasome, within the intestine, including controlling colitis in intestinal macrophage and the regulatory function in goblet cell mucus secretion. Thus, the exact role and nature of GSDMD's regulatory function in maintaining intestinal immune homeostasis and defending against pathogens remain elucidation. Here, we uncover that GSDMD plays a key role in defending against intestinal Citrobacter rodentium infection, with high expression in intestinal epithelial and lamina propria myeloid cells. Our results show that GSDMD specifically acts in intestinal epithelial cells to fight the infection, independently of its effects on antimicrobial peptides or mucin secretion. Instead, the resistance is mediated through GSDMD's N-terminal fragments, highlighting its importance in intestinal immunity. However, the specific underlying mechanism of GSDMD N-terminal activity in protection against intestinal bacterial infections still needs further study to clarify in the future.
Collapse
Affiliation(s)
- Honghui Li
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jie Pu
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Dongxue Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lu Liu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
2
|
Li Y, Ling P, Li Y, Wang Y, Li G, Qiu C, Wang J, Gong K. miR-138-5p ameliorates intestinal barrier disruption caused by acute superior mesenteric vein thrombosis injury by inhibiting the NLRP3/HMGB1 axis. PeerJ 2024; 12:e16692. [PMID: 38406274 PMCID: PMC10893868 DOI: 10.7717/peerj.16692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/28/2023] [Indexed: 02/27/2024] Open
Abstract
Background Acute superior mesenteric venous thrombosis (ASMVT) decreases junction-associated protein expression and intestinal epithelial cell numbers, leading to intestinal epithelial barrier disruption. Pyroptosis has also recently been found to be one of the important causes of mucosal barrier defects. However, the role and mechanism of pyroptosis in ASMVT are not fully understood. Methods Differentially expressed microRNAs (miRNAs) in the intestinal tissues of ASMVT mice were detected by transcriptome sequencing (RNA-Seq). Gene expression levels were determined by RNA extraction and reverse transcription-quantitative PCR (RT-qPCR). Western blot and immunofluorescence staining analysis were used to analyze protein expression. H&E staining was used to observe the intestinal tissue structure. Cell Counting Kit-8 (CCK-8) and fluorescein isothiocyanate/propidine iodide (FITC/PI) were used to detect cell viability and apoptosis, respectively. Dual-luciferase reporter assays prove that miR-138-5p targets NLRP3. Results miR-138-5p expression was downregulated in ASMVT-induced intestinal tissues. Inhibition of miR-138-5p promoted NLRP3-related pyroptosis and destroyed tight junctions between IEC-6 cells, ameliorating ASMVT injury. miR-138-5p targeted to downregulate NLRP3. Knockdown of NLRP3 reversed the inhibition of proliferation, apoptosis, and pyroptosis and the decrease in tight junction proteins caused by suppression of miR-138-5p; however, this effect was later inhibited by overexpressing HMGB1. miR-138-5p inhibited pyroptosis, promoted intestinal epithelial tight junctions and alleviated ASMVT injury-induced intestinal barrier disruption via the NLRP3/HMGB1 axis.
Collapse
Affiliation(s)
- Yuejin Li
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Ping Ling
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yu Li
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yongzhi Wang
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Guosan Li
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Changtao Qiu
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jianghui Wang
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Kunmei Gong
- The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
3
|
Shadab A, Mahjoor M, Abbasi-Kolli M, Afkhami H, Moeinian P, Safdarian AR. Divergent functions of NLRP3 inflammasomes in cancer: a review. Cell Commun Signal 2023; 21:232. [PMID: 37715239 PMCID: PMC10503066 DOI: 10.1186/s12964-023-01235-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023] Open
Abstract
The cancer is a serious health problem, which is The cancer death rate (cancer mortality) is 158.3 per 100,000 men and women per year (based on 2013-2017 deaths). Both clinical and translational studies have demonstrated that chronic inflammation is associated with Cancer progression. However, the precise mechanisms of inflammasome, and the pathways that mediate this phenomenon are not fully characterized. One of the most recently identified signaling pathways, whose activation seems to affect many metabolic disorders, is the "inflammasome" a multiprotein complex composed of NLRP3 (nucleotide-binding domain and leucine-rich repeat protein 3), ASC (apoptosis associated speck-like protein containing a CARD), and procaspase-1. NLRP3 inflammasome activation leads to the processing and secretion of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18. The goal of this paper is to review new insights on the effects of the NLRP3 inflammasome activation in the complex mechanisms of crosstalk between different organs, for a better understanding of the role of chronic inflammation in cancer pathogenesis. We will provide here a perspective on the current research on NLRP3 inflammasome, which may represent an innovative therapeutic target to reverse the malignancy condition consequences of the inflammation. Video Abstract.
Collapse
Affiliation(s)
- Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abbasi-Kolli
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Parisa Moeinian
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Reza Safdarian
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno TACT), Universal Scientific Education and Research Network (USERN) Chicago, Chicago, IL, USA.
- Department of Immunology and Microbiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran.
| |
Collapse
|
4
|
JC2-11, a benzylideneacetophenone derivative, attenuates inflammasome activation. Sci Rep 2022; 12:22484. [PMID: 36577816 PMCID: PMC9797494 DOI: 10.1038/s41598-022-27129-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
Dysregulation of inflammasome activation induces chronic and excess inflammation resulting in several disorders, such as metabolic disorders and cancers. Thus, screening for its regulator derived from natural materials has been conducted progressively. JC2-11 (JC) was designed to enhance the antioxidant activity based on a chalcone, which is abundant in edible plants and a precursor of flavonoids. This study examined the effects of JC on inflammasome activation in human and murine macrophages. JC inhibited the secretion of interleukin (IL)-1β and lactate dehydrogenases, and the cleavage of caspase-1 and gasdermin D in response to the tested activators (i.e., NLRP3, NLRC4, AIM2, and non-canonical inflammasome triggers). In addition, JC attenuated IL-1β secretion from lipopolysaccharide (LPS)-injected mice, an inflammasome-mediating disease model. Mechanistically, JC blocked the expression of the inflammasome components during the priming step of the inflammasome, and interrupted the production of mitochondrial reactive oxygen species. In addition, JC inhibited the activity of caspase-1. In conclusion, JC may be a candidate pan-inflammasome inhibitor.
Collapse
|
5
|
Ahn H, Lee G, Lee GS. Lower Temperatures Exacerbate NLRP3 Inflammasome Activation by Promoting Monosodium Urate Crystallization, Causing Gout. Cells 2021; 10:cells10081919. [PMID: 34440688 PMCID: PMC8394355 DOI: 10.3390/cells10081919] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Gout is a recurrent and chronic form of arthritis caused by the deposition of monosodium urate (MSU) crystals in the joints. Macrophages intake MSU crystals, the trigger for NLRP3 inflammasome activation, which leads to the release of interleukin (IL)-1β and results in the flaring of gout. The effects of temperature, an environmental factor for MSU crystallization, on IL-1β secretion have not been well studied. This study examined the effects of temperature on inflammasome activation. Specific triggers activated canonical inflammasomes (NLRP3, NLRC4, and AIM2) in murine macrophages at various temperatures (25, 33, 37, 39, and 42 °C). The maturation of IL-1β and caspase-1 was measured as an indicator for inflammasome activation. As expected, the optimal temperature of inflammasome activation was 37 °C. The MSU crystal-mediated activation of inflammasome increased at temperatures lower than 37 °C and decreased at higher temperatures. MSU crystals at lower temperatures enhanced IL-1β secretion via the NLRP3 inflammasome pathway. A lower temperature promoted the formation of MSU crystals without changing phagocytosis. Overall, lower temperatures form more MSU crystals and enhance NLRP3 inflammasome activation. In light of these findings, it is possible that hyperthermia therapy may reduce gout flaring.
Collapse
Affiliation(s)
| | | | - Geun-Shik Lee
- Correspondence: ; Tel.: +82-3-3250-8683; Fax: +82-3-3244-2367
| |
Collapse
|
6
|
Ahn H, Lee G, Kim J, Park J, Kang SG, Yoon SI, Lee E, Lee GS. NLRP3 Triggers Attenuate Lipocalin-2 Expression Independent with Inflammasome Activation. Cells 2021; 10:cells10071660. [PMID: 34359830 PMCID: PMC8305203 DOI: 10.3390/cells10071660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
Lipocalin-2 (LCN2), a small secretory glycoprotein, is upregulated by toll-like receptor (TLR) signaling in various cells and tissues. LCN2 inhibits bacterial growth by iron sequestration and regulates the innate immune system. Inflammasome activates the inflammatory caspases leading to pyroptosis and cytokine maturation. This study examined the effects of inflammasome activation on LCN2 secretion in response to TLR signaling. The triggers of NLRP3 inflammasome activation attenuated LCN2 secretion while it induced interleukin-1β in mouse macrophages. In mice, NLRP3 inflammasome activation inhibited TLR-mediated LCN2 secretion. The inhibition of NLRP3 triggers on LCN2 secretion was caused by the inhibited transcription and translation of LCN2. At the same time, no changes in the other cytokines and IκBζ, a well-known transcriptional factor of Lcn2 transcription, were observed. Overall, NLRP3 triggers are a regulator of LCN2 expression suggesting a new linkage of inflammasome activation and LCN2 secretion in the innate immunity.
Collapse
Affiliation(s)
- Huijeong Ahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
| | - Gilyoung Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
| | - Jeongeun Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
| | - Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
| | - Seung Goo Kang
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (S.G.K.); (S.-I.Y.)
| | - Sung-Il Yoon
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (S.G.K.); (S.-I.Y.)
| | - Eunsong Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (H.A.); (G.L.); (J.K.); (J.P.); (E.L.)
- Correspondence: ; Tel.: +82-3-3250-8683; Fax: +82-3-3244-2367
| |
Collapse
|
7
|
Lam HYP, Liang TR, Peng SY. Ameliorative effects of Schisandrin B on Schistosoma mansoni-induced hepatic fibrosis in vivo. PLoS Negl Trop Dis 2021; 15:e0009554. [PMID: 34161342 PMCID: PMC8259995 DOI: 10.1371/journal.pntd.0009554] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/06/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Schistosomiasis is second only to malaria as the most devastating parasitic disease in the world. It is caused by the helminths Schistosoma mansoni (S. mansoni), S. haematobium, or S. japonicum. Typically, patients with schistosomiasis suffer from symptoms of liver fibrosis and hepatosplenomegaly. Currently, patients were treated with praziquantel. Although praziquantel effectively kills the worm, it cannot prevent re-infection or resolve liver fibrosis. Also, current treatment options are not ample to completely cure liver fibrosis and splenic damages. Moreover, resistance of praziquantel has been reported in vivo and in vitro studies. Therefore, finding new effective treatment agents is urgently needed. Schisandrin B (Sch B) of Schisandra chinensis has been shown to protect against different liver injuries including fatty liver disease, hepatotoxicity, fibrosis, and hepatoma. We herein investigate the potential of using Sch B to treat S. mansoni-induced liver fibrosis. Results from the present study demonstrate that Sch B is beneficial in treating S. mansoni-induced liver fibrosis and splenic damages, through inhibition of inflammasome activation and apoptosis; and aside from that regulates host immune responses. Besides, Sch B treatment damages male adult worm in the mice, consequently helps to reduce egg production and lessen the parasite burden.
Collapse
Affiliation(s)
- Ho Yin Pekkle Lam
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ting-Ruei Liang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Shih-Yi Peng
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
- * E-mail:
| |
Collapse
|
8
|
Korean Red Ginseng attenuates ultraviolet-mediated inflammasome activation in keratinocytes. J Ginseng Res 2021; 45:456-463. [PMID: 34025139 PMCID: PMC8134848 DOI: 10.1016/j.jgr.2021.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/07/2021] [Accepted: 02/09/2021] [Indexed: 01/08/2023] Open
Abstract
Background Keratinocytes form a physical barrier and act as an innate immune cell in skin. Keratinocytes secrete pro-inflammatory cytokines, such as interleukin (IL)-1β, resulting from inflammasome activation when exposed to ultraviolet (UV) irradiation. Korean Red Ginseng extracts (RGE) have been well-studied as modulators of inflammasome activation in immune cells, such as macrophages. In the study, we elucidated the role of RGE on the UV-mediated inflammasome activation in keratinocytes compared with that in macrophages. Methods Human skin keratinocyte cells (HaCaT), human epidermal keratinocytes (HEK), human monocyte-like cells (THP-1), and mouse macrophages were treated with RGE or a saponin fraction (SF) or non-saponin fraction (NS) of RGE before and after UV irradiation. The secretion levels of IL-1β, as an indicator of inflammasome activation, were analyzed. Results The treatment of RGE or SF in macrophages after UV irradiation inhibited IL-1β secretion, but similar treatment in HaCaT cells did not. However, the treatment of RGE or SF in HaCaT cells in the presence of poly I:C, a toll-like receptor (TLR) 3 ligand, before UV exposure elicited the inhibition of the IL-1β secretion. The inhibition was caused by the disruption by RGE or SF of the TLR mediating up-regulation of the pro-IL-1β and NLRP3 genes during the priming step. Conclusion RGE and its saponins inhibit IL-1β secretion in response to UV exposure in both keratinocytes and macrophages. In particular, RGE treatment interrupted only the priming step in keratinocytes, although it did attenuate both the priming and activation steps in macrophages.
Collapse
|
9
|
Tersigni C, Vatish M, D'Ippolito S, Scambia G, Di Simone N. Abnormal uterine inflammation in obstetric syndromes: molecular insights into the role of chemokine decoy receptor D6 and inflammasome NLRP3. Mol Hum Reprod 2021; 26:111-121. [PMID: 32030415 DOI: 10.1093/molehr/gaz067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
The adaptation of the uterine environment into a favorable immunological and inflammatory milieu is a physiological process needed in normal pregnancy. A uterine hyperinflammatory state, whether idiopathic or secondary to hormonal or organic uterine disorders (polycystic ovary syndromes, endometriosis/adenomyosis and fibroids), negatively influences the interactions between decidua and trophoblast, early in gestation, and between chorion and decidua later in pregnancy. Abnormal activation of uterine inflammatory pathways not only contributes to the pathogenesis of the obstetric syndromes, i.e. recurrent pregnancy loss (RPL), pre-term delivery (PTD) and pre-eclampsia (PE), but also to correlates with severity. In this review, we summarize recent advances in the knowledge of uterine molecular mechanisms of inflammatory modulation in normal pregnancy and obstetric syndromes (RPL, PTD and PE). In particular, we focus on two regulators of uterine/placental inflammation: the NLRP3 inflammasome and the chemokines decoy receptor D6. We performed comprehensive review of the literature in PubMed and Google Scholar databases from 1994 to 2018. The available evidence suggests that: (i) the expression of inflammasome NLRP3 is increased in the endometrium of women with unexplained RPL, in the chorioamniotic membranes of women with PTL and in the placenta of women with PE; (ii) there is a role for abnormal expression and function of D6 decoy receptor at the feto-maternal interface in cases of RPL and PTD and (iii) the function of placental D6 decoy receptor is impaired in PE. A wider comprehension of the inflammatory molecular mechanisms involved in the pathogenesis of the obstetric syndromes might lead to the identification of new potential therapeutic targets.
Collapse
Affiliation(s)
- Chiara Tersigni
- U.O.C. di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy.,Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, OX3 9DU, Oxford, UK
| | - Silvia D'Ippolito
- U.O.C. di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy.,Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Giovanni Scambia
- Università Cattolica del Sacro Cuore, Rome 00168, Italy.,U.O.C. di Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Nicoletta Di Simone
- U.O.C. di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy.,Università Cattolica del Sacro Cuore, Rome 00168, Italy
| |
Collapse
|
10
|
Dey Sarkar R, Sinha S, Biswas N. Manipulation of Inflammasome: A Promising Approach Towards Immunotherapy of Lung Cancer. Int Rev Immunol 2021; 40:171-182. [PMID: 33508984 DOI: 10.1080/08830185.2021.1876044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic inflammation has emerged as a key player at different stages of cancer development. A prominent signaling pathway for acute and chronic inflammation is the activation of the caspase-1 inflammasomes. These are complexes that assemble on activation of certain nucleotide-binding domain, leucine-rich repeat containing proteins (NLRs), AIM2-like receptors (ALRs), or pyrin due to activation via PAMPs or DAMPs. Of these, five complexes-NLRP1, NLRP3, NLRC4, Pyrin, and AIM2 are of importance in the context of cancer for their activities in modulating immune responses, cell proliferation, and apoptosis. Inflammasomes have emerged as clinically relevant in multiple forms of cancer making them highly promising targets for cancer therapy. As lungs are a tissue niche that is prone to inflammation owing to its exposure to external substances, inflammasomes play a vital role in the development and pathogenesis of lung cancer. Therefore, manipulation of inflammasome by various immunomodulatory means could prove a full-proof strategy for the treatment of lung cancer. Here, in this review, we tried to explore the various strategies to target the inflammasomes for the treatment of lung cancer.
Collapse
Affiliation(s)
- Rupak Dey Sarkar
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Samraj Sinha
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, Kolkata, India
| |
Collapse
|
11
|
Ahn H, Lee GS. Riboflavin, vitamin B2, attenuates NLRP3, NLRC4, AIM2, and non-canonical inflammasomes by the inhibition of caspase-1 activity. Sci Rep 2020; 10:19091. [PMID: 33154451 PMCID: PMC7645791 DOI: 10.1038/s41598-020-76251-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Riboflavin is commonly taken as a nutritional supplement, and it converts to coenzymes during the process of energy production from carbohydrates, fats, and proteins. Although riboflavin is considered to be an anti-inflammatory vitamin because of its antioxidant properties, the effects of riboflavin on inflammasome have been not reported. Inflammasome, a cytosolic surveillance protein complex, leads to the activation of caspase-1, cytokine maturation, and pyroptosis. In the present study, riboflavin attenuated the indicators of NLRP3 inflammasome activation in macrophages, such as the maturation and secretion of interleukin (IL)-1β, IL-18, and caspase-1; the formation of Asc pyroptosome; and the cleavage of gasdermin D. In addition, the oral and peritoneal administration of riboflavin inhibited the peritoneal production of IL-1β and IL-18 in a mouse model. Mechanistically, riboflavin prevented mitochondrial perturbations, such as mitochondrial ROS production and mitochondrial DNA release, which trigger the NLRP3 inflammasome assembly. Riboflavin was further confirmed to disrupt the activity of caspase-1, and it also inhibited the AIM2, NLRC4, and non-canonical inflammasomes. Therefore, riboflavin has both an antioxidant effect and an anti-inflammasome property that regulates the inflammatory response.
Collapse
Affiliation(s)
- Huijeong Ahn
- Laboratory of Inflammatory Diseases, Department of Physiology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Geun-Shik Lee
- Laboratory of Inflammatory Diseases, Department of Physiology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
12
|
Zhou F, Li C, Zhang SY. NLRP3 inflammasome: a new therapeutic target for high-risk reproductive disorders? Chin Med J (Engl) 2020; 134:20-27. [PMID: 33395071 PMCID: PMC7862815 DOI: 10.1097/cm9.0000000000001214] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT The NOD-like receptor protein 3 (NLRP3) inflammasome is a key regulator of the host's immune response, and many immune and metabolic disorders are linked to its activation. This review aimed to investigate and clarify the relationship between this inflammasome and high-risk reproductive disorders. Papers cited here were retrieved from PubMed up to August 2020 using the keywords "NLRP3" or "NALP3", "caspase-1", "endometriosis", "gestational diabetes", "interleukin (IL)-18", "IL-1β", "pre-eclampsia (PE)", "preterm birth", "polycystic ovarian syndrome (PCOS)", "recurrent spontaneous abortion (RSA)", and combinations of these terms. The results show that NLRP3 inflammasome is associated with various high-risk reproductive disorders and many inflammatory factors are secreted during its activation, such as IL-1β induced during the development of endometriosis. PCOS is also associated with activation of the NLRP3 inflammasome, especially in overweight patients. It also participates in the pathogenesis of RSA and is activated in fetal membranes before preterm birth. The placentas of pregnant women with PE show higher expression of the NLRP3 inflammasome, and gestational diabetes mellitus occurs simultaneously with its activation. Current evidence suggest that the NLRP3 inflammasome plays an important role in female reproductive disorders. New treatment and management methods targeting it might help reduce the incidence of such disorders and improve neonatal outcomes.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | | | | |
Collapse
|
13
|
Recent insights on modulation of inflammasomes by adipokines: a critical event for the pathogenesis of obesity and metabolism-associated diseases. Arch Pharm Res 2020; 43:997-1016. [PMID: 33078304 DOI: 10.1007/s12272-020-01274-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Aberrant production of adipokines, a group of adipocytes-derived hormones, is considered one of the most important pathological characteristics of obesity. In individuals with obesity, beneficial adipokines, such as adiponectin are downregulated, whereas leptin and other pro-inflammatory adipokines are highly upregulated. Hence, the imbalance in levels of these adipokines is thought to promote the development of obesity-linked complications. However, the mechanisms by which adipokines contribute to the pathogenesis of various diseases have not been clearly understood. Inflammasomes represent key signaling platform that triggers the inflammatory and immune responses through the processing of the interleukin family of pro-inflammatory cytokines in a caspase-1-dependent manner. Beyond their traditional function as a component of the innate immune system, inflammasomes have been recently integrated into the pathological process of multiple metabolism- and obesity-related disorders such as cardiovascular diseases, diabetes, fatty liver disease, and cancer. Interestingly, emerging evidence also highlights the role of adipokines in the modulation of inflammasomes activation, making it a promising mechanism underlying distinct biological actions of adipokines in diseases driven by inflammation and metabolic disorders. In this review, we summarize the effects of adipokines, in particular adiponectin, leptin, visfatin and apelin, on inflammasomes activation and their implications in the pathophysiology of obesity-linked complications.
Collapse
|
14
|
Ahn H, Han BC, Lee SH, Lee GS. Fructose-arginine, a non-saponin molecule of Korean Red Ginseng, attenuates AIM2 inflammasome activation. J Ginseng Res 2020; 44:808-814. [PMID: 33192124 PMCID: PMC7655492 DOI: 10.1016/j.jgr.2020.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/22/2020] [Accepted: 06/26/2020] [Indexed: 01/29/2023] Open
Abstract
Background Korean Red Ginseng extract (RGE) has been reported to act as an inflammasome modulator. Ginsenosides, saponin molecules of RGE, selectively inhibit activation of NLRP3 and AIM2 inflammasomes, while non-saponin molecules of RGE upregulate inflammasome components associated with the initiation of NLRP3 inflammasome activation. In this study, we investigated the effect of non-saponin components of RGE on AIM2 inflammasome activation. Methods The role of non-saponins of RGE on AIM2 inflammasomes was tested in mouse bone marrow-derived macrophages, a human monocyte-like cell line, and a mouse animal model. Cells or mice were transfected with dsDNA or inoculated with Listeria monocytogenes to activate AIM2 inflammasomes. Several indices of inflammasome activation were examined via immunoblot or ELISA analysis. Results The non-saponin fraction and saponin-eliminating fraction (SEF) of RGE selectively attenuated the activation of AIM2 inflammasomes, but not that of NLRP3 or NLRC4 inflammasomes. Fructose-arginine, an amino-sugar, was shown to be effective against AIM2 inflammasome activation. Conclusion Non-saponins of RGE, such as fructose-arginine, might be effective in regulating infectious and autoimmune diseases resulting from AIM2 inflammasome activation.
Collapse
Affiliation(s)
- Huijeong Ahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung-Cheol Han
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea.,Korea Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Seung-Ho Lee
- Korea Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
15
|
Ruhee RT, Roberts LA, Ma S, Suzuki K. Organosulfur Compounds: A Review of Their Anti-inflammatory Effects in Human Health. Front Nutr 2020; 7:64. [PMID: 32582751 PMCID: PMC7280442 DOI: 10.3389/fnut.2020.00064] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Abstract
Phytonutrients are widely recognized for providing protective human health benefits. Among the phytonutrients, epidemiological and experimental studies show that dietary organosulfur compounds (OSC) play a significant role in preventing various human pathological progressions, including chronic inflammation, by decreasing inflammatory mediators such as nitric oxide (NO), prostaglandin (PG)E2, interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and IL-17, which are all typical hallmarks of inflammation. Evidence supports OSC in reducing the expression of these markers, thereby attenuating chronic inflammatory processes. Nuclear factor-kappa B (NF-κB) is a key regulating factor during inflammation, and novel evidence shows that OSC downregulates this transcriptional factor, thus contributing to the anti-inflammatory response. In vitro and in vivo studies show that inflammation is mechanistically linked with acute and chronic pathological conditions including cancer, diabetes, obesity, neural dysfunction, etc. Furthermore, a considerable number of experiments have demonstrated that the anti-inflammatory properties of OSC occur in a dose-dependent manner. These experiments also highlight indirect mechanisms as well as potent co-functions for protective roles as antioxidants, and in providing chemoprotection and neuroprotection. In this brief review, we provided an overview of the anti-inflammatory effects of OSC and elucidated probable mechanisms that are associated with inflammation and chronic disorders.
Collapse
Affiliation(s)
| | - Llion Arwyn Roberts
- School of Allied Health Sciences, Griffith University, Gold Coast, QLD, Australia.,School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Sihui Ma
- Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan
| | | |
Collapse
|
16
|
Ahn H, Kim J, Lee H, Lee E, Lee GS. Characterization of equine inflammasomes and their regulation. Vet Res Commun 2020; 44:51-59. [PMID: 32297137 DOI: 10.1007/s11259-020-09772-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Inflammasome, a cytosolic multi-protein complex, assembly is a response to sensing intracellular pathogenic and endogenic danger signals followed by caspase-1 activation, which maturates precursor cytokines such as interleukin (IL)-1β. Most inflammasome research has been undertaken in humans and rodents, and inflammasomes in veterinary species have not been well-characterized. In this study, we observed the effects of well-known inflammasome activators on equine peripheral blood monocytes (PBMCs). The NLRP3 inflammasome triggers include ATP, nigericin, aluminum crystals, and monosodium urate crystals, and NLRP3 activation induces IL-1β secretion in a dose-dependent manner. Activators of NLRC4 and AIM2 inflammasomes include cytosolic flagellin and dsDNA, and their activation induces IL-1β secretion. The bacterial inflammasome triggers Salmonella Typhimurium and Listeria monocytogenes also induce IL-β releases. To elucidate the role of potassium efflux as an upstream signal of NLRP3 inflammasome activation, equine PBMCs were treated with blockers of potassium efflux in the presence of NLRP3 triggers. As a result, the IL-1β secretion stemming from equine NLRP3 inflammasome activation was not completely attenuated by the inhibition of potassium efflux. Taken together, the results indicate that equine PBMCs normally secrete IL-1β in response to well-known inflammasome activators, although equine NLRP3 inflammasome activation might not be dependent on potassium efflux.
Collapse
Affiliation(s)
- Huijeong Ahn
- College of Veterinary Medicine, Institute of Veterinary Science, Kangwon National University, 24341, Chuncheon, Republic of Korea
| | - Jeongeun Kim
- College of Veterinary Medicine, Institute of Veterinary Science, Kangwon National University, 24341, Chuncheon, Republic of Korea
| | - Hansae Lee
- College of Veterinary Medicine, Institute of Veterinary Science, Kangwon National University, 24341, Chuncheon, Republic of Korea
| | - Eunsong Lee
- College of Veterinary Medicine, Institute of Veterinary Science, Kangwon National University, 24341, Chuncheon, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine, Institute of Veterinary Science, Kangwon National University, 24341, Chuncheon, Republic of Korea. .,Laboratory of Inflammatory Diseases, Department of Physiology, College of Veterinary Medicine, Kangwon National University, 24341, Chuncheon, Republic of Korea.
| |
Collapse
|
17
|
Yu X, Wu Y, Zhang J, Jirimutu, Zulipikaer A, Chen J. Pre-evaluation of humoral immune response of Bactrian camels by the quantification of Th2 cytokines using real-time PCR. J Biomed Res 2020; 34:387-394. [PMID: 32611846 PMCID: PMC7540241 DOI: 10.7555/jbr.34.20190035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/13/2019] [Indexed: 01/12/2023] Open
Abstract
With the increasing immunological studies on camels due to the advantage of their single-chain antibodies for humanizations, it is demanding to develop an easy-to-handle evaluation method of their humoral immune response before proceeding with immunization of foreign antigens that may be toxic to camels. In this study, we quantitatively determined the expression levels of T-helper 2 (Th2) cytokines in peripheral blood lymphocytes obtained from Bactrian camels by real-time PCR. The recorded kinetic profiles resulting from the immunization of ovalbumin (OVA) indicated that after immunization, Th2 cytokines including interleukin (IL) families such as IL-4, IL-10, and IL-13 in the camels were up-regulated by a factor of 1.78, 3.15, and 1.22, respectively, which was validated by traditional enzyme-linked immunosorbent assay (ELISA) methods. Unlike ELISA which requires specific enzyme-labeled antibodies, this established method based on the minimal amount of blood samples holds an advantage in the preliminary evaluation of camel humoral immune response with desirable precision, which is meaningful for biomedical explorations of camel-derived antibodies.
Collapse
Affiliation(s)
- Xinyu Yu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuan Wu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Jiarong Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jirimutu
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia 010018, China
| | | | - Jin Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
18
|
Gruber JV, Holtz R. In vitro expression of NLRP inflammasome-induced active Caspase-1 expression in normal human epidermal keratinocytes (NHEK) by various exogenous threats and subsequent inhibition by naturally derived ingredient blends. J Inflamm Res 2019; 12:219-230. [PMID: 31692589 PMCID: PMC6716588 DOI: 10.2147/jir.s215776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
Background The discovery of the nod-like receptor protein (NLRP) inflammasomes in 2002 has led to the rapid identification of these unique cellular proteins as key targets for studies on innate inflammation pathways. The NLRP inflammasomes have been shown to be expressed in normal human epidermal keratinocytes (NHEK) and human dermal fibroblasts (HDF). NLRP inflammasomes in keratinocytes are interesting as these skin cells are the first living cells in the skin to contact external exogenous threats such as UV energy, chemicals, physical trauma, and bacteria and viruses. Activation of the NLRP Inflammasomes by exogenous threats results in the release of active Caspase-1 (ACasp-1), a key protease enzyme, which targets inactive forms of IL-1β, IL-18 as well as IL-1α and IL-33. Purpose This article discusses efforts to examine the release of active Caspase-1 from NHEKs activated by various exogenous threats including UVB energy, ATP, Nigericin and Urban Dust. The work further examines if, after inflammasome activation and Caspase-1 release, certain naturally derived botanical ingredients known to have anti-inflammatory effects can function to inhibit upregulation of active Caspase-1. Methods NHEK were treated with various doses of UVB, ATP and Nigericin and with a single dose of Urban Dust. ACasp-1 expression was measured after 3 and 20 hours using the Promega Caspase Glo-1 bioluminescent assay. After confirmation that 60 mJ/cm2 of UVB and 5mM of ATP were effective to activate NHEK ACasp-1 release after 20 hrs, these conditions were employed to examine the influence of three botanical blends of ingredients on their ability to inhibit ACasp-1 expression. Results Initial results demonstrate that NHEKs can be activated to release active Caspase-1 by ATP and UVB, but not by Nigericin or Urban Dust. In addition, it was unexpectedly found that, while ATP and UVB activated NHEKs, the release of ACasp-1-did not happen within the first 3 hours after exposure but did become significant after 20 hours. Additional results indicate that a blend of polysaccharides and two blends of antioxidants, one oil-soluble and the other water-soluble, known for their anti-inflammatory effects, can reduce expression of active Caspase-1 in activated NHEKs when applied extracellularly. Conclusion Expression of NLRP activated release of ACasp-1 was found to be influenced by UVB and ATP but not by Nigericin or Urban Dust. The effects were also time dependent. Several botanical extract blends were found to reduce ACasp-1 expression in previously activated NHEKs. Links between these inflammatory effects and processes of cellular inflammaging are discussed.
Collapse
Affiliation(s)
- James V Gruber
- Research and Development, BotanicalsPlus, Little Falls, NJ 07424, USA
| | - Robert Holtz
- Research and Development, Bioinnovation Laboratories, Inc, Lakewood, CO 80235, USA
| |
Collapse
|