101
|
Zhang Q, Sun J, Fu Y, He W, Li Y, Tan H, Xu H, Jiang X. Guttiferone K Exerts the Anti-inflammatory Effect on Mycobacterium Tuberculosis- (H37Ra-) Infected Macrophages by Targeting the TLR/IRAK-1 Mediated Akt and NF- κB Pathway. Mediators Inflamm 2020; 2020:8528901. [PMID: 33100904 PMCID: PMC7569438 DOI: 10.1155/2020/8528901] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/26/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) remains a great threat to global health, killing more people than any other single infectious agent and causing uncontrollable inflammation in the host. Poorly controlled inflammatory processes can be deleterious and result in immune exhaustion. The current tuberculosis (TB) control is facing the challenge of drugs deficiency, especially in the context of increasingly multidrug resistant (MDR) TB. Under this circumstance, alternative host-directed therapy (HDT) emerges timely which can be exploited to improve the efficacy of TB treatment and disease prognosis by targeting the host. Here, we established the in vitro infection model of Mtb macrophages with H37Ra strain to seek effective anti-TB active agent. The present study showed that Guttiferone K, isolated from Garcinia yunnanensis, could significantly inhibit Mtb-induced inflammation in RAW264.7 and primary peritoneal macrophages. It was evidenced by the decreased production of inflammatory mediators, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Further studies with immunoblotting and immunofluorescence revealed that Guttiferone K obviously inhibits the nuclear factor-kappa B (NF-κB) both in RAW264.7 and primary peritoneal macrophages relying on the TLR/IRAK-1 pathway. Guttiferone K could also suppress the NLRP3 inflammasome activity and induce autophagy by inhibiting the protein kinase B (p-Akt) and mammalian target of rapamycin (mTOR) phosphorylation at Ser473 and Ser2448 in both cell lines. Thus, Guttiferone K possesses significant anti-inflammatory effect, alleviating Mtb-induced inflammation with an underlying mechanism that targeting on the TLR/IRAK-1 pathway and inhibiting the downstream NF-κB and Akt/mTOR signaling pathways. Together, Guttiferone K can be an anti-inflammatory agent candidate for the design of new adjunct HDT drugs fighting against tuberculosis.
Collapse
Affiliation(s)
- Qingwen Zhang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, 201318 Shanghai, China
| | - Jinxia Sun
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Yan Fu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Weigang He
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Yinhong Li
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Hongsheng Tan
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, 200240 Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Xin Jiang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| |
Collapse
|
102
|
Chen X, Nie K, Zhang X, Tan S, Zheng Q, Wang Y, Chen X, Tang Z, Liu R, Yan M, Liu Z, Lin J, Xu J, Zhang N, Wang H, Yang J. The recombinant anti-TNF-α fusion protein ameliorates rheumatoid arthritis by the protective role of autophagy. Biosci Rep 2020; 40:BSR20194515. [PMID: 32880389 PMCID: PMC7502693 DOI: 10.1042/bsr20194515] [Citation(s) in RCA: 5] [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: 01/07/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 01/08/2023] Open
Abstract
The currently used anti-cytokine therapeutic antibodies cannot selectively neutralize pathogenic cytokine signaling that cause collateral damage to protective signaling cascades carrying the potential for unwanted side effects. The variable domains of heavy-chain only antibodies (HCAbs) discovered in Camelidae are stable and display to be fully functional in antigen-binding against variable targets, which seem to be attractive candidates for the next-generation biologic drug study. The purpose of our study was to establish a simple prokaryotic expression system for large-scale expression, purification, and refolding of the recombinant anti-tumor necrosis factor α (TNF-α) fusion protein (FVH1-1) from inclusion bodies. Over 95% purity of the recombinant anti-TNF-α fusion proteins was obtained by just one purification step in our developed prokaryotic expression system, while the results of surface plasmon resonance (SPR) established the high-efficiency potent binding ability of FVH1-1 to human TNF-α. The counteraction of TNF-α cytotoxic effect experiment on the mouse fibroblast fibrosarcoma cell line (L929) confirmed that the expressed FVH1-1 were able to selectively and highly combine with human recombinant TNF-α (hTNF-α) in vitro. Western blot results showed that FVH1-1 can inhibit the activation of caspase-9 and PARP, which are the apoptotic signaling pathway proteins activated by hTNF-α. Meanwhile, lysosome autophagy signaling pathways stimulated by hTNF-α were inhibited by FVH1-1, which down-regulated the expression of LC3II/LC3I and up-regulated the expression of P62, indicating that the autophagy linked with TNF-α-induced apoptosis in response to rheumatoid arthritis. The results of the AIA rat model experiment presented that FVH1-1 can reduce the degree of joint swelling and inflammatory factors to a certain extent in vivo.
Collapse
Affiliation(s)
- Xiaole Chen
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
| | - Kaimei Nie
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Zhang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuangyu Tan
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingmei Zheng
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yaduan Wang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaofeng Chen
- Department of Neurorehabilitation, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhiyu Tang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Rui Liu
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Mengru Yan
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhiwei Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianbo Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianhua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Nanwen Zhang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - He Wang
- School of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Juhua Yang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
103
|
Thiriot JD, Martinez-Martinez YB, Endsley JJ, Torres AG. Hacking the host: exploitation of macrophage polarization by intracellular bacterial pathogens. Pathog Dis 2020; 78:5739920. [PMID: 32068828 DOI: 10.1093/femspd/ftaa009] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages play an integral role in host defenses against intracellular bacterial pathogens. A remarkable plasticity allows for adaptation to the needs of the host to orchestrate versatile innate immune responses to a variety of microbial threats. Several bacterial pathogens have adapted to macrophage plasticity and modulate the classical (M1) or alternative (M2) activation bias towards a polarization state that increases fitness for intracellular survival. Here, we summarize the current understanding of the host macrophage and intracellular bacterial interface; highlighting the roles of M1/M2 polarization in host defense and the mechanisms employed by several important intracellular pathogens to modulate macrophage polarization to favor persistence or proliferation. Understanding macrophage polarization in the context of disease caused by different bacterial pathogens is important for the identification of targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joseph D Thiriot
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Yazmin B Martinez-Martinez
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Janice J Endsley
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Alfredo G Torres
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA.,Department of Pathology, University of Texas Medical Branch , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| |
Collapse
|
104
|
Zhang Y, Zhao Y, Zhang J, Yang G. Mechanisms of NLRP3 Inflammasome Activation: Its Role in the Treatment of Alzheimer's Disease. Neurochem Res 2020; 45:2560-2572. [PMID: 32929691 DOI: 10.1007/s11064-020-03121-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease of progressive dementia which is characterized pathologically by extracellular neuritic plaques containing aggregated amyloid beta (Aβ) and intracellular hyperphosphorylated tau protein tangles in cerebrum. It has been confirmed that microglia-specific nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome-mediated chronic neuroinflammation plays a crucial role in the pathogenesis of AD. Stimulated by Aβ deposition, NLRP3 assembles and activates within microglia in the AD brain, leading to caspase-1 activation along with downstream interleukin (IL)-1β secretion, and subsequent inflammatory events. Activation of the NLRP3 inflammasome mediates microglia to exhibit inflammatory M1 phenotype, with high expression of caspase-1 and IL-1β. This leads to Aβ deposition and neuronal loss in the amyloid precursor protein (APP)/human presenilin-1 (PS1) mouse model of AD. However, NLRP3 or caspase-1 deletion in APP/PS1 mice promotes microglia to transform to an anti-inflammatory M2 phenotype, with decreased secretion of caspase-1 and IL-1β. It also results in improved cognition, enhanced Aβ clearance, and a lower cerebral inflammatory response. This result suggests that the NLRP3 inflammasome may be an appropriate target for reducing neuroinflammation and alleviating pathological processes in AD. In the present review, we summarize the generally accepted regulatory mechanisms of NLRP3 inflammasome activation, and explore its role in neuroinflammation. Furthermore, we speculate on the possible roles of microglia-specific NLRP3 activation in AD pathogenesis and consider potential therapeutic interventions targeting the NLRP3 inflammasome in AD.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Yuan Zhao
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Jian Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Guofeng Yang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China.
| |
Collapse
|
105
|
Zhao W, Shi CS, Harrison K, Hwang IY, Nabar NR, Wang M, Kehrl JH. AKT Regulates NLRP3 Inflammasome Activation by Phosphorylating NLRP3 Serine 5. THE JOURNAL OF IMMUNOLOGY 2020; 205:2255-2264. [PMID: 32929041 DOI: 10.4049/jimmunol.2000649] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/17/2020] [Indexed: 02/05/2023]
Abstract
The cytosolic pattern recognition receptor NLRP3 senses host-derived danger signals and certain microbe-derived products in both humans and rodents. NLRP3 activation assembles an inflammasome complex that contains the adapter proteins ASC and caspase-1, whose activation triggers the maturation and release of the proinflammatory cytokines IL-1β and IL-18. S5 phosphorylation of NLRP3 prevents its oligomerization and activation, whereas dephosphorylation of this residue by the phosphatase PP2A allows NLRP3 activation. However, the protein kinase that mediates NLRP3 S5 phosphorylation is unknown. In this study, we show that AKT associates with NLRP3 and phosphorylates it on S5, limiting NLRP3 oligomerization. This phosphorylation event also stabilizes NLRP3 by reducing its ubiquitination on lysine 496, which inhibits its proteasome-mediated degradation by the E3 ligase Trim31. Pharmacologic manipulation of AKT kinase activity reciprocally modulates NLRP3 inflammasome-mediated IL-1β production. Inhibition of AKT reduced IL-1β production following the i.p. injection of LPS into mice. We propose that AKT, Trim31, and PP2A together modulate NLRP3 protein levels and the tendency to oligomerize, thereby setting a tightly regulated threshold for NLRP3 activation.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892.,State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China; and.,Department of Prosthodontics, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chong-Shan Shi
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kathleen Harrison
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Il-Young Hwang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Neel R Nabar
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Min Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China; and
| | - John H Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
106
|
Ziegler ME, Claytor B, Bell M, Casas L, Widgerow AD. Gene Expression Changes in the Skin of Patients Undergoing Medial Thigh Liposuction With Pre-Surgical and Post-Surgical Application of Topical Products. Aesthet Surg J Open Forum 2020; 2:ojaa033. [PMID: 33791656 PMCID: PMC7671262 DOI: 10.1093/asjof/ojaa033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Skin topical preconditioning before and after surgical procedures is a relatively new concept, particularly in relation to the efficient removal of tissue breakdown products. Clinical trials demonstrate improvements, such as less induration, when surgery is combined with topical product preconditioning and with usage post-surgery. OBJECTIVES This trial aimed to assess the efficacy of such a regimen at the molecular level through gene expression studies in combination with clinical assessments. METHODS Six women who underwent medial thigh liposuction administered either a bland moisturizer or the experimental topical products to each side of the surgical area twice daily. Biopsies were taken before any topical application, at 2 and 4 weeks after liposuction. An inflammation-related gene expression analysis was conducted to compare the different conditions. In addition, the degree of induration was assessed in a blinded manner. RESULTS Compared with the bland moisturizer, the experimental group demonstrated a hastened immune inflammatory response moving more rapidly to an anti-inflammatory reversal at 2 weeks followed by a wound healing extracellular remodeling effect at 4 weeks. This matched the clinical picture depicting less induration with the treatment. CONCLUSIONS For patients undergoing body procedures, a topical treatment with the Alastin induces an accelerated healing response, inducing the clearance of "waste" products and the induction of anti-inflammatory genes. Furthermore, this topical treatment stimulates extracellular matrix remodeling, which ultimately leads to less induration. LEVEL OF EVIDENCE 5
Collapse
Affiliation(s)
| | | | | | | | - Alan D Widgerow
- Corresponding Author:Dr Alan D. Widgerow, 3129 Tiger Run Court Suite #109, Carlsbad, CA 92010, USA. E-mail: ; Instagram: @alanwidge
| |
Collapse
|
107
|
Dankers W, Hasnat MA, Swann V, Alharbi A, Lee JP, Cristofaro MA, Gantier MP, Jones SA, Morand EF, Flynn JK, Harris J. Necrotic cell death increases the release of macrophage migration inhibitory factor by monocytes/macrophages. Immunol Cell Biol 2020; 98:782-790. [PMID: 32654231 DOI: 10.1111/imcb.12376] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/16/2020] [Accepted: 07/09/2020] [Indexed: 12/01/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory molecule with both cytokine and noncytokine activity. MIF is constitutively released from multiple cell types via an unconventional secretory pathway that is not well defined. Here, we looked at MIF release from human and mouse monocytes/macrophages in response to different stimuli. While MIF release was not significantly altered in response to lipopolysaccharide or heat-killed Escherichia coli, cytotoxic stimuli strongly promoted release of MIF. MIF release was highly upregulated in cells undergoing necrosis, necroptosis and NLRP3 inflammasome-dependent pyroptosis. Our data suggest that cell death represents a major route for MIF release from myeloid cells. The functional significance of these findings and their potential importance in the context of autoimmune and inflammatory diseases warrant further investigation.
Collapse
Affiliation(s)
- Wendy Dankers
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Md Abul Hasnat
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Vanesa Swann
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Arwaf Alharbi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute for Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Jacinta Pw Lee
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Megan A Cristofaro
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Michael P Gantier
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute for Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Sarah A Jones
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Eric F Morand
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Jacqueline K Flynn
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - James Harris
- Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
108
|
Ye J, Lu S, Wang M, Ge W, Liu H, Qi Y, Fu J, Zhang Q, Zhang B, Sun G, Sun X. Hydroxysafflor Yellow A Protects Against Myocardial Ischemia/Reperfusion Injury via Suppressing NLRP3 Inflammasome and Activating Autophagy. Front Pharmacol 2020; 11:1170. [PMID: 32848777 PMCID: PMC7406911 DOI: 10.3389/fphar.2020.01170] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/17/2020] [Indexed: 12/24/2022] Open
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a serious threat to human health. Hydroxysafflor yellow A (HSYA), the main water-soluble ingredient extracted from Carthami flos (Carthamus tinctorius L.), has therapeutic potential for treating MI/R injury. However, the mechanisms of HSYA−mediated protection from MI/R injury are incompletely understood. In the present study, we investigated the effects and the underlying mechanisms of HSYA during MI/R. Adult Sprague-Dawley rats were subjected to left anterior descending artery ligation for 30 min followed by 24 h of reperfusion with or without HSYA treatment. The protective effect of HSYA was detected by 2,3,5-triphenyl tetrazolium chloride (TTC) staining, hematoxylin eosin (HE) staining, and myocardial enzymes detections. Serum levels of inflammatory factors such as TNF-α, interleukin (IL)-1β, and IL-18, were detected using ELISA kits. The expression of NLRP3 and other related proteins in the myocardium was detected by western blot and immunohistochemistry. The expression of autophagy-related proteins, including Atg5, BECN1, P62, and LC3B, was detected by western blot to evaluate the effect of HSYA on autophagy. Results showed that HSYA decreased the myocardial infarct size and attenuated the cardiac dysfunction in rats after I/R. In addition, HSYA inhibited myocardial apoptosis compared with the I/R group, decreased the levels of inflammatory cytokines in rat serum, reduced NLRP3 inflammasome expression, and induced autophagy. Mechanistically, our results demonstrated that HSYA can activate AMPK to improve autophagy and inhibit NLRP3 inflammasome by inhibiting the mTOR pathway. This work provides strong data supporting for the clinical applications of HSYA in MI/R injury.
Collapse
Affiliation(s)
- Jingxue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shan Lu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenxiu Ge
- College of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Haitao Liu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yaodong Qi
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhua Fu
- Pneumology Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiong Zhang
- Pneumology Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bengang Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
109
|
Li Z, Guo J, Bi L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed Pharmacother 2020; 130:110542. [PMID: 32738636 DOI: 10.1016/j.biopha.2020.110542] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
NOD-like receptor family pyrin domain containing 3 (NLRP3) is an intracellular receptor that senses foreign pathogens and endogenous danger signals. It assembles with apoptosis-associated speck-like protein containing a CARD (ASC) and caspase-1 to form a multimeric protein called the NLRP3 inflammasome. Among its various functions, the NLRP3 inflammasome can induce the release of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 while also promoting gasdermin D (GSDMD)-mediated pyroptosis. Previous studies have established a vital role for the NLRP3 inflammasome in innate and adaptive immune system as well as its contribution to several autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), Sjögren's syndrome (SS), systemic sclerosis (SSc), and ankylosing spondylitis (AS). In this review, we briefly introduce the biological features of the NLRP3 inflammasome and present the mechanisms underlying its activation and regulation. We also summarize recent studies that have reported on the roles of NLRP3 inflammasome in the immune system and several autoimmune diseases, with a focus on therapeutic and clinical applications.
Collapse
Affiliation(s)
- Zhe Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jialong Guo
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
110
|
Kumari P, Russo AJ, Shivcharan S, Rathinam VA. AIM2 in health and disease: Inflammasome and beyond. Immunol Rev 2020; 297:83-95. [PMID: 32713036 DOI: 10.1111/imr.12903] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022]
Abstract
Nucleic acid sensing is a critical mechanism by which the immune system monitors for pathogen invasion. A set of germline-encoded innate immune receptors detect microbial DNA in various compartments of the cell, such as endosomes, the cytosol, and the nucleus. Sensing of microbial DNA through these receptors stimulates, in most cases, interferon regulatory factor-dependent type I IFN synthesis followed by JAK/STAT-dependent interferon-stimulated gene expression. In contrast, the detection of DNA in the cytosol by AIM2 assembles a macromolecular complex called the inflammasome, which unleashes the proteolytic activity of a cysteine protease caspase-1. Caspase-1 cleaves and activates the pro-inflammatory cytokines such as IL-1β and IL-18 and a pore-forming protein, gasdermin D, which triggers pyroptosis, an inflammatory form of cell death. Research over the past decade has revealed that AIM2 plays essential roles not only in host defense against pathogens but also in inflammatory diseases, autoimmunity, and cancer in inflammasome-dependent and inflammasome-independent manners. This review discusses the latest advancements in our understanding of AIM2 biology and its functions in health and disease.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Sonia Shivcharan
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| |
Collapse
|
111
|
Interplay between Cellular and Molecular Mechanisms Underlying Inflammatory Bowel Diseases Development-A Focus on Ulcerative Colitis. Cells 2020; 9:cells9071647. [PMID: 32659925 PMCID: PMC7408467 DOI: 10.3390/cells9071647] [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: 05/16/2020] [Revised: 06/26/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are defined by the continuous inflammation of the gastrointestinal tract. During inflammation, the number of pathogens in the intestinal epithelium increases, leading to inflammasome assembly. Inflammasome activation is meant to protect the intestinal epithelial barrier from further damage by maintaining homeostasis. Although its purpose is to protect the cells, excessive nucleotide-binding oligomerization domain-like receptor and pyrin domain-containing protein 3 (NLRP3) inflammasome assembly is responsible for the synthesis of a high number of pro-inflammatory cytokines. The activation of two crucial pathways, autophagy process, and unfolded protein response, is initiated for restoring homeostasis. Aberrant expression of miRNAs and lncRNAs also interfere with the pathogenic mechanisms of IBD, as these non-coding transcripts play key roles in regulation of biological processes, such as inflammation and immunity. This review thoroughly describes the cellular and molecular mechanism that trigger and perpetuate inflammation in ulcerative colitis (UC) patients.
Collapse
|
112
|
Yina S, Zhongjie C, Kaiyu C, Chenghua L, Xiaodong Z. Target of rapamycin signaling inhibits autophagy in sea cucumber Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2020; 102:480-488. [PMID: 32437859 DOI: 10.1016/j.fsi.2020.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 06/11/2023]
Abstract
Autophagy mediated by mTOR pathway is a particularly important immune defense mechanism in the pathogens infected mammals. However, the role of TOR in echinoderm autophagy is largely unknown. Here, a cDNA encoding TOR protein was cloned and characterized from sea cucumber Apostichopus japonicus (designated as AjTOR) and its biological functions were also investigated. The AjTOR gene encoded a peptide of 2499 amino acids with the representative domains of DUF3385, FAT, FRB, PI3Kc, and FATC, which exhibited highly conservation with vertebrate orthologs. Phylogenetic analysis supported that AjTOR belonged to a new member of TOR family. Moreover, tissues distribution analysis indicated that AjTOR was ubiquitously expressed in all the tested tissues, with the highest transcription in muscle. Vibrio splendidus infection in vivo and LPS challenge in vitro could both significantly down-regulate the mRNA expression of AjTOR. What's more, transmission electron microscopy observations showed that rapamycin treatment resulted in rapid formation of autophagosomes in coelomocytes both at 3 and 6 h, however, injection with mTOR activator of MHY1485 showed an inhibitory effect on autophagosomes formation compared to the control, suggesting blocking the expression of AjTOR could accelerates autophagy signals. Our findings supported that AjTOR served as a negative regulator in sea cucumber authophay.
Collapse
Affiliation(s)
- Shao Yina
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Che Zhongjie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chen Kaiyu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Li Chenghua
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| | - Zhao Xiaodong
- School of Ocean, Yantai University, Yantai, 264005, PR China
| |
Collapse
|
113
|
Wang X, Shao QH, Zhou H, Wu JL, Quan WQ, Ji P, Yao YW, Li D, Sun ZJ. Ginkgolide B inhibits lung cancer cells promotion via beclin-1-dependent autophagy. BMC Complement Med Ther 2020; 20:194. [PMID: 32576183 PMCID: PMC7310550 DOI: 10.1186/s12906-020-02980-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/01/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Ginkgolide B (GKB) is a major active component of the extracts of Ginkgo biloba leaves, and it has been used as an anti-cancer agent. However, it is unknown whether GKB has the therapeutic effects on lung cancer. Here, we studied the effects of GKB on lung cancer cells. METHODS The effects of GKB on lung cancer cell proliferation and invasion were analyzed by cell counting kit (CCK-8) and cell invasion assays, respectively. Apoptosis was detected by flow cytometry. Western blot analysis was used to confirm the expression of autophagy-associated proteins in GKB-treated cells. Immunofluorescence analysis was used to analyze the level of light chain 3B (LC3B). RESULTS Treatment with GKB time-dependently inhibited the proliferation and decreased the invasive capacity of A549 and H1975 cells. GKB induced apoptosis of these cells, but there was no significant effect on apoptosis compared to the control treatment. GKB-induced inhibition of cell proliferation and GKB-induced cell death were due to autophagy rather than apoptosis. GKB-induced autophagy of lung cancer cells was dependent on beclin-1, and autophagy-induced inhibition of the NLRP3 inflammasome contributed to the anti-tumor effect of GKB. CONCLUSIONS GKB-mediated autophagy of lung cancer cells is beclin-1-dependent and results in inhibition of the NLRP3 inflammasome. Therefore, GKB might be a potential therapeutic candidate for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacy, Putuo People’s Hospital, Shanghai, 200060 China
| | - Qi-Hui Shao
- grid.24516.340000000123704535Department of Traditional Chinese Medicine, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Hao Zhou
- Department of Pharmacy, Putuo People’s Hospital, Shanghai, 200060 China
| | - Jun-Lu Wu
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Wen-Qiang Quan
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Ping Ji
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Yi-Wen Yao
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Dong Li
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Zu-Jun Sun
- grid.24516.340000000123704535Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| |
Collapse
|
114
|
Carmona-Gutierrez D, Bauer MA, Zimmermann A, Kainz K, Hofer SJ, Kroemer G, Madeo F. Digesting the crisis: autophagy and coronaviruses. MICROBIAL CELL (GRAZ, AUSTRIA) 2020; 7:119-128. [PMID: 32391393 PMCID: PMC7199282 DOI: 10.15698/mic2020.05.715] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/08/2023]
Abstract
Autophagy is a catabolic pathway with multifaceted roles in cellular homeostasis. This process is also involved in the antiviral response at multiple levels, including the direct elimination of intruding viruses (virophagy), the presentation of viral antigens, the fitness of immune cells, and the inhibition of excessive inflammatory reactions. In line with its central role in immunity, viruses have evolved mechanisms to interfere with or to evade the autophagic process, and in some cases, even to harness autophagy or constituents of the autophagic machinery for their replication. Given the devastating consequences of the current COVID-19 pandemic, the question arises whether manipulating autophagy might be an expedient approach to fight the novel coronavirus SARS-CoV-2. In this piece, we provide a short overview of the evidence linking autophagy to coronaviruses and discuss whether such links may provide actionable targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Maria A. Bauer
- Institute for Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andreas Zimmermann
- Institute for Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioHealth Graz, Graz, Austria
| | - Katharina Kainz
- Institute for Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Sebastian J. Hofer
- Institute for Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Frank Madeo
- Institute for Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioHealth Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
115
|
Wang L, Tan Z, Zhang Y, Kady Keita N, Liu H, Zhang Y. ADAM12 silencing promotes cellular apoptosis by activating autophagy in choriocarcinoma cells. Int J Oncol 2020; 56:1162-1174. [PMID: 32319603 PMCID: PMC7115740 DOI: 10.3892/ijo.2020.5007] [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: 09/16/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
ADAM metallopeptidase domain 12 (ADAM12) has been demonstrated to mediate cell proliferation and apoptosis resistance in several types of cancer cells. However, the effect of ADAM12 silencing on the proliferation and apoptosis of choriocarcinoma cells remains unknown. The present study revealed that ADAM12 silencing significantly inhibited cellular activity and proliferation in the human choriocarcinoma JEG3 cell line and increased the rate of apoptosis. In addition, ADAM12 silencing significantly increased the expression levels of the autophagy proteins microtubule-associated protein-light-chain 3 (LC3B) and autophagy related 5 (ATG5) and the fluorescence density of LC3B in JEG-3 cells. However, the suppression of autophagy by 3-methyladenine could block ADAM12 silencing-induced cellular apoptosis. ADAM12 silencing reduced the levels of the inflammatory factors interleukin-1β, interferon-γ and TNF-α, and inactivated nuclear p65-NF-κB and p-mTOR in JEG-3 cells. The downregulation of p-mTOR expression by ADAM12 silencing was rescued in 3-methyladenine-treated JEG-3 cells, indicating that mTOR might participate in the autophagy-mediated pro-apoptotic effect of ADAM12 silencing. In conclusion, ADAM12 silencing promoted cellular apoptosis in human choriocarcinoma JEG3 cells, which might be associated with autophagy and the mTOR response. These findings indicate that ADAM12 silencing might be a potential novel therapeutic target for choriocarcinoma.
Collapse
Affiliation(s)
- Lin Wang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhihui Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ying Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Nankoria Kady Keita
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Huining Liu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
116
|
Lu W, Mei J, Yang J, Wu Z, Liu J, Miao P, Chen Y, Wen Z, Zhao Z, Kong H, Wu C, Yang Y, Chen M. ApoE deficiency promotes non-alcoholic fatty liver disease in mice via impeding AMPK/mTOR mediated autophagy. Life Sci 2020; 252:117601. [PMID: 32304762 DOI: 10.1016/j.lfs.2020.117601] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/11/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
AIM This work was to investigate the relationship between ApoE and autophagy regulated by AMPK/mTOR pathway in the pathological process of NAFLD. MAIN METHODS Both WT and ApoE-/- mice were divided into two groups and allocated into either a normal chow (ND) or a high-fat diet (HFD) for 8 weeks. After that, we detected the indicators of lipid accumulation, hepatic injury, mitochondrial function hallmark, autophagy, apoptosis, inflammation, and oxidative stress by commercially available kits, immunohistochemistry, immunofluorescent staining, and western blot. KEY FINDING We found the lipid levels of serum and liver, and hepatic injury were significantly increased in the ApoE-/--HFD group compared to other groups. ApoE-/- mice exhibited increased deposition of fat in liver tissue. The PGC1α, NRF1, ATP, p-AMPK, AMPK, Beclin1, and LC3 levels were downregulated and ROS, p-mTOR, and mTOR were increased in the ApoE-/--HFD group compared to WT-HFD group. When treated with AMPK and autophagy activators, AICAR and rapamycin, these pathologies and protein levels can be rescued. The expression levels of apoptosis-related proteins, inflammation, and oxidative stress were increased in the ApoE-/--HFD group compared to the WT-HFD group. SIGNIFICANCE Our results indicated that ApoE deficiency can regulate AMPK/mTOR pathway, which leads to NAFLD most likely by modulating hepatic mitochondrial function.
Collapse
Affiliation(s)
- Wanpeng Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jinyu Mei
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Juan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhihan Wu
- First Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Jiayuan Liu
- First Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Pengyu Miao
- First Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Yiliang Chen
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhenfan Wen
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongting Zhao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Hua Kong
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Chao Wu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, 117 Meishan Road, Hefei 230031, China
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
117
|
Emran AA, Tseng HY, Coleman MC, Tiffen J, Cook S, McGuire HM, Gallagher S, Feng C, Hersey P. Do innate killing mechanisms activated by inflammasomes have a role in treating melanoma? Pigment Cell Melanoma Res 2020; 33:660-670. [PMID: 32027447 PMCID: PMC7497247 DOI: 10.1111/pcmr.12870] [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: 11/27/2019] [Revised: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 12/18/2022]
Abstract
Melanoma, as for many other cancers, undergoes a selection process during progression that limits many innate and adaptive tumor control mechanisms. Immunotherapy with immune checkpoint blockade overcomes one of the escape mechanisms but if the tumor is not eliminated other escape mechanisms evolve that require new approaches for tumor control. Some of the innate mechanisms that have evolved against infections with microorganisms and viruses are proving to be active against cancer cells but require better understanding of how they are activated and what inhibitory mechanisms may need to be targeted. This is particularly so for inflammasomes which have evolved against many different organisms and which recruit a number of cytotoxic mechanisms that remain poorly understood. Equally important is understanding of where these mechanisms will fit into existing treatment strategies and whether existing strategies already involve the innate killing mechanisms.
Collapse
Affiliation(s)
- Abdullah Al Emran
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Hsin-Yi Tseng
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Mikaela C Coleman
- Immunology and Host Defence Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Camperdown, New South Wales, Australia
| | - Jessamy Tiffen
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Stuart Cook
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Helen M McGuire
- Ramaciotti Facility for Human Systems Biology, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Stuart Gallagher
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Carl Feng
- Immunology and Host Defence Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Camperdown, New South Wales, Australia
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, The Centenary Institute, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
118
|
Gusev EY, Zotova NV. Cellular Stress and General Pathological Processes. Curr Pharm Des 2020; 25:251-297. [PMID: 31198111 DOI: 10.2174/1381612825666190319114641] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023]
Abstract
From the viewpoint of the general pathology, most of the human diseases are associated with a limited number of pathogenic processes such as inflammation, tumor growth, thrombosis, necrosis, fibrosis, atrophy, pathological hypertrophy, dysplasia and metaplasia. The phenomenon of chronic low-grade inflammation could be attributed to non-classical forms of inflammation, which include many neurodegenerative processes, pathological variants of insulin resistance, atherosclerosis, and other manifestations of the endothelial dysfunction. Individual and universal manifestations of cellular stress could be considered as a basic element of all these pathologies, which has both physiological and pathophysiological significance. The review examines the causes, main phenomena, developmental directions and outcomes of cellular stress using a phylogenetically conservative set of genes and their activation pathways, as well as tissue stress and its role in inflammatory and para-inflammatory processes. The main ways towards the realization of cellular stress and its functional blocks were outlined. The main stages of tissue stress and the classification of its typical manifestations, as well as its participation in the development of the classical and non-classical variants of the inflammatory process, were also described. The mechanisms of cellular and tissue stress are structured into the complex systems, which include networks that enable the exchange of information with multidirectional signaling pathways which together make these systems internally contradictory, and the result of their effects is often unpredictable. However, the possible solutions require new theoretical and methodological approaches, one of which includes the transition to integral criteria, which plausibly reflect the holistic image of these processes.
Collapse
Affiliation(s)
- Eugeny Yu Gusev
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation
| | - Natalia V Zotova
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation.,Department of Medical Biochemistry and Biophysics, Ural Federal University named after B.N.Yeltsin, Yekaterinburg, Russian Federation
| |
Collapse
|
119
|
Kang R, Zeh H, Lotze M, Tang D. The Multifaceted Effects of Autophagy on the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:99-114. [PMID: 32030650 DOI: 10.1007/978-3-030-35727-6_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is composed of cancer cells, noncancer cells (e.g., immune cells, stromal cells, endothelial cells, and adipocytes), and various mediators (e.g., cytokines, chemokines, growth factors, and humoral factors) that work together to support cancer growth, progression, and resistance to therapies. Autophagy is an evolutionarily conserved degradation mechanism by which various cytosolic cargos (e.g., damaged organelles, unused molecules, or invaded pathogens) are engulfed by double-membrane autophagosomes, and then delivered into the lysosome for degradation and recycling. The level of autophagy is a crucial threshold to either promote cell survival or induce cell death in response to environmental stresses. Autophagy plays a context-dependent role in tumorigenesis and anticancer therapy via shaping the inflammatory, hypoxic, immunosuppressive, and metabolic tumor microenvironment. In particular, impaired autophagy flux is associated with chronic inflammation, immunosuppression, stromal formation, cancer stemness, angiogenesis, metastasis, and metabolic reprogramming in the tumor microenvironment. Understanding the molecular machinery of autophagy and its communication with hallmarks of cancer could lead to potential new anticancer strategies or drugs.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
120
|
Chávez MN, Morales RA, López-Crisosto C, Roa JC, Allende ML, Lavandero S. Autophagy Activation in Zebrafish Heart Regeneration. Sci Rep 2020; 10:2191. [PMID: 32042056 PMCID: PMC7010704 DOI: 10.1038/s41598-020-59106-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a key role in the maintenance of overall cellular health. While it has been suggested that autophagy may elicit cardioprotective and pro-survival modulating functions, excessive activation of autophagy can also be detrimental. In this regard, the zebrafish is considered a hallmark model for vertebrate regeneration, since contrary to adult mammals, it is able to faithfully regenerate cardiac tissue. Interestingly, the role that autophagy may play in zebrafish heart regeneration has not been studied yet. In the present work, we hypothesize that, in the context of a well-established injury model of ventricular apex resection, autophagy plays a critical role during cardiac regeneration and its regulation can directly affect the zebrafish regenerative potential. We studied the autophagy events occurring upon injury using electron microscopy, in vivo tracking of autophagy markers, and protein analysis. Additionally, using pharmacological tools, we investigated how rapamycin, an inducer of autophagy, affects regeneration relevant processes. Our results show that a tightly regulated autophagic response is triggered upon injury and during the early stages of the regeneration process. Furthermore, treatment with rapamycin caused an impairment in the cardiac regeneration outcome. These findings are reminiscent of the pathophysiological description of an injured human heart and hence put forward the zebrafish as a model to study the poorly understood double-sword effect that autophagy has in cardiac homeostasis.
Collapse
Affiliation(s)
- Myra N Chávez
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Rodrigo A Morales
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel L Allende
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
121
|
Macrophage-Based Therapies for Atherosclerosis Management. J Immunol Res 2020; 2020:8131754. [PMID: 32411803 PMCID: PMC7204102 DOI: 10.1155/2020/8131754] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/21/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis (AS), a typical chronic inflammatory vascular disease, is the main pathological basis of ischemic cardio/cerebrovascular disease (CVD). Long-term administration was characterized with low efficacy and serious side effects, while the macrophages with attractive intrinsic homing target have great potential in the efficient and safe management of AS. In this review, we focused on the systematical summary of the macrophage-based therapies in AS management, including macrophage autophagy, polarization, targeted delivery, microenvironment-triggered drug release, and macrophage- or macrophage membrane-based drug carrier. In conclusion, macrophage-based therapies have great promise to effectively manage AS in future research and clinic translation.
Collapse
|
122
|
Hsieh CY, Li LH, Lam Y, Fang Z, Gan CH, Rao YK, Chiu HW, Wong WT, Ju TC, Chen FH, Chernikov OV, Liu ML, Hsu CH, Hua KF. Synthetic 4-Hydroxy Auxarconjugatin B, a Novel Autophagy Inducer, Attenuates Gouty Inflammation by Inhibiting the NLRP3 Inflammasome. Cells 2020; 9:cells9020279. [PMID: 31979265 PMCID: PMC7072356 DOI: 10.3390/cells9020279] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/29/2022] Open
Abstract
Gouty arthritis results from the generation of uric acid crystals within the joints. These uric acid crystals activate the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome, which is involved in chronic inflammatory diseases, including gouty arthritis. This study identified the polyenylpyrrole derivative 4-hydroxy auxarconjugatin B (4-HAB), a novel autophagy inducer, which attenuated uric acid crystals-mediated activation of the NLRP3 inflammasome in vitro and in vivo. 4-HAB dose-dependently reduced the release of interleukin (IL)-1β, IL-18, active caspase-1 and apoptosis-associated speck-like protein (ASC) in uric acid crystals-activated macrophages. In a mechanistic study, 4-HAB was shown to inhibit uric acid crystals-induced mitochondrial damage, lysosomal rupture and ASC oligomerization. Additionally, 4-HAB inhibited the NLRP3 inflammasome through Sirt1-dependent autophagy induction. Furthermore, the anti-inflammatory properties of 4-HAB were confirmed in a mouse model of uric acid crystals-mediated peritonitis by the reduced levels of neutrophil influx, IL-1β, active caspase-1, IL-6 and MCP-1 in lavage fluids. In conclusion, 4-HAB attenuates gouty inflammation, in part by attenuating activation of the NLRP3 inflammasome through the Sirt1/autophagy induction pathway.
Collapse
Affiliation(s)
- Chih-Yu Hsieh
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan;
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11483, Taiwan
| | - Yulin Lam
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Zhanxiong Fang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Chin Heng Gan
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Yerra Koteswara Rao
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Hsiao-Wen Chiu
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Wei-Ting Wong
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Tz-Chuen Ju
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan;
| | - Fang-Hsin Chen
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Oleg V. Chernikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, Vladivostok 690022, Russia;
| | - May-Lan Liu
- Department of Nutritional Science, Toko University, Chiayi 61363, Taiwan;
| | - Chung-Hua Hsu
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Chinese Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11483, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Correspondence: ; Tel.: +886-3931-7630
| |
Collapse
|
123
|
Peng Z, Zhan H, Shao Y, Xiong Y, Zeng L, Zhang C, Liu Z, Huang Z, Su H, Yang Z. 13-Methylberberine improves endothelial dysfunction by inhibiting NLRP3 inflammasome activation via autophagy induction in human umbilical vein endothelial cells. Chin Med 2020; 15:8. [PMID: 31993073 PMCID: PMC6977264 DOI: 10.1186/s13020-020-0286-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atherosclerosis, the underlying cause of the majority of cardiovascular diseases, is a lipid-driven, inflammatory disease of the large arteries. Atherosclerotic cardiovascular disease (ASCVD) threatens human lives due to high morbidity and mortality. Many studies have demonstrated that atherosclerosis is accelerated via activation of the NLRP3 inflammasome. The NLRP3 inflammasome plays a critical role in the development of vascular inflammation and atherosclerosis. In atherosclerotic plaques, excessive generation of reactive oxygen species (ROS) activates the NLRP3 inflammasome. 13-Methylberberine (13-MB) is a newly synthesized compound used in traditional Chinese medicine that has outstanding antibacterial, antitumor, and antiobesity activities, especially anti-inflammatory activity. However, the role of 13-MB in atherosclerosis needs to be explored. METHODS CCK-8 assays and flow cytometry were conducted to determine the cell viability and apoptotic profiles of human umbilical vein endothelial cells (HUVECs) treated with 13-MB. Carboxy-DCFH-DA and JC-10 assays were used to measure ROS and determine mitochondrial membrane potential. Western blot analysis was performed to investigate proteins that are associated with the NLRP3 inflammasome and autophagy. ELISA was used to detect and quantify inflammatory cytokines related to the NLRP3 inflammasome. Transfection and confocal microscopy were conducted to observe autophagy. RESULTS Pretreatment with 13-MB markedly reduced cytotoxicity and apoptosis, as well as intracellular ROS production, in H2O2-induced HUVECs. Moreover, 13-MB showed a protective effect in maintaining mitochondrial membrane potential. 13-MB also suppressed NLRP3 inflammasome activation and promoted autophagy induction in HUVECs. CONCLUSION 13-MB exerts cytoprotective effects in an H2O2-induced cell injury model by inhibiting NLRP3 inflammasome activation via autophagy induction in HUVECs. These anti-inflammatory and autophagy induction activities may provide valuable evidence for further investigating the potential role of 13-MB in atherosclerosis.
Collapse
Affiliation(s)
- Zhihua Peng
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Hong Zhan
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Yijia Shao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
| | - Yan Xiong
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Lijin Zeng
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Cong Zhang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhihao Liu
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhenhua Huang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhen Yang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| |
Collapse
|
124
|
De Biase D, Piegari G, Prisco F, Cimmino I, Pirozzi C, Mattace Raso G, Oriente F, Grieco E, Papparella S, Paciello O. Autophagy and NLRP3 inflammasome crosstalk in neuroinflammation in aged bovine brains. J Cell Physiol 2020; 235:5394-5403. [PMID: 31903559 DOI: 10.1002/jcp.29426] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
NLRP3 inflammasome is a multiprotein complex that can sense several stimuli such as autophagy dysregulation and increased reactive oxygen species production stimulating inflammation by priming the maturation of proinflammatory cytokines interleukin-1β and interleukin-18 in their active form. In the aging brain, these cytokines can mediate the innate immunity response priming microglial activation. Here, we describe the results of immunohistochemical and molecular analysis carried out on bovine brains. Our results support the hypothesis that the age-related impairment in cellular housekeeping mechanisms and the increased oxidative stress can trigger the inflammatory danger sensor NLRP3. Moreover, according to the recent scientific literature, we demonstrate the presence of an age-related proinflammatory environment in aged brains consisting in an upregulation of interleukin-1β, an increased microglial activation and increased NLRP3 expression. Finally, we suggest that bovine may potentially be a pivotal animal model for brain aging studies.
Collapse
Affiliation(s)
- Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Piegari
- Department of Veterinary Medicine and Animal Production, University of Naples "Federico II", Naples, Italy
| | - Francesco Prisco
- Department of Veterinary Medicine and Animal Production, University of Naples "Federico II", Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | | | - Francesco Oriente
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | | | - Serenella Papparella
- Department of Veterinary Medicine and Animal Production, University of Naples "Federico II", Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
125
|
Qiu Q, Yang Z, Cao F, Yang C, Hardy P, Yan X, Yang S, Xiong W. Activation of NLRP3 inflammasome by lymphocytic microparticles via TLR4 pathway contributes to airway inflammation. Exp Cell Res 2019; 386:111737. [PMID: 31759058 DOI: 10.1016/j.yexcr.2019.111737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 01/23/2023]
Abstract
The presence of elevated T lymphocytic microparticles (TLMPs) during respiratory illness is associated with airway and lung inflammation and epithelial injuries. Although inflammasome and IL-1β signaling are crucial in airway inflammation, little was known about their regulatory mechanism. We hypothesized that TLMPs trigger inflammasome activation and IL-1β production in bronchial and alveolar epithelial cells to induce airway and lung inflammation. In this study, TLMPs induced IL-1β and IL-18 secretion through NLRP3 inflammasome activation and upregulated TLR4 mRNA and protein expression in alveolar (A549) and human airway epithelial (16HBE) cells. Pretreatment with CLI-095, a specific inhibitor of TLR4 signaling, dramatically diminished the TLMP-induced release of IL-1β and IL-18 by inhibiting the formation of NLRP3/ASC/pro-caspase-1 inflammasome in a dose-dependent manner. The TLMP-induced autophagy inhibition in epithelial cells was dependent on the PI3K/Akt signaling pathway, which significantly increased NLRP3 expression and enhanced TLMP-induced inflammation. TLR4, IL-1β, and IL-18 proteins harbored in TLMPs were nonessential for the pro-inflammatory effect. In conclusion, TLMPs induce bronchial and alveolar epithelial cell secretion of IL-1β and IL-18 cytokines by activating the TLR4 and PI3K/Akt signaling pathways and inhibiting autophagy. These effects lead to NLRP3 inflammasome formation and accumulation. TLMPs may be regarded as deleterious markers of airway and lung damage in respiratory diseases.
Collapse
Affiliation(s)
- Qian Qiu
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China; Research Institute of Tuberculosis, Chongqing Public Health Medical Center, Chongqing, 400036, China
| | - Zaixing Yang
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Fuli Cao
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Chun Yang
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Pierre Hardy
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Xiaofeng Yan
- Research Institute of Tuberculosis, Chongqing Public Health Medical Center, Chongqing, 400036, China
| | - Song Yang
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Wei Xiong
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
126
|
Danielski LG, Giustina AD, Bonfante S, Barichello T, Petronilho F. The NLRP3 Inflammasome and Its Role in Sepsis Development. Inflammation 2019; 43:24-31. [DOI: 10.1007/s10753-019-01124-9] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
127
|
Javaheri A, Bajpai G, Picataggi A, Mani S, Foroughi L, Evie H, Kovacs A, Weinheimer CJ, Hyrc K, Xiao Q, Ballabio A, Lee JM, Matkovich SJ, Razani B, Schilling JD, Lavine KJ, Diwan A. TFEB activation in macrophages attenuates postmyocardial infarction ventricular dysfunction independently of ATG5-mediated autophagy. JCI Insight 2019; 4:127312. [PMID: 31672943 DOI: 10.1172/jci.insight.127312] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
Lysosomes are at the epicenter of cellular processes critical for inflammasome activation in macrophages. Inflammasome activation and IL-1β secretion are implicated in myocardial infarction (MI) and resultant heart failure; however, little is known about how macrophage lysosomes regulate these processes. In mice subjected to cardiac ischemia/reperfusion (IR) injury and humans with ischemic cardiomyopathy, we observed evidence of lysosomal impairment in macrophages. Inducible macrophage-specific overexpression of transcription factor EB (TFEB), a master regulator of lysosome biogenesis (Mϕ-TFEB), attenuated postinfarction remodeling, decreased abundance of proinflammatory macrophages, and reduced levels of myocardial IL-1β compared with controls. Surprisingly, neither inflammasome suppression nor Mϕ-TFEB-mediated attenuation of postinfarction myocardial dysfunction required intact ATG5-dependent macroautophagy (hereafter termed "autophagy"). RNA-seq of flow-sorted macrophages postinfarction revealed that Mϕ-TFEB upregulated key targets involved in lysosomal lipid metabolism. Specifically, inhibition of the TFEB target, lysosomal acid lipase, in vivo abrogated the beneficial effect of Mϕ-TFEB on postinfarction ventricular function. Thus, TFEB reprograms macrophage lysosomal lipid metabolism to attenuate remodeling after IR, suggesting an alternative paradigm whereby lysosome function affects inflammation.
Collapse
Affiliation(s)
- Ali Javaheri
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Geetika Bajpai
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Antonino Picataggi
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Smrithi Mani
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Layla Foroughi
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Hosannah Evie
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Attila Kovacs
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Carla J Weinheimer
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | | | - Qingli Xiao
- Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Jin-Moo Lee
- Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Scot J Matkovich
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Babak Razani
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine.,John Cochran Veterans Affairs Medical Center, Saint Louis, Missouri, USA
| | - Joel D Schilling
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Kory J Lavine
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine
| | - Abhinav Diwan
- Cardiovascular Division and Center for Cardiovascular Research, Department of Medicine.,John Cochran Veterans Affairs Medical Center, Saint Louis, Missouri, USA
| |
Collapse
|
128
|
Huang X, Kuang S, Shen Z, Liang M, Lin Z. High glucose disrupts autophagy lysosomal pathway in gingival epithelial cells via ATP6V0C. J Periodontol 2019; 91:705-714. [PMID: 31471894 DOI: 10.1002/jper.19-0262] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/05/2019] [Accepted: 07/07/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Xin Huang
- Department of PeriodontologyGuanghua School and Hospital of StomatologySun Yat‐Sen University Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityNorth Campus Guangzhou Guangdong China
| | - Shuhong Kuang
- Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityNorth Campus Guangzhou Guangdong China
- Department of Operative Dentistry and EndodonticsGuanghua School and Hospital of StomatologySun Yat‐Sen University Guangzhou Guangdong China
| | - Zongshan Shen
- Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityNorth Campus Guangzhou Guangdong China
- Department of Operative Dentistry and EndodonticsGuanghua School and Hospital of StomatologySun Yat‐Sen University Guangzhou Guangdong China
| | - Min Liang
- Department of PeriodontologyGuanghua School and Hospital of StomatologySun Yat‐Sen University Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityNorth Campus Guangzhou Guangdong China
| | - Zhengmei Lin
- Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityNorth Campus Guangzhou Guangdong China
- Department of Operative Dentistry and EndodonticsGuanghua School and Hospital of StomatologySun Yat‐Sen University Guangzhou Guangdong China
| |
Collapse
|
129
|
Kumar V. Sepsis roadmap: What we know, what we learned, and where we are going. Clin Immunol 2019; 210:108264. [PMID: 31655168 DOI: 10.1016/j.clim.2019.108264] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 07/02/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening condition originating as a result of systemic blood infection causing, one or more organ damage due to the dysregulation of the immune response. In 2017, the world health organization (WHO) declared sepsis as a disease of global health priority, needing special attention due to its high prevalence and mortality around the world. Most of the therapeutics targeting sepsis have failed in the clinics. The present review highlights the history of the sepsis, its immunopathogenesis, and lessons learned after the failure of previously used immune-based therapies. The subsequent section, where to go describes in details the importance of the complement system (CS), autophagy, inflammasomes, and microbiota along with their targeting to manage sepsis. These systems are interconnected to each other, thus targeting one may affect the other. We are in an urgent need for a multi-targeting therapeutic approach for sepsis.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
130
|
Shi CS, Nabar NR, Huang NN, Kehrl JH. SARS-Coronavirus Open Reading Frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Dis 2019. [PMID: 31231549 DOI: 10.1038/s41420‐019‐0181‐7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The SARS (severe acute respiratory syndrome) outbreak was caused by a coronavirus (CoV) named the SARS-CoV. SARS pathology is propagated both by direct cytotoxic effects of the virus and aberrant activation of the innate immune response. Here, we identify several mechanisms by which a SARS-CoV open reading frame (ORF) activates intracellular stress pathways and targets the innate immune response. We show that ORF8b forms insoluble intracellular aggregates dependent on a valine at residue 77. Aggregated ORF8b induces endoplasmic reticulum (ER) stress, lysosomal damage, and subsequent activation of the master regulator of the autophagy and lysosome machinery, Transcription factor EB (TFEB). ORF8b causes cell death in epithelial cells, which is partially rescued by reducing its ability to aggregate. In macrophages, ORF8b robustly activates the NLRP3 inflammasome by providing a potent signal 2 required for activation. Mechanistically, ORF8b interacts directly with the Leucine Rich Repeat domain of NLRP3 and localizes with NLRP3 and ASC in cytosolic dot-like structures. ORF8b triggers cell death consistent with pyroptotic cell death in macrophages. While in those cells lacking NLRP3 accumulating ORF8b cytosolic aggregates cause ER stress, mitochondrial dysfunction, and caspase-independent cell death.
Collapse
Affiliation(s)
- Chong-Shan Shi
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Neel R Nabar
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ning-Na Huang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - John H Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
131
|
Shi CS, Nabar NR, Huang NN, Kehrl JH. SARS-Coronavirus Open Reading Frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Discov 2019; 5:101. [PMID: 31231549 PMCID: PMC6549181 DOI: 10.1038/s41420-019-0181-7] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
The SARS (severe acute respiratory syndrome) outbreak was caused by a coronavirus (CoV) named the SARS-CoV. SARS pathology is propagated both by direct cytotoxic effects of the virus and aberrant activation of the innate immune response. Here, we identify several mechanisms by which a SARS-CoV open reading frame (ORF) activates intracellular stress pathways and targets the innate immune response. We show that ORF8b forms insoluble intracellular aggregates dependent on a valine at residue 77. Aggregated ORF8b induces endoplasmic reticulum (ER) stress, lysosomal damage, and subsequent activation of the master regulator of the autophagy and lysosome machinery, Transcription factor EB (TFEB). ORF8b causes cell death in epithelial cells, which is partially rescued by reducing its ability to aggregate. In macrophages, ORF8b robustly activates the NLRP3 inflammasome by providing a potent signal 2 required for activation. Mechanistically, ORF8b interacts directly with the Leucine Rich Repeat domain of NLRP3 and localizes with NLRP3 and ASC in cytosolic dot-like structures. ORF8b triggers cell death consistent with pyroptotic cell death in macrophages. While in those cells lacking NLRP3 accumulating ORF8b cytosolic aggregates cause ER stress, mitochondrial dysfunction, and caspase-independent cell death.
Collapse
Affiliation(s)
- Chong-Shan Shi
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Neel R Nabar
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ning-Na Huang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - John H Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
132
|
Evodiamine alleviates kidney ischemia reperfusion injury in rats: A biochemical and histopathological study. J Cell Biochem 2019; 120:17159-17166. [DOI: 10.1002/jcb.28976] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/14/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022]
|
133
|
Matteucci KC, Pereira GJS, Weinlich R, Bortoluci KR. Frontline Science: Autophagy is a cell autonomous effector mechanism mediated by NLRP3 to controlTrypanosoma cruziinfection. J Leukoc Biol 2019; 106:531-540. [DOI: 10.1002/jlb.hi1118-461r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Kely C. Matteucci
- Departamento de Ciências Biológicas e Centro de Terapia Celular e Molecular (CTC‐Mol)UNIFESP São Paulo Brazil
| | | | - Ricardo Weinlich
- Instituto de Ensino e PesquisaHospital Israelita Albert Einstein São Paulo Brazil
| | - Karina R. Bortoluci
- Departamento de Ciências Biológicas e Centro de Terapia Celular e Molecular (CTC‐Mol)UNIFESP São Paulo Brazil
| |
Collapse
|
134
|
Kumar V. The complement system, toll-like receptors and inflammasomes in host defense: three musketeers’ one target. Int Rev Immunol 2019; 38:131-156. [PMID: 31066339 DOI: 10.1080/08830185.2019.1609962] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vijay Kumar
- Children’s Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, QLD, Australia
| |
Collapse
|
135
|
Rapamycin improves the neuroprotection effect of inhibition of NLRP3 inflammasome activation after TBI. Brain Res 2019; 1710:163-172. [DOI: 10.1016/j.brainres.2019.01.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 01/02/2023]
|
136
|
Harris J, VanPatten S, Deen NS, Al-Abed Y, Morand EF. Rediscovering MIF: New Tricks for an Old Cytokine. Trends Immunol 2019; 40:447-462. [DOI: 10.1016/j.it.2019.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
|
137
|
Tan DX. Aging: An evolutionary competition between host cells and mitochondria. Med Hypotheses 2019; 127:120-128. [PMID: 31088635 DOI: 10.1016/j.mehy.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/20/2019] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
Here, a new theory of aging is proposed. This new theory is referred as the Host-Mitochondria Intracellular Innate Immune Theory of Aging (HMIIITA). The main point of this theory is that the aging is rooted from an evolutionary competition, that is, a never ending coevolutionary race between host cells and mitochondria. Mitochondria are the descendants of bacteria. The host cells will inevitably sense their bacterial origin, particularly their circular mtDNA. The host intracellular innate immune pressure (HIIIP) aims to eliminate mtDNA as more as possible while mitochondria have to adapt the HIIIP for survival. Co-evolution is required for both of them. From biological point of view, the larger, the mtDNA, the higher, the chance, it becomes the target of HIIIP. As a result, mitochondria have to reduce their mtDNA size via deletion. This process has last for 1.5-2 billion yeas and the result is that mitochondria have lost excessive 95% of their DNA. This mtDNA deletion is not associated with free radical attack but a unique trait acquired during evolution. In the postmitotic cells, the deletion is passively selected by the mitochondrial fission-fusion cycles. Eventually, the accumulation of deletion will significantly jeopardize the mitochondrial function. The dysfunctional mitochondria no longer provide sufficient ATP to support host cells' continuous demanding for growth. At this stage, the cell or the organism aging is inevitable.
Collapse
Affiliation(s)
- Dun-Xian Tan
- The Department of Cell System and Anatomy, The University of Texas, Health, San Antonio, TX 78229, USA.
| |
Collapse
|
138
|
Kumar V. A STING to inflammation and autoimmunity. J Leukoc Biol 2019; 106:171-185. [PMID: 30990921 DOI: 10.1002/jlb.4mir1018-397rr] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022] Open
Abstract
Various intracellular pattern recognition receptors (PRRs) recognize cytosolic pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Cyclic GMP-AMP synthase (cGAS), a cytosolic PRR, recognizes cytosolic nucleic acids including dsDNAs. The recognition of dsDNA by cGAS generates cyclic GMP-AMP (GAMP). The cGAMP is then recognized by STING generating type 1 IFNs and NF-κB-mediated generation of pro-inflammatory cytokines and molecules. Thus, cGAS-STING signaling mediated recognition of cytosolic dsDNA causing the induction of type 1 IFNs plays a crucial role in innate immunity against cytosolic pathogens, PAMPs, and DAMPs. The overactivation of this system may lead to the development of autoinflammation and autoimmune diseases. The article opens with the introduction of different PRRs involved in the intracellular recognition of dsDNA and gives a brief introduction of cGAS-STING signaling. The second section briefly describes cGAS as intracellular PRR required to recognize intracellular nucleic acids (dsDNA and CDNs) and the formation of cGAMP. The cGAMP acts as a second messenger to activate STING- and TANK-binding kinase 1-mediated generation of type 1 IFNs and the activation of NF-κB. The third section of the article describes the role of cGAS-STING signaling in the induction of autoinflammation and various autoimmune diseases. The subsequent fourth section describes both chemical compounds developed and the endogenous negative regulators of cGAS-STING signaling required for its regulation. Therapeutic targeting of cGAS-STING signaling could offer new ways to treat inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
139
|
Glucosamine inhibits IL-1β expression by preserving mitochondrial integrity and disrupting assembly of the NLRP3 inflammasome. Sci Rep 2019; 9:5603. [PMID: 30944389 PMCID: PMC6447579 DOI: 10.1038/s41598-019-42130-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 03/21/2019] [Indexed: 12/18/2022] Open
Abstract
The NLRP3 inflammasome promotes the pathogenesis of metabolic, neurodegenerative and infectious diseases. Increasing evidences show that the NLRP3 inflammasome is a promising therapeutic target in inflammatory diseases. Glucosamine is widely used as a dietary supplement to promote the health of cartilage tissue and is expected to exert anti-inflammatory activity in joint inflammation, which is a nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome-associated complication. Here, we investigated whether GlcN inhibits the NLRP3 inflammasome and dissected the underlying molecular mechanisms. We found that GlcN suppressed the NLRP3 inflammasome in mouse and human macrophages. A mechanistic study revealed that GlcN inhibited the expression of NLRP3 and IL-1β precursor by reducing reactive oxygen species generation and NF-κB activation in lipopolysaccharide-activated macrophages. GlcN also suppressed mitochondrial reactive oxygen species generation and mitochondrial integrity loss in NLRP3-activated macrophages. Additionally, GlcN disrupted NLRP3 inflammasome assembly by inhibiting NLRP3 binding to PKR, NEK7 and ASC. Furthermore, oral administration of GlcN reduced peritoneal neutrophils influx and lavage fluids concentrations of IL-1β, IL-6 MCP-1 and TNF-α in uric acid crystal-injected mice. These results indicated that GlcN might be a novel dietary supplement for the amelioration of NLRP3 inflammasome-associated complications.
Collapse
|
140
|
Feng Y, Liu B, Zheng X, Chen L, Chen W, Fang Z. The protective role of autophagy in sepsis. Microb Pathog 2019; 131:106-111. [PMID: 30935962 DOI: 10.1016/j.micpath.2019.03.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Sepsis is characterized by life-threatening organ dysfunction caused by a deregulated host response to infection. Autophagy is one of the innate immune defense mechanisms against microbial attack. Previous studies have demonstrated that autophagy is activated initially in sepsis, followed by a subsequent phase of impairment. A number of sepsis-related studies have shown that autophagy plays a protective role in multiple organ injuries partly by clearing pathogens, regulating inflammation and metabolism, inhibiting apoptosis and suppressing immune reactions. In this review, we present a general overview of and recent advances in the role of autophagy in sepsis and consider the therapeutic potential of autophagy activators in treating sepsis.
Collapse
Affiliation(s)
- Ying Feng
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China; Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Boyi Liu
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Xiang Zheng
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Li Chen
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Wei Chen
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Zhicheng Fang
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China.
| |
Collapse
|
141
|
Wortmann M, Xiao X, Wabnitz G, Samstag Y, Hakimi M, Böckler D, Dihlmann S. AIM2 levels and DNA-triggered inflammasome response are increased in peripheral leukocytes of patients with abdominal aortic aneurysm. Inflamm Res 2019; 68:337-345. [DOI: 10.1007/s00011-019-01212-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/03/2018] [Accepted: 01/01/2019] [Indexed: 02/08/2023] Open
|
142
|
Lee J, Kim HS. The Role of Autophagy in Eosinophilic Airway Inflammation. Immune Netw 2019; 19:e5. [PMID: 30838160 PMCID: PMC6399092 DOI: 10.4110/in.2019.19.e5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/17/2019] [Accepted: 01/29/2019] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a homeostatic mechanism that discards not only invading pathogens but also damaged organelles and denatured proteins via lysosomal degradation. Increasing evidence suggests a role for autophagy in inflammatory diseases, including infectious diseases, Crohn's disease, cystic fibrosis, and pulmonary hypertension. These studies suggest that modulating autophagy could be a novel therapeutic option for inflammatory diseases. Eosinophils are a major type of inflammatory cell that aggravates airway inflammatory diseases, particularly corticosteroid-resistant inflammation. The eosinophil count is a useful tool for assessing which patients may benefit from inhaled corticosteroid therapy. Recent studies demonstrate that autophagy plays a role in eosinophilic airway inflammatory diseases by promoting airway remodeling and loss of function. Genetic variant in the autophagy gene ATG5 is associated with asthma pathogenesis, and autophagy regulates apoptotic pathways in epithelial cells in individuals with chronic obstructive pulmonary disease. Moreover, autophagy dysfunction leads to severe inflammation, especially eosinophilic inflammation, in chronic rhinosinusitis. However, the mechanism underlying autophagy-mediated regulation of eosinophilic airway inflammation remains unclear. The aim of this review is to provide a general overview of the role of autophagy in eosinophilic airway inflammation. We also suggest that autophagy may be a new therapeutic target for airway inflammation, including that mediated by eosinophils.
Collapse
Affiliation(s)
- Jinju Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hun Sik Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.,Department of Microbiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.,Stem Cell Immunomodulation Research Center (SCIRC), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
143
|
Autophagy attenuates tubulointerstital fibrosis through regulating transforming growth factor-β and NLRP3 inflammasome signaling pathway. Cell Death Dis 2019; 10:78. [PMID: 30692509 PMCID: PMC6349890 DOI: 10.1038/s41419-019-1356-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 01/24/2023]
Abstract
Renal fibrosis is the final common pathway of various renal injuries and it leads to chronic kidney disease. Autophagy is a cellular process of degradation of damaged cytoplasmic components and regulates cell death and proliferation. Cellular response during unilateral ureteral obstruction (UUO) is tubular segment specific. Thus the role of autophagy on renal tubulointerstitial fibrosis (TIF) after UUO may be different according to segment of nephron. The role of autophagy during UUO remains unclear especially in distal tubules. In this study, we investigated the role of autophagy in distal tubules on renal TIF using conditional knockout mice in which Atg7 was genetically ablated specifically in distal tubular epithelial cell (TEC). In green fluorescent protein (GFP)-LC3 transgenic mice, GFP-LC3 puncta was highly expressed in distal tubular cells of the obstructed kidneys after UUO. Genetic deletion of Atg7 specifically in distal TEC increased renal tubulointerstial fibrosis and epithelial-mesenchymal transition-like phenotype change after UUO through Smad4-dependent transforming growth factor (TGF)-β pathway. Distal tubule-specific autophagy-deficient mice increased the accumulation of damaged mitochondria and SQSTM1/p62-positive aggregates in the obstructed kidney and resulted in increased expression of NLRP3 inflammasome, interleukin (IL) 1-β and caspase-1. Distal TEC-specific Atg7 deletion enhanced apoptosis of TECs after UUO. In summary, our data showed that autophagy in distal TEC plays a protective role in development of renal tubulointerstial fibrosis through regulating the expression of TGF-β an IL1-β after UUO.
Collapse
|
144
|
Vural A, Nabar NR, Hwang IY, Sohn S, Park C, Karlsson MCI, Blumer JB, Kehrl JH. Gα i2 Signaling Regulates Inflammasome Priming and Cytokine Production by Biasing Macrophage Phenotype Determination. THE JOURNAL OF IMMUNOLOGY 2019; 202:1510-1520. [PMID: 30683698 DOI: 10.4049/jimmunol.1801145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022]
Abstract
Macrophages exist as innate immune subsets that exhibit phenotypic heterogeneity and functional plasticity. Their phenotypes are dictated by inputs from the tissue microenvironment. G-protein-coupled receptors are essential in transducing signals from the microenvironment, and heterotrimeric Gα signaling links these receptors to downstream effectors. Several Gαi-coupled G-protein-coupled receptors have been implicated in macrophage polarization. In this study, we use genetically modified mice to investigate the role of Gαi2 on inflammasome activity and macrophage polarization. We report that Gαi2 in murine bone marrow-derived macrophages (BMDMs) regulates IL-1β release after activation of the NLRP3, AIM2, and NLRC4 inflammasomes. We show this regulation stems from the biased polarity of Gαi2 deficient (Gnai2 -/-) and RGS-insensitive Gαi2 (Gnai2 G184S/G184S) BMDMs. We determined that although Gnai2 G184S/G184S BMDMs (excess Gαi2 signaling) have a tendency toward classically activated proinflammatory (M1) phenotype, Gnai2-/- BMDMs (Gαi2 deficient) are biased toward alternatively activated anti-inflammatory (M2) phenotype. Finally, we find that Gαi2-deficient macrophages have increased Akt activation and IFN-β production but defects in ERK1/2 and STAT3 activation after LPS stimulation. Gαi2-deficient macrophages also exhibit increased STAT6 activation after IL-4 stimulation. In summary, our data indicates that excess Gαi2 signaling promotes an M1 macrophage phenotype, whereas Gαi2 signaling deficiency promotes an M2 phenotype. Understanding Gαi2-mediated effects on macrophage polarization may bring to light insights regarding disease pathogenesis and the reprogramming of macrophages for the development of novel therapeutics.
Collapse
Affiliation(s)
- Ali Vural
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Neel R Nabar
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; .,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; and
| | - Il-Young Hwang
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Silke Sohn
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; and
| | - Chung Park
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; and
| | - Joe B Blumer
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
145
|
Nam SA, Kim WY, Kim JW, Kang MG, Park SH, Lee MS, Kim HW, Yang CW, Kim J, Kim YK. Autophagy in FOXD1 stroma-derived cells regulates renal fibrosis through TGF-β and NLRP3 inflammasome pathway. Biochem Biophys Res Commun 2019; 508:965-972. [DOI: 10.1016/j.bbrc.2018.11.090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
|
146
|
Merkley SD, Chock CJ, Yang XO, Harris J, Castillo EF. Modulating T Cell Responses via Autophagy: The Intrinsic Influence Controlling the Function of Both Antigen-Presenting Cells and T Cells. Front Immunol 2018; 9:2914. [PMID: 30619278 PMCID: PMC6302218 DOI: 10.3389/fimmu.2018.02914] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a homeostatic and inducible process affecting multiple aspects of the immune system. This intrinsic cellular process is involved in MHC-antigen (Ag) presentation, inflammatory signaling, cytokine regulation, and cellular metabolism. In the context of T cell responses, autophagy has an influential hand in dictating responses to self and non-self by controlling extrinsic factors (e.g., MHC-Ag, cytokine production) in antigen-presenting cells (APC) and intrinsic factors (e.g., cell signaling, survival, cytokine production, and metabolism) in T cells. These attributes make autophagy an attractive therapeutic target to modulate T cell responses. In this review, we examine the impact autophagy has on T cell responses by modulating multiple aspects of APC function; the importance of autophagy in the activation, differentiation and homeostasis of T cells; and discuss how the modulation of autophagy could influence T cell responses.
Collapse
Affiliation(s)
- Seth D Merkley
- Clinical and Translational Science Center, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - Cameron J Chock
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Albuquerque, NM, United States.,Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - James Harris
- Rheumatology Group, Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Clayton, VIC, Australia
| | - Eliseo F Castillo
- Clinical and Translational Science Center, University of New Mexico Health Sciences Albuquerque, NM, United States.,Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Albuquerque, NM, United States.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine Albuquerque, NM, United States
| |
Collapse
|
147
|
Wu X, Zhang J, Ma C, Li W, Zeng J, Wang Y, Deng G. A role for Wnt/β-catenin signalling in suppressing Bacillus Calmette-Guerin-induced macrophage autophagy. Microb Pathog 2018; 127:277-287. [PMID: 30550847 DOI: 10.1016/j.micpath.2018.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022]
Abstract
Mycobacterium tuberculosis (Mtb)-induced autophagy of alveolar macrophages has been confirmed to play a central role in the pathogenesis of tuberculosis. Growing evidence indicates that excessive or uncontrolled autophagic activity, which results in type II programmed cell death, can be regulated by many factors, including Wnt/β-catenin signalling. Wnt/β-catenin signalling has been demonstrated to be involved in multiple diseases through the regulation of autophagy; however, its exact role in regulating autophagy induced by Mtb remains unclear. Accordingly, this study examined the function of the Wnt/β-catenin signalling pathway in regulating Mycobacterium bovis Bacillus Calmette-Guerin (BCG)-induced autophagy in RAW264.7 macrophage cell line. In the present study, we found that BCG induced the autophagy of RAW264.7 cells in a time- and dose-dependent manner along with an accumulation of LC3 (Microtubule-associated protein 1 light chain 3) protein. Intriguingly, Wnt3a, a Wnt/β-catenin signalling ligand, significantly inhibited autophagy, with decreased autophagy rates and autophagic flux. An immunoblot analysis further revealed that Wnt/β-catenin signalling was capable of inhibiting the expression of the LC3 and autophagy-associated gene (Atg) cascade proteins in BCG-infected cells. Mechanistically, Wnt/β-catenin signalling may inhibit autophagy in BCG-infected macrophages by activating mTOR-dependent pathways. Our findings reveal the mechanisms of Wnt/β-catenin signalling regulates cellular autophagy induced by Mtb and provide novel insights into physiological and immune control of tuberculosis by modulating autophagy processes.
Collapse
Affiliation(s)
- Xiaoling Wu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Jiamei Zhang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Chenjie Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Wu Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Jin Zeng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| |
Collapse
|
148
|
Abstract
Sepsis was known to ancient Greeks since the time of great physician Hippocrates (460-377 BC) without exact information regarding its pathogenesis. With time and medical advances, it is now considered as a condition associated with organ dysfunction occurring in the presence of systemic infection as a result of dysregulation of the immune response. Still with this advancement, we are struggling for the development of target-based therapeutic approach for the management of sepsis. The advancement in understanding the immune system and its working has led to novel discoveries in the last 50 years, including different pattern recognition receptors. Inflammasomes are also part of these novel discoveries in the field of immunology which are <20 years old in terms of their first identification. They serve as important cytosolic pattern recognition receptors required for recognizing cytosolic pathogens, and their pathogen-associated molecular patterns play an important role in the pathogenesis of sepsis. The activation of both canonical and non-canonical inflammasome signaling pathways is involved in mounting a proinflammatory immune response via regulating the generation of IL-1β, IL-18, IL-33 cytokines and pyroptosis. In addition to pathogens and their pathogen-associated molecular patterns, death/damage-associated molecular patterns and other proinflammatory molecules involved in the pathogenesis of sepsis affect inflammasomes and vice versa. Thus, the present review is mainly focused on the inflammasomes, their role in the regulation of immune response associated with sepsis, and their targeting as a novel therapeutic approach.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Australia,
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia,
| |
Collapse
|
149
|
Huang JH, Liu CY, Wu SY, Chen WY, Chang TH, Kan HW, Hsieh ST, Ting JPY, Wu-Hsieh BA. NLRX1 Facilitates Histoplasma capsulatum-Induced LC3-Associated Phagocytosis for Cytokine Production in Macrophages. Front Immunol 2018; 9:2761. [PMID: 30559741 PMCID: PMC6286976 DOI: 10.3389/fimmu.2018.02761] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/09/2018] [Indexed: 11/13/2022] Open
Abstract
LC3-associated phagocytosis (LAP) is an emerging non-canonical autophagy process that bridges signaling from pattern-recognition receptors (PRRs) to autophagic machinery. LAP formation results in incorporation of lipidated LC3 into phagosomal membrane (termed LAPosome). Increasing evidence reveals that LAP functions as an innate defense mechanism against fungal pathogens. However, the molecular mechanism involved and the consequence of LAP in regulating anti-fungal immune response remain largely unexplored. Here we show that Histoplasma capsulatum is taken into LAPosome upon phagocytosis by macrophages. Interaction of H. capsulatum with Dectin-1 activates Syk and triggers subsequent NADPH oxidase-mediated reactive oxygen species (ROS) response that is involved in LAP induction. Inhibiting LAP induction by silencing LC3α/β or treatment with ROS inhibitor impairs the activation of MAPKs-AP-1 pathway, thereby reduces macrophage proinflammatory cytokine response to H. capsulatum. Additionally, we unravel the importance of NLRX1 in fungus-induced LAP. NLRX1 facilitates LAP by interacting with TUFM which associates with autophagic proteins ATG5-ATG12 for LAPosome formation. Macrophages from Nlrx1 -/- mice or TUFM-silenced cells exhibit reduced LAP induction and LAP-mediated MAPKs-AP-1 activation for cytokine response to H. capsulatum. Furthermore, inhibiting ROS production in Nlrx1 -/- macrophages almost completely abolishes H. capsulatum-induced LC3 conversion, indicating that both Dectin-1/Syk/ROS-dependent pathway and NLRX1-TUFM complex-dependent pathway collaboratively contribute to LAP induction. Our findings reveal new pathways underlying LAP induction by H. capsulatum for macrophage cytokine response.
Collapse
Affiliation(s)
- Juin-Hua Huang
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chu-Yu Liu
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sheng-Yang Wu
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Yu Chen
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tzu-Hsuan Chang
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hung-Wei Kan
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jenny P-Y Ting
- Departments of Genetics, Microbiology and Immunology, Lineberger Comprehensive Cancer Center, Center for Translational Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Betty A Wu-Hsieh
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
150
|
Caspases orchestrate microglia instrumental functions. Prog Neurobiol 2018; 171:50-71. [DOI: 10.1016/j.pneurobio.2018.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 12/16/2022]
|