1
|
Nadendla EK, Tweedell RE, Kasof G, Kanneganti TD. Caspases: structural and molecular mechanisms and functions in cell death, innate immunity, and disease. Cell Discov 2025; 11:42. [PMID: 40325022 PMCID: PMC12052993 DOI: 10.1038/s41421-025-00791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gary Kasof
- Cell Signaling Technology, Danvers, MA, USA
| | | |
Collapse
|
2
|
Wang Z, Wang Y, Yang S, Wang Z, Yang Q. Brucella suis S2 strain inhibits IRE1/caspase-12/caspase-3 pathway-mediated apoptosis of microglia HMC3 by affecting the ubiquitination of CALR. mSphere 2025; 10:e0094124. [PMID: 40019270 PMCID: PMC11934333 DOI: 10.1128/msphere.00941-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/14/2024] [Indexed: 03/01/2025] Open
Abstract
Neurobrucellosis represents a severe complication of brucellosis, posing a considerable risk to human health and quality of life. This condition arises from an increased susceptibility to chronic Brucella infection, a significant clinical challenge. One key factor contributing to chronic neurobrucellosis is the regulation of microglial apoptosis by Brucella; however, the exact molecular mechanisms remain largely unresolved. In this study, human microglial clone 3 (HMC3) cells were infected with Brucella suis vaccine strain S2 (B. suis S2) at varying multiplicity of infection (MOI) and durations to assess its effects on the IRE1/caspase-12/caspase-3 signaling pathway. Following the suppression of this pathway by B. suis S2, calreticulin (CALR) was identified through ubiquitin-modified proteomics (data accessible via ProteomeXchange, identifier PXD056006). To further investigate, CALR-overexpression and knockdown HMC3 cell lines were infected with B. suis S2 to elucidate the mechanism by which B. suis S2 inhibits apoptosis in HMC3 cells. In conclusion, our findings demonstrate that B. suis S2 suppresses HMC3 cell apoptosis via the IRE1/caspase-12/caspase-3 pathway by modulating CALR ubiquitination. This study provides a theoretical basis for exploring the mechanisms of neurobrucellosis and offers insights into its clinical treatment.IMPORTANCENeurobrucellosis is a severe complication impacting the central nervous system (CNS) due to neurological deficits caused by Brucella, with primary clinical features including meningitis, encephalitis, brain abscesses, and demyelinating lesions. These nonspecific symptoms often lead to misdiagnosis or delayed diagnosis, increasing the risk of recurrent or chronic neurobrucellosis infections. Consequently, persistent infection and relapse are critical challenges in the clinical management of neurobrucellosis, which are closely linked to Brucella's survival and replication within microglia. Interestingly, Brucella may inhibit microglia apoptosis by mitigating endoplasmic reticulum (ER) stress, though the precise molecular mechanisms remain largely unexplored. Thus, this study will elucidate the specific mechanisms by which Brucella suppresses microglial apoptosis and provide deeper insights into the molecular pathogenesis and clinical treatment of neurobrucellosis.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Experimental Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yanbai Wang
- Department of Neurology, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shulong Yang
- Department of Orthopedics, The People’s Hospital of Wuhai, Wuhai, China
| | - Zhenhai Wang
- Department of Neurology, The General Hospital of Ningxia Medical University, Yinchuan, China
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qian Yang
- Department of Experimental Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| |
Collapse
|
3
|
Fu X, Xu W, Yang Y, Li D, Shi W, Li X, Chen N, Lv Q, Shi Y, Xu J, Xu J, Yan Y, Shi F, Li X. Diverse strategies utilized by coronaviruses to evade antiviral responses and suppress pyroptosis. Int J Biol Macromol 2025; 296:139743. [PMID: 39798756 DOI: 10.1016/j.ijbiomac.2025.139743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Viral infections trigger inflammasome-mediated caspase-1 activation. Nevertheless, limited understanding exists regarding how viruses use the active caspase-1 to evade host immune response. Here, we use porcine epidemic diarrhea virus (PEDV) as a model of coronaviruses (CoVs) to illustrate the intricate regulation of CoVs to combat IFN-I signaling and pyroptosis. Our findings demonstrate that PEDV infection stabilizes caspase-1 expression via papain-like protease PLP2's deubiquitinase activity. This stabilization of caspase-1 disrupts IFN-I signaling by cleaving RIG-I at the D189 residue. Furthermore, we demonstrate that 6-thioguanine (6TG), a PLP2 inhibitor, reverses the inhibitory effect on IFN-I signaling mediated by PLP2 and significantly reduces PEDV replication. Additionally, PLP2 degrades GSDMD-p30 by removing its K27-linked ubiquitin chain at K275 to restrain pyroptosis. Papain-like proteases from other genera of CoVs (PDCoV and SARS-CoV-2) have the similar activity to degrade GSDMD-p30. We further demonstrate that SARS-CoV-2 N protein induced NLRP3 inflammasome activation also uses the active caspase-1 to counter IFN-I signaling by cleaving RIG-I. Therefore, our work unravels a novel antagonistic mechanism employed by CoVs to evade host antiviral response.
Collapse
Affiliation(s)
- Xinyu Fu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Weilv Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yang Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Danyue Li
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wen Shi
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xinyue Li
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Nan Chen
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qian Lv
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yuhua Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jinxia Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jidong Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China; Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China; Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Fushan Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China; Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China; Zhejiang University-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Xinchang, China.
| | - Xiaoliang Li
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China; Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China; Hainan Institute of Zhejiang University, Sanya, China; Zhejiang University-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Xinchang, China.
| |
Collapse
|
4
|
Yu H, He L, Wu X, Zhang M, Chen Z, Sun J, Xu X. Porcine epidemic diarrhea coronavirus infection activates caspase-8 to enhance innate immunity by blocking CYLD-mediated deubiquitination of RIG-I. Virology 2025; 604:110443. [PMID: 39908775 DOI: 10.1016/j.virol.2025.110443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
Caspase-8 is best known as an initiator caspase that induces apoptosis. However, recent studies suggest that caspase-8 can modulate innate antiviral immunity in a context-dependent manner. Porcine epidemic diarrhea virus (PEDV), a coronavirus that rapidly replicates in porcine intestinal epithelial cells, triggers apoptosis, in part through caspase-8 activation. In this study, we investigated the role of caspase-8 activation in regulating antiviral responses in IPEC-DQ cells, a porcine intestinal epithelial cell line. We found that inhibition of caspase-8 activity using Z-Vad (a pan-caspase inhibitor) and Z-IETD (a caspase-8-specific inhibitor) reduced PEDV-induced phosphorylation of TBK1, IRF3, and the p65 subunit of NF-κB, and inhibited the nuclear translocation of p65 and IRF3. Similarly, caspase-8 knockout inhibited PEDV-induced phosphorylation of p65, IRF3, and TBK1, as well as RIG-I expression and IRF3- and NF-κB-driven luciferase activity. Notably, inhibition of caspase-3 with Z-DEVD had no effect on PEDV-induced TBK1 and IRF3 phosphorylation. Mechanistically, caspase-8 cleaves and inactivates CYLD, a deubiquitinase that removes K63-linked ubiquitin from RIG-I. Caspase-8 knockout and Z-Vad blocked PEDV-induced RIG-I K63 ubiquitination. While Z-Vad inhibited cleavage of the viral N protein and promoted PEDV replication, neither Z-IETD nor caspase-8 knockout affected N protein cleavage or virus replication. Our results suggest that caspase-8 activation enhances innate antiviral immunity in PEDV-infected intestinal epithelial cells without affecting viral replication, likely due to viral manipulation of IFN signaling.
Collapse
Affiliation(s)
- Huidi Yu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Linshan He
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Xuemei Wu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Miao Zhang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Jing Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China.
| | - Xiulong Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, Jiangsu Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China.
| |
Collapse
|
5
|
Studstill C, Huang N, Sundstrom S, Moscoso S, Zhang H, Damania B, Moody C. Apoptotic Caspases Suppress Expression of Endogenous Retroviruses in HPV31+ Cells That Are Associated with Activation of an Innate Immune Response. Viruses 2024; 16:1695. [PMID: 39599810 PMCID: PMC11598866 DOI: 10.3390/v16111695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Avoidance of an immune response is critical to completion of the human papillomavirus (HPV) life cycle, which occurs in the stratified epithelium and is linked to epithelial differentiation. We previously demonstrated that high-risk HPVs use apoptotic caspases to suppress an antiviral innate immune response during the productive phase of the life cycle. We found that caspase-8 and caspase-3 suppress a type I IFN-β and type III IFN-λ response by disabling the MDA5/MAVS double-stranded RNA (dsRNA) sensing pathway, indicating that immunogenic RNAs increase upon differentiation in HPV+ cells. In this study, we demonstrate that caspase inhibition results in aberrant transcription of a subset of endogenous retroviruses (ERVs) that have been shown to activate an IFN response through dsRNA-sensing pathways. We show that the increase in ERV transcription is accompanied by an enrichment in dsRNA formation. Additionally, we demonstrate that the robust increase in ERV expression requires activation of the JAK/STAT-signaling pathway, indicating that this subset of ERVs is IFN-inducible. Overall, these results suggest a model by which caspase activity blocks the reactivation of ERVs through the JAK/STAT pathway, protecting HPV+ cells from an increase in immunogenic dsRNAs that otherwise would trigger an IFN response that inhibits productive viral replication.
Collapse
Affiliation(s)
- Caleb Studstill
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ning Huang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shelby Sundstrom
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samantha Moscoso
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Huirong Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Blossom Damania
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cary Moody
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Fang Y, Gong Z, You M, Peng K. Identification of a novel caspase cleavage motif AEAD. Virol Sin 2024; 39:755-766. [PMID: 39098717 PMCID: PMC11738786 DOI: 10.1016/j.virs.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/16/2023] [Indexed: 08/06/2024] Open
Abstract
Infections of many viruses induce caspase activation to regulate multiple cellular pathways, including programmed cell death, immune signaling and etc. Characterizations of caspase cleavage sites and substrates are important for understanding the regulation mechanisms of caspase activation. Here, we identified and analyzed a novel caspase cleavage motif AEAD, and confirmed its caspase dependent cleavage activity in natural substrate, such as nitric oxide-associated protein 1 (NOA1). Fusing the enhanced green fluorescent protein (EGFP) with the mitochondrial marker protein Tom20 through the AEAD motif peptide localized EGFP to the mitochondria. Upon the activation of caspase triggered by Sendai virus (SeV) or herpes simplex virus type 1 (HSV-1) infection, EGFP diffusely localized to the cell due to the caspase-mediated cleavage, thus allowing visual detection of the virus-induced caspase activation. An AEAD peptide-derived inhibitor Z-AEAD-FMK were developed, which significantly inhibited the activities of caspases-1, -3, -6, -7, -8 and -9, exhibiting a broad caspase inhibition effect. The inhibitor further prevented caspases-mediated cleavage of downstream substrates, including BID, PARP1, LMNA, pro-IL-1β, pro-IL-18, GSDMD and GSDME, protecting cells from virus-induced apoptotic and pyroptotic cell death. Together, our findings provide a new perspective for the identification of novel caspase cleavage motifs and the development of new caspase inhibitors and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Yujie Fang
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhou Gong
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences, Wuhan, 430071, China
| | - Miaomiao You
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Provincial Key Laboratory of Jiangxia, Wuhan, 430207, China.
| |
Collapse
|
7
|
Yu H, Sun Y, Zhang J, Zhang W, Liu W, Liu P, Liu K, Sun J, Liang H, Zhang P, Wang X, Liu X, Xu X. Influenza A virus infection activates caspase-8 to enhance innate antiviral immunity by cleaving CYLD and blocking TAK1 and RIG-I deubiquitination. Cell Mol Life Sci 2024; 81:355. [PMID: 39158695 PMCID: PMC11335283 DOI: 10.1007/s00018-024-05392-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/10/2024] [Accepted: 07/27/2024] [Indexed: 08/20/2024]
Abstract
Caspase-8, an aspartate-specific cysteine protease that primarily functions as an initiator caspase to induce apoptosis, can downregulate innate immunity in part by cleaving RIPK1 and IRF3. However, patients with caspase-8 mutations or deficiency develop immunodeficiency and are prone to viral infections. The molecular mechanism underlying this controversy remains unknown. Whether caspase-8 enhances or suppresses antiviral responses against influenza A virus (IAV) infection remains to be determined. Here, we report that caspase-8 is readily activated in A549 and NL20 cells infected with the H5N1, H5N6, and H1N1 subtypes of IAV. Surprisingly, caspase-8 deficiency and two caspase-8 inhibitors, Z-VAD and Z-IETD, do not enhance but rather downregulate antiviral innate immunity, as evidenced by decreased TBK1, IRF3, IκBα, and p65 phosphorylation, decreased IL-6, IFN-β, MX1, and ISG15 gene expression; and decreased IFN-β production but increased virus replication. Mechanistically, caspase-8 cleaves and inactivates CYLD, a tumor suppressor that functions as a deubiquitinase. Caspase-8 inhibition suppresses CYLD cleavage, RIG-I and TAK1 ubiquitination, and innate immune signaling. In contrast, CYLD deficiency enhances IAV-induced RIG-I and TAK1 ubiquitination and innate antiviral immunity. Neither caspase-3 deficiency nor treatment with its inhibitor Z-DEVD affects CYLD cleavage or antiviral innate immunity. Our study provides evidence that caspase-8 activation in two human airway epithelial cell lines does not silence but rather enhances innate immunity by inactivating CYLD.
Collapse
Affiliation(s)
- Huidi Yu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Yuling Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Jingting Zhang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Wenhui Zhang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Wei Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Penggang Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Kaituo Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, Jiangsu Province, China
| | - Jing Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Hailiang Liang
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Pinghu Zhang
- College of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xiulong Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, Jiangsu Province, China.
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China.
| |
Collapse
|
8
|
Sun Y, Ji L, Liu W, Sun J, Liu P, Wang X, Liu X, Xu X. Influenza virus infection activates TAK1 to suppress RIPK3-independent apoptosis and RIPK1-dependent necroptosis. Cell Commun Signal 2024; 22:372. [PMID: 39044278 PMCID: PMC11264382 DOI: 10.1186/s12964-024-01727-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Many DNA viruses develop various strategies to inhibit cell death to facilitate their replication. However, whether influenza A virus (IAV), a fast-replicating RNA virus, attenuates cell death remains unknown. Here, we report that IAV infection induces TAK1 phosphorylation in a murine alveolar epithelial cell line (LET1) and a murine fibroblastoma cell line (L929). The TAK1-specific inhibitor 5Z-7-Oxzeneonal (5Z) and TAK1 knockout significantly enhance IAV-induced apoptosis, as evidenced by increased PARP, caspase-8, and caspase-3 cleavage. TAK1 inhibition also increases necroptosis as evidenced by increased RIPK1S166, RIPK3T231/S232, and MLKLS345 phosphorylation. Mechanistically, TAK1 activates IKK, which phosphorylates RIPK1S25 and inhibits its activation. TAK1 also activates p38 and its downstream kinase MK2, which phosphorylates RIPK1S321 but does not affect RIPK1 activation. Further investigation revealed that the RIPK1 inhibitor Nec-1 and RIPK1 knockout abrogate IAV-induced apoptosis and necroptosis; re-expression of wild-type but not kinase-dead (KD)-RIPK1 restores IAV-induced cell death. ZBP1 knockout abrogates IAV-induced cell death, whereas RIPK3 knockout inhibits IAV-induced necroptosis but not apoptosis. 5Z treatment enhances IAV-induced cell death and slightly reduces the inflammatory response in the lungs of H1N1 virus-infected mice and prolongs the survival of IAV-infected mice. Our study provides evidence that IAV activates TAK1 to suppress RIPK1-dependent apoptosis and necroptosis, and that RIPK3 is required for IAV-induced necroptosis but not apoptosis in epithelial cells.
Collapse
Affiliation(s)
- Yuling Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Lei Ji
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Wei Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Jing Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Penggang Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Xiulong Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
| |
Collapse
|
9
|
Xiong F, Wang C, Lu J, Bai G, Zhou D, Ling J. 4-PBA exerts brain-protective effects against sepsis-associated encephalopathy in a mouse model of sepsis. Exp Neurol 2024; 375:114738. [PMID: 38395217 DOI: 10.1016/j.expneurol.2024.114738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Neuroinflammation assumes a pivotal role in both the etiological underpinnings and the dynamic progression of sepsis-associated encephalopathy (SAE). The occurrence of cognitive deficits with SAE is associated with neuroinflammation. 4-phenyl butyrate (4-PBA) may control inflammation by inhibiting endoplasmic reticulum stress (ERS). The primary objective of this investigation is to scrutinize the effectiveness of 4-PBA in mitigating neuroinflammation induced by lipopolysaccharides (LPS) and its consequent impact on cognitive function decline. METHODS LPS-injected mice with SAE and LPS-treated BV2 cell were established to serve as experimental paradigms, both contributing to the investigative framework of the study. Cognitive functions were assessed by behavioral tests. Hippocampal neuronal damage was assessed using Golgi staining and Nissl staining. Quantitative PCR assay and immunofluorescence were used to analyze neuroinflammation. Mitochondrial function was examined using transmission electron microscopy. Protein expression analysis was conducted through the application of western blotting methodology, serving as the investigative approach to elucidate molecular signatures in the experimental framework. Endoplasmic reticulum and mitochondrial calcium flow were detected using flow cytometry. To delve deeper into the mechanistic intricacies, the administration of 4μ8c was employed to selectively impede the IRE1α/Xbp1s pathway, constituting a strategic intervention aimed at elucidating underlying regulatory processes. RESULT Expression levels of ERS-related proteins exhibited a significant upregulation in hippocampal tissues of LPS-treated mice when compared to wild-type (WT) counterparts. The administration of 4-PBA notably ameliorated memory deficits in LPS-treated mice. Furthermore, 4-PBA treatment was found to alleviate oxidative stress and neuroinflammation. Mechanistically, the IRE1α/Xbp1s-Ca2+ signaling pathway played a crucial role in mediating the beneficial effects of mitigating oxidative stress and maintaining mitochondrial calcium homeostasis, with inhibition of the IRE-related pathway displaying opposing effects. CONCLUSION Our results suggest that administration of 4-PBA treatment significantly attenuates ERS, alleviates cognitive decline, reduces inflammatory damage, and restores mitochondrial dynamics via the IRE1α/Xbp1s-Ca2+-associated pathway, which provides a new potential therapeutic approach to SAE.
Collapse
Affiliation(s)
- Feng Xiong
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China
| | - Cailin Wang
- Department of Neurology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jun Lu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China
| | - Guangyang Bai
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China.
| | - Jianmin Ling
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong university of science and technology, Wuhan 430000, China.
| |
Collapse
|
10
|
González R, Félix MA. Caenorhabditis elegans immune responses to microsporidia and viruses. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 154:105148. [PMID: 38325500 DOI: 10.1016/j.dci.2024.105148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
The model organism Caenorhabditis elegans is susceptible to infection by obligate intracellular pathogens, specifically microsporidia and viruses. These intracellular pathogens infect intestinal cells, or, for some microsporidia, epidermal cells. Strikingly, intestinal cell infections by viruses or microsporidia trigger a common transcriptional response, activated in part by the ZIP-1 transcription factor. Among the strongest activated genes in this response are ubiquitin-pathway members and members of the pals family, an intriguing gene family with cross-regulations of different members of genomic clusters. Some of the induced genes participate in host defense against the pathogens, for example through ubiquitin-mediated inhibition. Other mechanisms defend the host specifically against viral infections, including antiviral RNA interference and uridylation. These various immune responses are altered by environmental factors and by intraspecific genetic variation of the host. These pathogens were first isolated 15 years ago and much remains to be discovered using C. elegans genetics; also, other intracellular pathogens of C. elegans may yet to be discovered.
Collapse
Affiliation(s)
- Rubén González
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France.
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France
| |
Collapse
|
11
|
Zhang R, Tao X, Sun R, Dai T, Xi X, Sun W, Song L, Gong W. Cognitive-exercise dual-task promotes cognitive function recovery in chronic cerebral ischemia male rats through regulating PI3K/Akt signaling pathway via inhibition of EphrinA3/EphA4. J Neurosci Res 2024; 102. [PMID: 38284844 DOI: 10.1002/jnr.25275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 01/30/2024]
Abstract
Chronic cerebral ischemia (CCI) can lead to vascular cognitive impairment, but therapeutic options are limited. Cognitive-exercise dual-task (CEDT), as a potential rehabilitation intervention, can attenuate cognitive impairment. However, the related mechanisms remain unclear. In this study, 2-vessel occlusion (2-VO) in male SD rats was performed to establish the CCI model. The rats were treated with cognitive, exercise, or CEDT intervention for 21 days. The Morris water maze (MWM) test was used to assess cognitive ability. TUNEL staining was used to detect the neuronal apoptosis. Immunofluorescence, RT-qPCR and Western blot were used to detect the protein or mRNA levels of EphrinA3, EphA4, p-PI3K, and p-Akt. The results showed that CEDT could improve performance in the MWM test, reverse the increased expression of EphrinA3 and EphA4, and the reduced expression of p-PI3K and p-Akt in CCI rats, which was superior to exercise and cognitive interventions. In vitro, oxygenglucose deprivation (OGD) challenge of astrocytes and neuronal cells were used to mimic cerebral ischemia. Immunofluorescence assay revealed that the levels of MAP-2, p-PI3K, and p-Akt were reduced in EphrinA3 overexpressed cells after OGD stimulation. Finally, the knock-down of EphrinA3 by shRNA significantly promoted the recovery of cognitive function and activation of PI3K/Akt after CEDT treatment in CCI rats. In conclusion, our study suggests that CEDT promotes cognitive function recovery after CCI by regulating the signaling axis of EphrinA3/EphA4/PI3K/Akt.
Collapse
Affiliation(s)
- Rong Zhang
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
- The Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Xue Tao
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Ruifeng Sun
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Tengteng Dai
- The Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - XiaoShuang Xi
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Weishuang Sun
- Rehabilitation Medicine Academy, Weifang Medical University, Weifang, China
| | - Li Song
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Zhang X, Sun C, Hao J, Cao L, Zhang X, Du J, Han Q. Metformin inhibits EV71‑induced pyroptosis by upregulating DEP domain‑containing mTOR‑interacting protein. Exp Ther Med 2023; 26:388. [PMID: 37456175 PMCID: PMC10347180 DOI: 10.3892/etm.2023.12087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/02/2023] [Indexed: 07/18/2023] Open
Abstract
Enterovirus 71 (EV71) infection is one of the main causes of severe hand, foot and mouth disease (HFMD), which is usually accompanied by a marked inflammatory response. The excessive inflammatory response has been implicated to serve an important role in EV71-caused HFMD. Pyroptosis is a type of inflammatory programmed cell death. Therefore, a novel treatment strategy against EV71 infection could aim to alleviate the inflammatory response through inhibition of EV71-induced pyroptosis. The present study revealed that metformin had this therapeutic potential. A cell model of EV71 infection was established, cell viability was measured by CCK8 assay, cell damage was measured by LDH release kit, and the dead and dying cells were excluded by propidium iodide staining. The intracellular levels of DEP domain-containing mTOR interacting protein (DEPTOR) and pyroptosis-associated molecules were measured by western blot analysis, the NLRP3 expression was assessed by immunofluorescence labeling, and virus titers in cell culture supernatants were determined by a cell culture infectious dose 50 assay. The results demonstrated that EV71 infection could induce pyroptosis in a time- and dose-dependent manner, and metformin could inhibit EV71-induced pyroptosis. The mechanism of metformin inhibiting EV71-induced pyroptosis was explored next. Subsequent experiments indicated that metformin could increase the levels of DEPTOR, which were decreased by EV71. Finally, overexpression of DEPTOR in cells could reduce EV71-induced pyroptosis. Overall, the present study demonstrated that metformin could exert a novel pharmacodynamic anti-pyroptosis effect in the treatment of EV71 infection by upregulating DEPTOR expression.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
| | - Chenxi Sun
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jinfang Hao
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Li Cao
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xinyan Zhang
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jianping Du
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Qian Han
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi 032200, P.R. China
- Graduate School, Fenyang Hospital Provincial and Municipal Joint Construction Key Laboratory, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
13
|
Kim CU, Lim D, Kim YS, Ku B, Kim DJ. Influenza viral matrix 1 protein aggravates viral pathogenicity by inducing TLR4-mediated reactive oxygen species production and apoptotic cell death. Cell Death Dis 2023; 14:228. [PMID: 36990977 PMCID: PMC10060384 DOI: 10.1038/s41419-023-05749-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023]
Abstract
Influenza virus is one of the most challenging viruses threating human health. Since infection with influenza virus triggers inflammatory responses and induces cell death, the molecular and cellular mechanisms by which the virus-infected cells undergo apoptotic and necrotic cell death have been widely studied. However, most of the studies have focused on the molecular events occurring in the cytosol and there is limited information on the physiological correlation between virus-induced cell death and the viral pathogenesis in vivo. In this study, we demonstrate that the influenza virus matrix 1 (M1) protein is released from virus-infected cells and triggers apoptotic cell death of lung epithelial and pulmonary immune cells, through the activation of Toll-like receptor 4 (TLR4) signaling. Treatment with M1 protein led to robust cellular inflammatory responses, such as the production of proinflammatory cytokines and cellular reactive oxygen species (ROS), and induction of cell death. When M1 protein was administered in vivo, it induced the activation of inflammatory responses and cell death in the lungs. Furthermore, the administration of M1 aggravated lung pathology and mortality of the virus-infected mice in a TLR4-dependent manner. These results demonstrate that M1 is an important pathogenic factor contributing to influenza virus pathogenicity by enhancing cell death in the lungs, thereby expanding our understanding of the molecular mechanism of influenza virus-induced cell death through the interaction with an innate immune receptor.
Collapse
Affiliation(s)
- Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Dahwan Lim
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young Sang Kim
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.
- University of Science and Technology (UST), Daejeon, South Korea.
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea.
- University of Science and Technology (UST), Daejeon, South Korea.
| |
Collapse
|
14
|
Unnisa A, Greig NH, Kamal MA. Inhibition of Caspase 3 and Caspase 9 Mediated Apoptosis: A Multimodal Therapeutic Target in Traumatic Brain Injury. Curr Neuropharmacol 2023; 21:1001-1012. [PMID: 35339178 PMCID: PMC10227914 DOI: 10.2174/1570159x20666220327222921] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/17/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the significant causes of death and morbidity, and it is hence a focus of translational research. Apoptosis plays an essential part in the pathophysiology of TBI, and its inhibition may help overcome TBI's negative consequences and improve functional recovery. Although physiological neuronal death is necessary for appropriate embryologic development and adult cell turnover, it can also drive neurodegeneration. Caspases are principal mediators of cell death due to apoptosis and are critical for the required cleavage of intracellular proteins of cells committed to die. Caspase-3 is the major executioner Caspase of apoptosis and is regulated by a range of cellular components during physiological and pathological conditions. Activation of Caspase-3 causes proteolyzation of DNA repair proteins, cytoskeletal proteins, and the inhibitor of Caspase-activated DNase (ICAD) during programmed cell death, resulting in morphological alterations and DNA damage that define apoptosis. Caspase-9 is an additional crucial part of the intrinsic pathway, activated in response to several stimuli. Caspases can be altered post-translationally or by modulatory elements interacting with the zymogenic or active form of a Caspase, preventing their activation. The necessity of Caspase-9 and -3 in diverse apoptotic situations suggests that mammalian cells have at least four distinct apoptotic pathways. Continued investigation of these processes is anticipated to disclose new Caspase regulatory mechanisms with consequences far beyond apoptotic cell death control. The present review discusses various Caspase-dependent apoptotic pathways and the treatment strategies to inhibit the Caspases potentially.
Collapse
Affiliation(s)
- Aziz Unnisa
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail, KSA;
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, NSW, Australia
| |
Collapse
|
15
|
Bagayoko S, Meunier E. Emerging roles of ferroptosis in infectious diseases. FEBS J 2022; 289:7869-7890. [PMID: 34670020 DOI: 10.1111/febs.16244] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/06/2021] [Accepted: 10/20/2021] [Indexed: 01/14/2023]
Abstract
In living organisms, lipid peroxidation is a continuously occurring cellular process and therefore involved in various physiological and pathological contexts. Among the broad variety of lipids, polyunsaturated fatty acids (PUFA) constitute a major target of oxygenation either when released as mediators by phospholipases or when present in membranous phospholipids. The last decade has seen the characterization of an iron- and lipid peroxidation-dependent cell necrosis, namely, ferroptosis, that involves the accumulation of peroxidized PUFA-containing phospholipids. Further studies could link ferroptosis in a very large body of (physio)-pathological processes, including cancer, neurodegenerative, and metabolic diseases. In this review, we mostly focus on the emerging involvement of lipid peroxidation-driven ferroptosis in infectious diseases, and the immune consequences. We also discuss the putative ability of microbial virulence factors to exploit or to dampen ferroptosis regulatory pathways to their own benefit.
Collapse
Affiliation(s)
- Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| |
Collapse
|
16
|
Saxena V, Arregui S, Kamocka MM, Hains DS, Schwaderer A. MAP3K7 is an innate immune regulatory gene with increased expression in human and murine kidney intercalated cells following uropathogenic Escherichia coli exposure. J Cell Biochem 2022; 123:1817-1826. [PMID: 35959632 PMCID: PMC9671826 DOI: 10.1002/jcb.30318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/28/2022] [Accepted: 08/01/2022] [Indexed: 01/09/2023]
Abstract
Understanding the mechanisms responsible for the kidney's defense against ascending uropathogen is critical to devise novel treatment strategies against increasingly antibiotic resistant uropathogen. Growing body of evidence indicate Intercalated cells of the kidney as the key innate immune epithelial cells against uropathogen. The aim of this study was to find orthologous and differentially expressed bacterial defense genes in human versus murine intercalated cells. We simultaneously analyzed 84 antibacterial genes in intercalated cells enriched from mouse and human kidney samples. Intercalated cell "reporter mice" were exposed to saline versus uropathogenic Escherichia coli (UPEC) transurethrally for 1 h in vivo, and intercalated cells were flow sorted. Human kidney intercalated cells were enriched from kidney biopsy using magnetic-activated cell sorting and exposed to UPEC in vitro for 1 h. RT2 antibacterial PCR array was performed. Mitogen-activated protein kinase kinase kinase 7 (MAP3K7) messenger RNA (mRNA) expression increased in intercalated cells of both humans and mice following UPEC exposure. Intercalated cell MAP3K7 protein expression was defined by immunofluorescence and confocal imaging analysis, was consistent with the increased MAP3K7 mRNA expression profiles defined by PCR. The presence of the orthologous innate immune gene MAP3K7/TAK1 suggests that it may be a key regulator of the intercalated cell antibacterial response and demands further investigation of its role in urinary tract infection pathogenesis.
Collapse
Affiliation(s)
- Vijay Saxena
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Samuel Arregui
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Malgorzata Maria Kamocka
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - David S. Hains
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA,Department of Pediatrics, Division of NephrologyRiley Hospital for ChildrenIndianapolisIndianaUSA
| | - Andrew Schwaderer
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA,Department of Pediatrics, Division of NephrologyRiley Hospital for ChildrenIndianapolisIndianaUSA
| |
Collapse
|
17
|
Wang Y, Karki R, Mall R, Sharma BR, Kalathur RC, Lee S, Kancharana B, So M, Combs KL, Kanneganti TD. Molecular mechanism of RIPK1 and caspase-8 in homeostatic type I interferon production and regulation. Cell Rep 2022; 41:111434. [PMID: 36198273 PMCID: PMC9630927 DOI: 10.1016/j.celrep.2022.111434] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/22/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
Type I interferons (IFNs) are essential innate immune proteins that maintain tissue homeostasis through tonic expression and can be upregulated to drive antiviral resistance and inflammation upon stimulation. However, the mechanisms that inhibit aberrant IFN upregulation in homeostasis and the impacts of tonic IFN production on health and disease remain enigmatic. Here, we report that caspase-8 negatively regulates type I IFN production by inhibiting the RIPK1-TBK1 axis during homeostasis across multiple cell types and tissues. When caspase-8 is deleted or inhibited, RIPK1 interacts with TBK1 to drive elevated IFN production, leading to heightened resistance to norovirus infection in macrophages but also early onset lymphadenopathy in mice. Combined deletion of caspase-8 and RIPK1 reduces the type I IFN signaling and lymphadenopathy, highlighting the critical role of RIPK1 in this process. Overall, our study identifies a mechanism to constrain tonic type I IFN during homeostasis which could be targeted for infectious and inflammatory diseases. Wang et al. report the mechanistic regulation of homeostatic type I IFN production by caspase-8 through the RIPK1-TBK1 axis. Hyper-activation of this pathway due to loss of caspase-8 has profound physiological impacts on natural resistance to viral infection and the pathogenesis of lymphadenopathy.
Collapse
|
18
|
Moody CA. Regulation of the Innate Immune Response during the Human Papillomavirus Life Cycle. Viruses 2022; 14:v14081797. [PMID: 36016419 PMCID: PMC9412305 DOI: 10.3390/v14081797] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022] Open
Abstract
High-risk human papillomaviruses (HR HPVs) are associated with multiple human cancers and comprise 5% of the human cancer burden. Although most infections are transient, persistent infections are a major risk factor for cancer development. The life cycle of HPV is intimately linked to epithelial differentiation. HPVs establish infection at a low copy number in the proliferating basal keratinocytes of the stratified epithelium. In contrast, the productive phase of the viral life cycle is activated upon epithelial differentiation, resulting in viral genome amplification, high levels of late gene expression, and the assembly of virions that are shed from the epithelial surface. Avoiding activation of an innate immune response during the course of infection plays a key role in promoting viral persistence as well as completion of the viral life cycle in differentiating epithelial cells. This review highlights the recent advances in our understanding of how HPVs manipulate the host cell environment, often in a type-specific manner, to suppress activation of an innate immune response to establish conditions supportive of viral replication.
Collapse
Affiliation(s)
- Cary A. Moody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
An J, Cao X, Feng M, Hu B, Wu J, Gang Y, Jian S, Wen C. MAPK/MAK/MRK overlapping kinase mediated apoptosis through caspase signaling pathway from Cristaria plicata. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104427. [PMID: 35460761 DOI: 10.1016/j.dci.2022.104427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/17/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
MAPK/MAK/MRK Overlapping Kinase (MOK) belongs to MAP kinase superfamily, which plays an important role in regulating cell growth, division, and differentiation. Caspase-3, as the final executor of apoptosis, has an important position in the caspase-mediated apoptotic signaling pathway. The full-length cDNA of MOK and caspase-3 were cloned from Cristaria plicata (designated CpMOK and CpCaspase-3). The CpMOK gene was sequence with a full-length of 1413 bp, encoding a total of 470 amino acids, and containing an S_TKc structural domain. CpCaspase-3 has a sequence of 2425 bp, encoding 322 amino acids, containing a CASc domain. Real-time fluorescence quantitative PCR analysis showed that CpMOK and CpCaspase-3 distributed in various tissues of C. plicata, and the highest expression of CpMOK and CpCaspase-3 mRNA was in hepatopancreas. The expression of CpMOK was significantly changed in hepatopancreas, gills, and kidneys by the construction of wound model as well as stimulation of LPS, PGN, Poly I: C and Aeromonas hydrophila. Subcellular localization experiments confirmed that CpMOK was localized in the nucleus. Furthermore, the double-stranded RNA (dsRNA) of CpMOK was constructed for interference experiment, and the results showed that the mRNA expression of apoptotic gene signals caspase-1, caspase-3, caspase-7, caspase-8, and caspase-9 were increased. The expression of caspase-1, -3, -7, -9, cytochrome C (Cyt-c) and tumor necrosis factor-α (TNF-α) was detected by ELISA. Fluorescent staining of apoptotic cells using the Tunnel method revealed an increase in the number of apoptotic cells after interference. These results suggested that CpMOK knockdown could induce caspase-mediated apoptosis in C. plicata, and the phosphorylation of the kinase was disrupted during the process.
Collapse
Affiliation(s)
- Jinhua An
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Xinying Cao
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Maolin Feng
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Baoqing Hu
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Jielian Wu
- Jiangxi Science and Technology Normal University, Nanchang, Jiangxi Province, 11318, China
| | - Yang Gang
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Shaoqing Jian
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Chungen Wen
- Department of Zoology, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China.
| |
Collapse
|
20
|
Apoptotic caspases suppress an MDA5-driven IFN response during productive replication of human papillomavirus type 31. Proc Natl Acad Sci U S A 2022; 119:e2200206119. [PMID: 35858339 PMCID: PMC9303994 DOI: 10.1073/pnas.2200206119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human papillomaviruses (HPVs) infect the basal proliferating cells of the stratified epithelium, but the productive phase of the life cycle (consisting of viral genome amplification, late gene expression, and virion assembly) is restricted to the highly differentiated suprabasal cells. While much is known regarding the mechanisms that HPVs use to block activation of an innate immune response in undifferentiated cells, little is known concerning how HPV prevents an interferon (IFN) response upon differentiation. Here, we demonstrate that high-risk HPVs hijack a natural function of apoptotic caspases to suppress an IFN response in differentiating epithelial cells. We show that caspase inhibition results in the secretion of type I and type III IFNs that can act in a paracrine manner to induce expression of interferon-stimulated genes (ISGs) and block productive replication of HPV31. Importantly, we demonstrate that the expression of IFNs is triggered by the melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral-signaling protein (MAVS)-TBK1 (TANK-binding kinase 1) pathway, signifying a response to double-stranded RNA (dsRNA). Additionally, we identify a role for MDA5 and MAVS in restricting productive viral replication during the normal HPV life cycle. This study identifies a mechanism by which HPV reprograms the cellular environment of differentiating cells through caspase activation, co-opting a nondeath function of proteins normally involved in apoptosis to block antiviral signaling and promote viral replication.
Collapse
|
21
|
Hepatoprotective Effect of Mitochondria-Targeted Antioxidant Mito-TEMPO against Lipopolysaccharide-Induced Liver Injury in Mouse. Mediators Inflamm 2022; 2022:6394199. [PMID: 35769207 PMCID: PMC9236847 DOI: 10.1155/2022/6394199] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/26/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
The liver is vulnerable to sepsis, and sepsis-induced liver injury is closely associated with poor survival of sepsis patients. Studies have found that the overproduction of reactive oxygen species (ROS) is the major cause of oxidative stress, which is the main pathogenic factor for the progression of septic liver injury. The mitochondria are a major source of ROS. Mito-TEMPO is a mitochondria-specific superoxide scavenger. The aim of this study was to investigate the effect of Mito-TEMPO on lipopolysaccharide- (LPS-) induced sepsis mice. We found that Mito-TEMPO pretreatment inhibited inflammation, attenuated LPS-induced liver injury, and enhanced the antioxidative capability in septic mice, as evidenced by the decreased MDA content and the increased SOD activity. In addition, Mito-TEMPO restored mitochondrial size and improved mitochondrial function. Finally, we found that the levels of pyroptosis-related proteins in the liver of LPS-treated mice were lower after pretreatment with Mito-TEMPO. The mechanisms could be related to Mito-TEMPO enhanced antioxidative capability and improved mitochondrial function, which reflects the ability to neutralize ROS.
Collapse
|
22
|
Wang C, Zhou W, Liu Y, Xu Y, Zhang X, Jiang C, Jiang M, Cao X. Nuclear translocation of RIG-I promotes cellular apoptosis. J Autoimmun 2022; 130:102840. [DOI: 10.1016/j.jaut.2022.102840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
|
23
|
Yue M, Xiao L, Yan R, Li X, Yang W. Pyroptosis in neurodegenerative diseases: What lies beneath the tip of the iceberg? Int Rev Immunol 2022:1-16. [PMID: 35312447 DOI: 10.1080/08830185.2022.2052064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Neurodegenerative diseases gradually receive attention with a rapidly aging global population. The hallmark of them is a progressive neuronal loss in the brain or peripheral nervous system due to complex reasons ranging from protein aggregation, immune dysregulation to abnormal cell death. The death style of nerve cell is no longer restricted to apoptosis, autophagy and necrosis as confirmed before. With the successive discoveries of the gasdermin (GSDM) protein family and key caspase molecules in the past several decades, pyroptosis emerges as a novel kind of programmed cell death. A substantial body of evidence has recognized the close connection between pyroptosis and the occurrence and development of neurodegenerative diseases. In this review, we summarize molecular mechanisms of pyroptosis, evidences for pyroptosis involvement in neurodegenerative diseases and finally we hope to provide a novel angle for clinical decision-making.
Collapse
Affiliation(s)
- Mengli Yue
- Department of Immunology, College of Basic Medical Sciences, JiLin University, Changchun City, Jilin Province, China
| | - Li Xiao
- Department of Immunology, College of Basic Medical Sciences, JiLin University, Changchun City, Jilin Province, China
| | - Rui Yan
- Department of Immunology, College of Basic Medical Sciences, JiLin University, Changchun City, Jilin Province, China
| | - Xinyi Li
- Department of Immunology, College of Basic Medical Sciences, JiLin University, Changchun City, Jilin Province, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, JiLin University, Changchun City, Jilin Province, China
| |
Collapse
|
24
|
Xiong Y, Tang YD, Zheng C. The crosstalk between the caspase family and the cGAS‒STING signaling pathway. J Mol Cell Biol 2021; 13:739-747. [PMID: 34718659 PMCID: PMC8718194 DOI: 10.1093/jmcb/mjab071] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
Edited by Jiarui Wu Cytosolic nucleic acid sensors are critical for sensing nucleic acids and initiating innate immunity during microbial infections and/or cell death. Over the last decade, several key studies have characterized the conserved mechanism of cyclic guanosine monophosphate‒adenosine monophosphate synthase (cGAS) and the downstream signaling adaptor stimulator of interferon genes (STING) initiating the innate immune signaling pathways. Aside from its primary involvement in microbial infections and inflammatory diseases, there is growing interest in the alternate roles of cGAS‒STING-mediated signaling. Caspase family members are powerful functional proteins that respond to cellular stress, including cell death signals, inflammation, and innate immunity. Recent studies have uncovered how the caspase family cooperates with the cGAS‒STING signaling pathway. Most caspase family members negatively regulate the cGAS‒STING signaling pathway. In turn, some caspase family members can also be modulated by cGAS‒STING. This review gives a detailed account of the interplay between the caspase family and the cGAS‒STING signaling pathway, which will shed light on developing novel therapeutics targeting the caspase family and cGAS‒STING signaling in antiviral innate immunity, cancer, inflammatory, and autoimmunity.
Collapse
Affiliation(s)
- Yongai Xiong
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yan-Dong Tang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Kong L, Zhang H, Lu C, Shi K, Huang H, Zheng Y, Wang Y, Wang D, Wang H, Huang W. AICAR, an AMP-Activated Protein Kinase Activator, Ameliorates Acute Pancreatitis-Associated Liver Injury Partially Through Nrf2-Mediated Antioxidant Effects and Inhibition of NLRP3 Inflammasome Activation. Front Pharmacol 2021; 12:724514. [PMID: 34531748 PMCID: PMC8438129 DOI: 10.3389/fphar.2021.724514] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Acute pancreatitis (AP) is a highly fatal acute inflammation and is often accompanied by multiple organ dysfunction syndrome (MODS). The liver, one of the most vulnerable extrapancreatic organs in AP, is the major organ involved in the evolution of the disease and correlates strongly with the occurrence of MODS. However, the etiology of pancreatitis-associated liver injury (PALI) has not been clarified and currently lacks an effective treatment. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) is a cell permeable nucleoside with pleiotropic effects on anti-inflammatory and antioxidant stress that binds with adenosine monophosphate protein kinase (AMPK) and induces AMPK activation. However, the role of AICAR in PALI remains elusive. Here, we show that activation of AMPK by AICAR, a direct AMPK agonist, significantly ameliorates sodium taurocholate-induced PALI in rats, whereas treatment of PALI rats with the AMPK antagonist Compound C profoundly exacerbates the degree of liver injury, suggesting that hepatic AMPK activation exerts an essential protective role in PALI. Mechanistically, AICAR induces AMPK activation, which in turn activates nuclear factor erythroid 2-related factor 2(Nrf2) -regulated hepatic antioxidant capacity and inhibits NLRP3 inflammasome-mediated pyrolysis, protecting rats from sodium taurocholate-induced PALI. In addition, Nrf2 deficiency strikingly weakens the beneficial effects of AICAR on alleviation of liver injury, oxidative stress and NLRP3 inflammasome activation in L-arginine-induced PALI mice. Thus, AICAR protects against PALI in rodents by triggering AMPK, which is mediated at least in part by Nrf2-modulated antioxidant effects and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Lijun Kong
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hewei Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaosheng Lu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Keqing Shi
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongjian Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yushu Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongqiang Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongwei Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Huang
- Department of Nutrition, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
Dejani NN, Elshabrawy HA, Bezerra Filho CDSM, de Sousa DP. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021; 11:biom11081254. [PMID: 34439920 PMCID: PMC8394099 DOI: 10.3390/biom11081254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
In 2019, COVID-19 emerged as a severe respiratory disease that is caused by the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The disease has been associated with high mortality rate, especially in patients with comorbidities such as diabetes, cardiovascular and kidney diseases. This could be attributed to dysregulated immune responses and severe systemic inflammation in COVID-19 patients. The use of effective antiviral drugs against SARS-CoV-2 and modulation of the immune responses could be a potential therapeutic strategy for COVID-19. Studies have shown that natural phenolic compounds have several pharmacological properties, including anticoronavirus and immunomodulatory activities. Therefore, this review discusses the dual action of these natural products from the perspective of applicability at COVID-19.
Collapse
Affiliation(s)
- Naiara Naiana Dejani
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | - Carlos da Silva Maia Bezerra Filho
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
- Correspondence: ; Tel.: +55-83-3216-7347
| |
Collapse
|
27
|
Fang Y, Peng K. Regulation of innate immune responses by cell death-associated caspases during virus infection. FEBS J 2021; 289:4098-4111. [PMID: 34089572 DOI: 10.1111/febs.16051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/04/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023]
Abstract
Viruses are obligate intracellular pathogens that rely on cellular machinery for successful replication and dissemination. The host cells encode a number of different strategies to sense and restrict the invading viral pathogens. Caspase-mediated programmed cell death pathways that are triggered by virus infection, such as apoptosis and pyroptosis, provide a means for the infected cells to limit viral proliferation, leading to suicidal cell death (apoptosis) or lytic cell death and alerting uninfected cells to mount anti-viral responses (pyroptosis). However, some viruses can employ activated caspases to dampen the anti-viral responses and facilitate viral replication through cleavage of critical molecules of the innate immune pathways. The regulation of innate immune responses by caspase activation during virus infection has recently become an important topic. In this review, we briefly introduce the characteristics of different classes of caspases and the cell death pathways regulated by these caspases. We then describe how viruses trigger or dampen caspase activation during infection and how these activated caspases regulate three major innate immune response pathways of viral infections: the retinoic acid-inducible gene I-like receptor, toll-like receptor and cyclic GMP-AMP synthase-stimulator of interferon genes pathways.
Collapse
Affiliation(s)
- Yujie Fang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Wang B, Zhang C, Yu C, Zhu Y, Tang Q, Huang H, Zhao Z. Enterovirus 71 Induces INF2 Cleavage via Activated Caspase-2 in Infected RD Cells. Front Microbiol 2021; 12:684953. [PMID: 34046026 PMCID: PMC8144320 DOI: 10.3389/fmicb.2021.684953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
Enterovirus 71 (EV71) is the major causative pathogen of hand, foot, and mouth disease. The lack of understanding of the virus’s pathogenesis hinders the development of anti-virus drugs and the control of EV71 infection. Our previous studies have demonstrated that both mitochondria and endoplasmic reticulum (ER) were altered significantly in EV71 infected cells, but the mechanism is still unclear. In this study, we investigated the effects of EV71 infection on the expression of INF2, a key regulator factor in ER-Mitochondria communication and mitochondrial fission. We found that INF2 was cleaved in EV71 infected RD cells. The INF2 cleavage occurred at Aspartic 1,051 of INF2 and is mediated by activated caspases, predominantly by activated caspase-2. The subcellular localization of INF2 and caspase-2 was significantly altered in infected cells. We speculate that caspase-2-mediated INF2 cleavage is involved in forming viral replication organelles (ROs) and is a positive feedback regulatory mechanism of mitochondrial disorders caused by EV71 infection.
Collapse
Affiliation(s)
- Bei Wang
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chongyang Zhang
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Congci Yu
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhu
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Tang
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Huang
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhendong Zhao
- National Health Commission (NHC) Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Role of pyroptosis in diabetic retinopathy and its therapeutic implications. Eur J Pharmacol 2021; 904:174166. [PMID: 33979651 DOI: 10.1016/j.ejphar.2021.174166] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022]
Abstract
Pyroptosis has recently been established as a term of programmed-inflammatory cell death. Pyroptosis is mainly divided into two molecular signaling pathways, including caspase-1-dependent canonical and caspase-4/5/11-dependent non-canonical inflammasome pathways. Extensive investigations have reported inflammasome activation facilitates the maturation and secretion of the inflammatory factors interleukin-1β/18 (IL-1β/18), cleavage of gasdermin D (GSDMD), and leading to the stimulation of pyroptosis-mediated cell death. Furthermore, accumulating studies report NLRP3 inflammasome activation plays a significant role in triggering the pyroptosis-mediated cell death and promotes the pathogenesis of diabetic retinopathy (DR). Our current review elaborates on the molecular mechanisms of pyroptosis-signaling pathways and their potential roles in the pathogenesis and impact of DR development. We also emphasize several investigational molecules regulating key steps in pyroptotic-cell death to create new comprehensions and findings to explore the pathogenesis of DR advancement. Our narrative review concisely suggests these potential pharmacological agents could be promising therapies to treat and manage DR in the future.
Collapse
|
30
|
Luo R, Delaunay‐Moisan A, Timmis K, Danchin A. SARS-CoV-2 biology and variants: anticipation of viral evolution and what needs to be done. Environ Microbiol 2021; 23:2339-2363. [PMID: 33769683 PMCID: PMC8251359 DOI: 10.1111/1462-2920.15487] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022]
Abstract
The global propagation of SARS-CoV-2 and the detection of a large number of variants, some of which have replaced the original clade to become dominant, underscores the fact that the virus is actively exploring its evolutionary space. The longer high levels of viral multiplication occur - permitted by high levels of transmission -, the more the virus can adapt to the human host and find ways to success. The third wave of the COVID-19 pandemic is starting in different parts of the world, emphasizing that transmission containment measures that are being imposed are not adequate. Part of the consideration in determining containment measures is the rationale that vaccination will soon stop transmission and allow a return to normality. However, vaccines themselves represent a selection pressure for evolution of vaccine-resistant variants, so the coupling of a policy of permitting high levels of transmission/virus multiplication during vaccine roll-out with the expectation that vaccines will deal with the pandemic, is unrealistic. In the absence of effective antivirals, it is not improbable that SARS-CoV-2 infection prophylaxis will involve an annual vaccination campaign against 'dominant' viral variants, similar to influenza prophylaxis. Living with COVID-19 will be an issue of SARS-CoV-2 variants and evolution. It is therefore crucial to understand how SARS-CoV-2 evolves and what constrains its evolution, in order to anticipate the variants that will emerge. Thus far, the focus has been on the receptor-binding spike protein, but the virus is complex, encoding 26 proteins which interact with a large number of host factors, so the possibilities for evolution are manifold and not predictable a priori. However, if we are to mount the best defence against COVID-19, we must mount it against the variants, and to do this, we must have knowledge about the evolutionary possibilities of the virus. In addition to the generic cellular interactions of the virus, there are extensive polymorphisms in humans (e.g. Lewis, HLA, etc.), some distributed within most or all populations, some restricted to specific ethnic populations and these variations pose additional opportunities for/constraints on viral evolution. We now have the wherewithal - viral genome sequencing, protein structure determination/modelling, protein interaction analysis - to functionally characterize viral variants, but access to comprehensive genome data is extremely uneven. Yet, to develop an understanding of the impacts of such evolution on transmission and disease, we must link it to transmission (viral epidemiology) and disease data (patient clinical data), and the population granularities of these. In this editorial, we explore key facets of viral biology and the influence of relevant aspects of human polymorphisms, human behaviour, geography and climate and, based on this, derive a series of recommendations to monitor viral evolution and predict the types of variants that are likely to arise.
Collapse
Affiliation(s)
- Ruibang Luo
- Department of Computer ScienceThe University of Hong KongBonham RoadPokfulamHong Kong
| | - Agnès Delaunay‐Moisan
- Université Paris‐Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC)Gif‐sur‐Yvette91198France
| | - Kenneth Timmis
- Institute of MicrobiologyTechnical University of BraunschweigBraunschweigGermany
| | - Antoine Danchin
- Kodikos Labs, Institut Cochin, 24 rue du Faubourg Saint‐JacquesParis75014France
- School of Biomedical Sciences, Li Kashing Faculty of MedicineUniversity of Hong Kong21 Sassoon RoadHong Kong
| |
Collapse
|
31
|
Riera Romo M. Cell death as part of innate immunity: Cause or consequence? Immunology 2021; 163:399-415. [PMID: 33682112 DOI: 10.1111/imm.13325] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Regulated or programmed cell death plays a critical role in the development and tissue organization and function. In addition, it is intrinsically connected with immunity and host defence. An increasing cellular and molecular findings cause a change in the concept of cell death, revealing an expanding network of regulated cell death modalities and their biochemical programmes. Likewise, recent evidences demonstrate the interconnection between cell death pathways and how they are involved in different immune mechanisms. This work provides an overview of the main cell death programmes and their implication in innate immunity not only as an immunogenic/inflammatory process, but also as an active defence strategy during immune response and at the same time as a regulatory mechanism.
Collapse
Affiliation(s)
- Mario Riera Romo
- Radiology Department, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Liu Y, Xu X, Wang X, Zhu T, Li J, Pang Y, Li Q. Analysis of the lamprey genotype provides insights into caspase evolution and functional divergence. Mol Immunol 2021; 132:8-20. [PMID: 33524772 DOI: 10.1016/j.molimm.2021.01.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/28/2022]
Abstract
The cysteine-containing aspartate specific proteinase (caspase) family plays important roles in apoptosis and the maintenance of homeostasis in lampreys. We conducted genomic and functional comparisons of six distinct lamprey caspase groups with human counterparts to determine how these expanded molecules evolved to adapt to the changing caspase-mediated signaling pathways. Our results showed that lineage-specific duplication and rearrangement were responsible for expanding lamprey caspases 3 and 7, whereas caspases 1, 6, 8, and 9 maintained a relatively stable genome and protein structure. Lamprey caspase family molecules displayed various expression patterns and were involved in the innate immune response. Caspase 1 and 7 functioned as a pattern recognition receptor with a broad-spectrum of microbial recognition and bactericidal effect. Additionally, caspases 1 and 7 may induce cell apoptosis in a time- and dose-dependent manner; however, apoptosis was inhibited by caspase inhibitors. Thus, these molecules may reflect the original state of the vertebrates caspase family. Our phylogenetic and functional data provide insights into the evolutionary history of caspases and illustrate their functional characteristics in primitive vertebrates.
Collapse
Affiliation(s)
- Ying Liu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Xiaoluan Xu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Xiaotong Wang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Ting Zhu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Jun Li
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Yue Pang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Qingwei Li
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| |
Collapse
|
33
|
Buenahora MR, Lafaurie GI, Perdomo SJ. Identification of HPV16-p16 INK4a mediated methylation in oral potentially malignant disorder. Epigenetics 2020; 16:1016-1030. [PMID: 33164635 DOI: 10.1080/15592294.2020.1834923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To evaluate the possible involvement of epigenetic modulation by HPV16-p16INK4a in oral potentially malignant disorder (OPMD). We generated DNA-methylation profiles, according to p16INK4a expression and HPV16 genotype (positive or negative), of OPMD samples and p16INK4a-HPV16 negative samples (used as control), using reduced-representation bisulphite sequencing (RRBS-Seq- Illumina) technology. Twelve samples, four for each group, as follows: 1) p16INK4a+ HPV16+; 2) p16INK4a+ HPV16-; 3) p16INK4a- HPV16-, were analysed in triplicate for DNA-methylation profiles. Fifty-four per cent of DMRs were hypermethylated and 46% were hypomethylated. An increase in methylation of loci in OPMD was independent of the presence of HPV. The hypermethylated genes in HPV+ samples were associated with signalling pathways such as NICD traffics to nucleus, signalling by NOTCH1 (p = 0.008), Interferon-gamma (p = 0.008) and Interleukin-6 signalling (p = 0.027). The hypomethylated genes in HPV infection were associated with TRAF3-dependent IRF activation pathway (p = 0.002), RIG-I/MDA5 mediated induction of IFN-alpha/beta pathways (p = 0.005), TRAF6 mediated IRF7 activation (p = 0.009), TRIF-mediated TLR3/TLR4 signalling (p = 0.011) and MyD88-independent cascade release of apoptotic factors (p = 0.011). Protein association analysis of DMRs in OPMD revealed 19 genes involved in the cell cycle regulation, immune system, and focal adhesion. Aberrantly methylated loci in OPMD were observed in p16INK4a positive samples which suggests that a shift in global methylation status may be important for cancer progression. The results suggest that HPV infection in OPMD induces modulation of genes related to the immune system and regulation of the cellular cycle.
Collapse
Affiliation(s)
- Maria Rosa Buenahora
- Unit of Oral Clinical Epidemiology, School of Dentistry, El Bosque University, Bogotá, Colombia
| | - Gloria Inés Lafaurie
- Unit of Basic Oral Investigation, School of Dentistry, El Bosque University, Bogotá, Colombia
| | - Sandra J Perdomo
- Cellular and Molecular Immunology Research Group, Universidad El Bosque, Bogotá, Colombia
| |
Collapse
|
34
|
Lahnsteiner F. Differences in immune components of blood, spleen and head kidney between diploid and auto- and allotriploid Salmonidae. Tissue Cell 2020; 67:101445. [PMID: 33099197 DOI: 10.1016/j.tice.2020.101445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 01/16/2023]
Abstract
Immune components were investigated in peripheral blood and in spleen and head kidney of autotriploid Salmo trutta f. lacustris, Salvelinus fontinalis, and Salvelinus umbla, and of allotriploid hybrids of S. trutta f. lacustris x Onchorynchus mykiss and S. fontinalis x O. mykiss in comparison to their diploid parents. In peripheral blood the number of lymphocytes was reduced in all investigated autotriploids and in the allotriploid S. trutta f. lacustris x O.mykiss, and the numbers of thrombocytes in autotriploid S. trutta f. lacustris and in both allotriploids. Alternative pathway of complement activity and immunoglobulin concentration were significantly decreased in all investigated auto- and allotriploids, lysozyme activity in autotriploid S. fontinalis and in both allotriploids. In the spleen of the 3 autotriploids the number of erythrocytes was increased, while the number of lymphoid precursor cells was decreased. In their head kidney the erythrocytes numbers were decreased and the numbers of erythropoietic precursor cells and the melanomacrophage centers were increased. Contrary, cytology of spleen and head kidney of the two allotriploid hybrids was similar to diploid controls. Caspase 1, caspase 6, lysozyme, and acid phosphatase activity and immunoglobulin concentration of spleen and head kidney showed specific changes which were related to cytological results. These data indicate alterations in immune system and in lymphoid organs of auto- and allotriploid Salmonidae.
Collapse
Affiliation(s)
- Franz Lahnsteiner
- Federal Agency for Water Management, Institute for Water Ecology, Fisheries and Lake Research, Scharfling 18, A-5310 Mondsee, Fishfarm Kreuzstein, Oberburgau 28, 4866, Unterach, Austria
| |
Collapse
|
35
|
Liu M, Zhou K, Xu Z, Ma H, Cao X, Yin X, Zeng W, Zahid A, Fu S, Ni K, Ye X, Zhou Y, Bai L, Zhou R, Jin T. Crystal structure of caspase-11 CARD provides insights into caspase-11 activation. Cell Discov 2020; 6:70. [PMID: 33083005 PMCID: PMC7552397 DOI: 10.1038/s41421-020-00201-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/20/2020] [Indexed: 01/16/2023] Open
Abstract
Murine caspase-11 is the centerpiece of the non-canonical inflammasome pathway that can respond to intracellular LPS and induce pyroptosis. Caspase-11 contains two components, an N-terminal caspase recruitment domain (CARD) and a C-terminal catalytic domain. The aggregation of caspase-11 is thought to promote the auto-processing and activation of caspase-11. However, the activation mechanism of caspase-11 remains unclear. In this study, we purified the caspase-11 CARD fused to an MBP tag and found it tetramerizes in solution. Crystallographic analysis reveals an extensive hydrophobic interface formed by the H1–2 helix mediating homotypic CARD interactions. Importantly, mutations of the helix H1–2 hydrophobic residues abolished the tetramerization of MBP-tagged CARD in solution and failed to induce pyroptosis in cells. Our study provides the first evidence of the homotypic interaction mode for an inflammatory caspase by crystal model. This finding demonstrates that the tetramerization of the N-terminal CARD can promote releasing of the catalytic domain auto-inhibition, leading to the caspase-11 activation.
Collapse
Affiliation(s)
- Muziying Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No.17 Lujiang Rd, Hefei, Anhui 230001 China.,Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Kang Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Zhihao Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No.17 Lujiang Rd, Hefei, Anhui 230001 China.,Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Huan Ma
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Xiaocong Cao
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Xueying Yin
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Weihong Zeng
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Ayesha Zahid
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Sicheng Fu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Kang Ni
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Xiaodong Ye
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No.17 Lujiang Rd, Hefei, Anhui 230001 China
| | - Li Bai
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, 200031 China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No.17 Lujiang Rd, Hefei, Anhui 230001 China.,Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, 200031 China
| |
Collapse
|
36
|
Appelberg S, Gupta S, Svensson Akusjärvi S, Ambikan AT, Mikaeloff F, Saccon E, Végvári Á, Benfeitas R, Sperk M, Ståhlberg M, Krishnan S, Singh K, Penninger JM, Mirazimi A, Neogi U. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg Microbes Infect 2020; 9:1748-1760. [PMID: 32691695 PMCID: PMC7473213 DOI: 10.1080/22221751.2020.1799723] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections engage cellular host pathways and innate immunity in infected cells remains largely elusive. We performed an integrative proteo-transcriptomics analysis in SARS-CoV-2 infected Huh7 cells to map the cellular response to the invading virus over time. We identified four pathways, ErbB, HIF-1, mTOR and TNF signaling, among others that were markedly modulated during the course of the SARS-CoV-2 infection in vitro. Western blot validation of the downstream effector molecules of these pathways revealed a dose-dependent activation of Akt, mTOR, S6K1 and 4E-BP1 at 24 hours post infection (hpi). However, we found a significant inhibition of HIF-1α through 24hpi and 48hpi of the infection, suggesting a crosstalk between the SARS-CoV-2 and the Akt/mTOR/HIF-1 signaling pathways. Inhibition of the mTOR signaling pathway using Akt inhibitor MK-2206 showed a significant reduction in virus production. Further investigations are required to better understand the molecular sequelae in order to guide potential therapy in the management of severe coronavirus disease 2019 (COVID-19) patients.
Collapse
Affiliation(s)
| | - Soham Gupta
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anoop T Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Flora Mikaeloff
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Elisa Saccon
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Marie Ståhlberg
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Shuba Krishnan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kamal Singh
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, Canada
| | - Ali Mirazimi
- Public Health Agency of Sweden, Solna, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,National Veterinary Institute, Uppsala, Sweden
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
37
|
Li J, Dong L, Zhu D, Zhang M, Wang K, Chen F. An effector caspase Sp-caspase first identified in mud crab Scylla paramamosain exhibiting immune response and cell apoptosis. FISH & SHELLFISH IMMUNOLOGY 2020; 103:442-453. [PMID: 32446967 DOI: 10.1016/j.fsi.2020.05.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
Apoptosis plays a key role in the immune defense against pathogen infection, and caspase is one of the most important protease enzyme families, which could initiate and execute apoptosis. Among crustaceans, several caspase genes have been reported. However, caspase in mud crab Scylla paramamosain, have not been identified yet. Here, in the present study, we characterized a new caspase, named as Sp-caspase, from S. paramamosain. The full-length cDNA sequence of Sp-caspase contained 966 bp open reading frame, encoding 322 amino acids, and its molecular weight was 36 kDa. This gene has three conserved domains of the caspase family, a prodomain, a large subunit P20 and a small subunit P10. Phylogenetic analysis showed that Sp-caspase was clustered into an effector caspase group. Sp-caspase mainly distributed in midgut, hepatopancreas, hemocytes and female ovaries, and the transcript was significantly regulated in different tissues after being challenged with Vibrio parahaemolyticus, Vibrio alginolyticus or LPS. After infection with V. alginolyticus, the apoptosis rate of hemocytes notably increased, while the mRNA level of Sp-caspase and hydrolysis activity of caspase 3/7 significantly decreased. Furthermore, in vitro assays showed that the recombinant protein tSp-caspase (deletion of Sp-caspase prodomain) could efficiently recognize and cleave human caspase 3/7 substrate Ac-DEVD-pNA, functioning as an effector caspase. Meanwhile, heterologous expression of Sp-caspase in several cell lines (HEK293T cells, HeLa cells and HighFive cells) could specifically induce cell apoptosis. Taken together, these data demonstrated that Sp-caspase could perform apoptosis as an effector caspase. In addition, it might be a negative regulator of hemocytes apoptosis under pathogen infection, which would contribute to homeostasis and immune defense of hemocytes in S. paramamosain.
Collapse
Affiliation(s)
- Jishan Li
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lixia Dong
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Depeng Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Min Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kejian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
38
|
Porcine Epidemic Diarrhea Virus and the Host Innate Immune Response. Pathogens 2020; 9:pathogens9050367. [PMID: 32403318 PMCID: PMC7281546 DOI: 10.3390/pathogens9050367] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a swine enteropathogenic coronavirus (CoV), is the causative agent of porcine epidemic diarrhea (PED). PED causes lethal watery diarrhea in piglets, which has led to substantial economic losses in many countries and is a great threat to the global swine industry. Interferons (IFNs) are major cytokines involved in host innate immune defense, which induce the expression of a broad range of antiviral effectors that help host to control and antagonize viral infections. PEDV infection does not elicit a robust IFN response, and some of the mechanisms used by the virus to counteract the host innate immune response have been unraveled. PEDV evades the host innate immune response by two main strategies including: (1) encoding IFN antagonists to disrupt innate immune pathway, and (2) hiding its viral RNA to avoid the exposure of viral RNA to immune sensors. This review highlights the immune evasion mechanisms employed by PEDV, which provides insights for the better understanding of PEDV-host interactions and developing effective vaccines and antivirals against CoVs.
Collapse
|
39
|
Zhu F, Ma X. Molecular characterization of troponin T in Scylla paramamosain and its role in Vibrio alginolyticus and white spot syndrome virus (WSSV) infection. FISH & SHELLFISH IMMUNOLOGY 2020; 99:392-402. [PMID: 32087277 DOI: 10.1016/j.fsi.2020.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
This study investigated the function of Troponin T (TnT) in the mud crab, Scylla paramamosain. The 1952 bp cDNA sequence of TnT was cloned from S. paramamosain using rapid amplification of cDNA ends (RACE) PCR. The quantitative real-time PCR analysis showed that TnT was highly expressed in the muscle and heart of S. paramamosain. Challenging with white spot syndrome virus (WSSV) or Vibrio alginolyticus (VA), two common pathogens that infect mud crabs, enhanced the expression of TnT in S. paramamosain. Knockdown of TnT using TnT-dsRNA led to up-regulating the expression of immune-related genes, such as c-type-lectin, toll-like-receptor, crustin antimicrobial peptide and prophenoloxidase. The cumulative mortality of WSSV- and VA-infected crabs was significantly increased following TnT knockdown. After WSSV or VA infection, TnT knockdown caused a significant reduction in phenoloxidase (PO) activity, superoxide dismutase (SOD) activity and total hemocyte count (THC), indicating a regulatory role of TnT in the innate immune response of S. paramamosain to pathogens. Apoptosis of hemocytes was higher in crabs treated with TnT-dsRNA compared with control crabs treated with phosphate-buffered saline. Knockdown of TnT increased apoptosis of hemocytes following VA infection, but reduced hemocyte apoptosis following WSSV infection. In summary, TnT may enhance the immune response of S. paramamosain to WSSV infection by regulating apoptosis, THC, PO activity and SOD activity. And TnT may play a positive role in the immune response against VA infection by regulating apoptosis, THC, SOD activity and PO activity.
Collapse
Affiliation(s)
- Fei Zhu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China.
| | - Xiongchao Ma
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China
| |
Collapse
|
40
|
Zhu JJ, Ramanathan P, Bishop EA, O’Donnell V, Gladue DP, Borca MV. Mechanisms of African swine fever virus pathogenesis and immune evasion inferred from gene expression changes in infected swine macrophages. PLoS One 2019; 14:e0223955. [PMID: 31725732 PMCID: PMC6855437 DOI: 10.1371/journal.pone.0223955] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/01/2019] [Indexed: 12/22/2022] Open
Abstract
African swine fever (ASF) is a swine disease caused by a large, structurally complex, double-stranded DNA virus, African swine fever virus (ASFV). In domestic pigs, acute infection by highly virulent ASF viruses causes hemorrhagic fever and death. Previous work has suggested that ASFV pathogenesis is primarily mediated by host cytokines produced by infected monocytes and macrophages. To better understand molecular mechanisms mediating virus pathogenesis and immune evasion, we used transcriptome analysis to identify gene expression changes after ASFV infection in ex vivo swine macrophages. Our results suggest that the cytokines of TNF family including FASLG, LTA, LTB, TNF, TNFSF4, TNFSF10, TNFSF13B and TNFSF18 are the major causative cytokine factors in ASF pathogenesis via inducing apoptosis. Other up-regulated proinflammatory cytokines (IL17F and interferons) and down-regulated anti-inflammatory cytokine (IL10) may also significantly contribute to ASF pathogenesis and cause excessive tissue inflammatory responses. The differential expression of genes also indicates that ASFV could evade both the innate and adaptive immune responses by (i) inhibiting MHC Class II antigen processing and presentation, (ii) avoiding CD8+ T effector cells and neutrophil extracellular traps via decreasing expression of neutrophil/CD8+ T effector cell-recruiting chemokines, (iii) suppressing M1 activation of macrophages, (iv) inducing immune suppressive cytokines, and (v) inhibiting the processes of macrophage autophagy and apoptosis. These results provide novel information to further investigate and better understand the mechanism of pathogenesis and immune evasion of this devastating swine disease.
Collapse
Affiliation(s)
- James J. Zhu
- USDA-ARS, FADRU, Plum Island Animal Disease Center, Orient, New York, United States of America
- * E-mail: (JJZ); (MVB)
| | - Palaniappan Ramanathan
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, Tennessee, United States of America
| | - Elizabeth A. Bishop
- USDA-ARS, FADRU, Plum Island Animal Disease Center, Orient, New York, United States of America
| | - Vivian O’Donnell
- USDA-APHIS, Plum Island Animal Disease Center, Orient, New York, United States of America
| | - Douglas P. Gladue
- USDA-ARS, FADRU, Plum Island Animal Disease Center, Orient, New York, United States of America
| | - Manuel V. Borca
- USDA-ARS, FADRU, Plum Island Animal Disease Center, Orient, New York, United States of America
- * E-mail: (JJZ); (MVB)
| |
Collapse
|
41
|
Chen YL, Han K, Huang X, Zhang Z, Wan X, Ren Q. Caspase-3C gene from red swamp crayfish, Procambarus clarki: Characterization and expression in response to pathogenic infection. FISH & SHELLFISH IMMUNOLOGY 2019; 94:792-799. [PMID: 31585244 DOI: 10.1016/j.fsi.2019.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/28/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
The caspase is an essential module in the Drosophila immune deficiency (IMD) pathway, which plays a crucial role in countering pathogen infection. In this study, a gene named PcCaspase-3C was found in Procambarus clarkia with a full-length of 4684 bp, including a 1572 bp opening reading frame, which encoded a putative protein of 523 amino acids. PcCaspase-3C contained a CASc domain constituted of 237 amino acids. The PcCaspase-3C gene was primarily expressed in heart, stomach, and intestine, while less in gonad, hepatopancreas, gills, and hemocytes, with the least expression in muscle. Infection with Staphyloccocus aureus, Vibrio parahaemolyticus or white spot syndrome virus (WSSV) induced an up-regulated expression of PcCaspase-3C in intestine or stomach to varying degrees. When PcCaspase-3C was silenced by double-stranded RNA, the expression of some antimicrobial peptides such as ALF2, ALF5, ALF6, Cru3, Cru4, and Lys was significantly inhibited. In addition, silencing of PcCaspase-3C accelerated infection with WSSV in vivo. According to these results, we suggest that PcCaspase-3C might play a crucial role in the immune response of P. clarkia against pathogenic bacterial and viral infections.
Collapse
Affiliation(s)
- Yu-Lei Chen
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian Province, 361021, China
| | - Keke Han
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Xin Huang
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Zhuoxing Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Xihe Wan
- Institute of Oceanology and Marine Fisheries, Jiangsu, PR China.
| | - Qian Ren
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu Province, 222005, China; College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
42
|
Takaoka A, Yamada T. Regulation of signaling mediated by nucleic acid sensors for innate interferon-mediated responses during viral infection. Int Immunol 2019; 31:477-488. [PMID: 30985869 PMCID: PMC7110195 DOI: 10.1093/intimm/dxz034] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
Type I and type III interferons are important anti-viral cytokines that are massively induced during viral infection. This dynamic process is regulated by many executors and regulators for efficient eradication of invading viruses and protection from harmful, excessive responses. An array of innate sensors recognizes virus-derived nucleic acids to activate their downstream signaling to evoke cytokine responses including interferons. In particular, a cytoplasmic RNA sensor RIG-I (retinoic acid-inducible gene I) is involved in the detection of multiple types of not only RNA viruses but also DNA viruses. Accumulating findings have revealed that activation of nucleic acid sensors and the related signaling mediators is regulated on the basis of post-translational modification such as ubiquitination, phosphorylation and ADP-ribosylation. In addition, long non-coding RNAs (lncRNAs) have been implicated as a new class of regulators in innate signaling. A comprehensive understanding of the regulatory mechanisms of innate sensor activation and its signaling in host-virus interaction will provide a better therapeutic strategy to efficiently control viral infection and maintain immune homeostasis.
Collapse
Affiliation(s)
- Akinori Takaoka
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Taisho Yamada
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
43
|
Naomaitai Ameliorated Brain Damage in Rats with Vascular Dementia by PI3K/PDK1/AKT Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2702068. [PMID: 30867669 PMCID: PMC6379870 DOI: 10.1155/2019/2702068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/21/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022]
Abstract
Background/Aims Naomaitai can improve blood perfusion and ameliorate the damage in the paraventricular white matter. This study was focused on observing the neuroprotective effect of Naomaitai on the vascular dementia of rat and exploring the action mechanism of PI3K/PDK1/AKT signaling pathway. Methods A vascular dementia model of rats was established by permanent, bilateral common carotid artery occlusion. Rats' behavior was tested by Neurological deficit score and the Morris water maze. The pathology and apoptosis were detected through HE staining and TUNEL assay. Myelin sheath loss and nerve fiber damage were detected by LFB staining. Inflammatory factors, oxidative stress, and brain damage markers were detected through ELISA. The expression of apoptosis-related proteins and PI3K/PDK1/AKT signaling pathway related proteins were measured by western blot. The expressions of PI3K, PDK1, AKT, and MBP in paraventricular white matter cells were detected by immunofluorescence. Results Naomaitai treatment decreased neurological function score in rats with vascular dementia, ameliorated paraventricular white matter damage caused by long-term hypoxia, and hypoperfusion reduced the brain injury markers S-100β and NSE contents, suppressed inflammatory reaction and oxidative stress, reduced IL-1β, IL-6, TNF-α, and MDA contents, and remarkably increased IL-10 and SOD contents. TUNEL and western blot assay showed that Naomaitai treatment decreased neuronal cell apoptosis, increased Bcl-2 expression, and reduced caspase-3 and Bax expression. Furthermore, we found Naomaitai inhibited PI3K and PDK1 expression and activated phosphorylated AKT protein in rats with vascular dementia. However, the protective effect of Naomatai in rats with vascular dementia was inhibited, and expression of PI3K signaling pathway-related proteins was blocked after administration of PI3K inhibitor. Conclusion Naomaitai can ameliorate brain damage in rats with vascular dementia, inhibit neuronal apoptosis, and have anti-inflammatory and antioxidative stress effects, which may be regulated by the PI3K/PDK1/AKT signaling pathway.
Collapse
|
44
|
Visualization of perforin/gasdermin/complement-formed pores in real cell membranes using atomic force microscopy. Cell Mol Immunol 2018; 16:611-620. [PMID: 30283066 PMCID: PMC6804747 DOI: 10.1038/s41423-018-0165-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/05/2023] Open
Abstract
Different types of pores ubiquitously form in cell membranes, leading to various types of cell death that profoundly influence the fate of inflammation and the disease status. However, these pores have never truly been visualized to date. Atomic force microscopy (AFM), which is emerging as a powerful tool to analyze the mechanical properties of biomolecules and cells, is actually an excellent imaging platform that allows biological samples to be visualized by probing surface roughness at the level of atomic resolution. Here, membrane pore structures were clearly visualized using AFM. This visualization not only describes the aperture and depth of the pore complexes but also highlights differences among the pores formed by perforin and gasdermins in tumor cell membranes and by complement in immune cell membranes. Additionally, this type of visualization also reveals the dynamic process of pore formation, fusion, and repair.
Collapse
|
45
|
Zheng Z, Xu W, Aweya JJ, Zhong M, Liu S, Lun J, Chen J, Zhang Y. Functional domains of Litopenaeus vannamei transglutaminase and their involvement in immunoregulation in shrimp. FISH & SHELLFISH IMMUNOLOGY 2018; 81:168-175. [PMID: 30010014 DOI: 10.1016/j.fsi.2018.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/08/2018] [Accepted: 07/11/2018] [Indexed: 06/08/2023]
Abstract
Shrimps, which mainly rely on their innate immune system to response to infectious pathogens, have clottable proteins as an important component of this system. While transglutaminases (TGase) are found in Litopenaeus vannamei and constitute part of the coagulation system, the specific immune-related roles played by its functional domains in the immunoregulation of shrimp has not been well understood. In the present study, we report that the Ig-like domain of L. vannamei transglutaminase (TGase-C) is the main immune-related domain among the three functional domains, as it had higher bacterial agglutinative activity against Vibrio parahaemolyticus and Streptococcus iniae. Using Co-immunoprecipitation and LC-MS/MS analysis, TGase-C was shown to interact with 474 proteins, of which 52 proteins were annotated to L. vannamei. More than half of the L. vannamei annotated proteins have immune-related functions, including apoptosis. Further analysis using pull-down assay revealed that TGase-C interacted with CAP-3 (a homologue of caspase 3). In addition, siRNA-mediated knockdown of LvTGase significantly (p < 0.01) increased the expression level of LvCAP-3 coupled with a significant (p < 0.01) increase in caspase 3/7 activity, suggesting that probably LvTGase participates in shrimp immune response by modulating the activity of LvCAP-3. These findings thus suggest the Ig-like functional domain of L. vannamei's transglutaminase is the domain that is involved in immunoregulation in shrimp.
Collapse
Affiliation(s)
- Zhou Zheng
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Wenning Xu
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Jude Juventus Aweya
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Mingqi Zhong
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Shangjie Liu
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Jingsheng Lun
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Jiehui Chen
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China
| | - Yueling Zhang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China.
| |
Collapse
|
46
|
Intestinal Lamina Propria CD4 + T Cells Promote Bactericidal Activity of Macrophages via Galectin-9 and Tim-3 Interaction during Salmonella enterica Serovar Typhimurium Infection. Infect Immun 2018; 86:IAI.00769-17. [PMID: 29844236 DOI: 10.1128/iai.00769-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/21/2018] [Indexed: 12/13/2022] Open
Abstract
The intestinal immune system is crucial for protection from pathogenic infection and maintenance of mucosal homeostasis. We studied the intestinal immune microenvironment in a Salmonella enterica serovar Typhimurium intestinal infection mouse model. Intestinal lamina propria macrophages are the main effector cells in innate resistance to intracellular microbial pathogens. We found that S Typhimurium infection augmented Tim-3 expression on intestinal lamina propria CD4+ T cells and enhanced galectin-9 expression on F4/80+ CD11b+ macrophages. Moreover, CD4+ T cells promoted the activation and bactericidal activity of intestinal F4/80+ CD11b+ macrophages via the Tim-3/galectin-9 interaction during S Typhimurium infection. Blocking the Tim-3/galectin-9 interaction with α-lactose significantly attenuated the bactericidal activity of intracellular S Typhimurium by macrophages. Furthermore, the Tim-3/galectin-9 interaction promoted the formation and activation of inflammasomes, which led to caspase-1 cleavage and interleukin 1β (IL-1β) secretion. The secretion of active IL-1β further improved bactericidal activity of macrophages and galectin-9 expression on macrophages. These results demonstrated the critical role of the cross talk between CD4+ T cells and macrophages, particularly the Tim-3/galectin-9 interaction, in antimicrobial immunity and the control of intestinal pathogenic infections.
Collapse
|
47
|
Shaalan A, Carpenter G, Proctor G. Caspases are key regulators of inflammatory and innate immune responses mediated by TLR3 in vivo. Mol Immunol 2018; 94:190-199. [PMID: 29331803 DOI: 10.1016/j.molimm.2017.12.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/14/2017] [Accepted: 12/19/2017] [Indexed: 01/02/2023]
Abstract
Understanding the key regulators which impact the innate immune response during initial phases of tissue injury, can advance the use of therapeutic approaches which aim at attenuating inflammation and organ damage. Recognition of microbial components by TLRs, initiates the transcription of innate immune signal pathways, that induce the expression of key inflammatory mediators: cytokines, chemokines and adhesion molecules. Beside regulating apoptotic cell death, recent studies have revealed distinct roles for caspases in the optimal production of inflammatory cytokines and host defense against injurious infections. Whether caspases can play an immune regulatory role in vivo has not been sufficiently investigated. This study aims to explore whether the pan caspase inhibitor z-VAD-fmk can control inflammation and cytokine production subsequent to challenging the innate immunity of the exocrine secretory tissues in vivo. Submandibular glands (SMGs) of the C57BL/6 mice were challenged with the TLR3 stimulant: polyinosinic-polycytidylic acid (poly (I:C)). Results obtained from the current study provide evidence that caspases can control immune responses downstream of TLR3 ligation. The present work proposes a novel mechanism that can prevent overactivation of the innate immunity, which typically leads to fatal immune disorders.
Collapse
Affiliation(s)
- Abeer Shaalan
- Mucosal and Salivary Biology Division, Dental Institute, King's College London, Guy's Hospital, Floor 17, Tower Wing, London SE1 9RT, UK.
| | - Guy Carpenter
- Mucosal and Salivary Biology Division, Dental Institute, King's College London, Guy's Hospital, Floor 17, Tower Wing, London SE1 9RT, UK
| | - Gordon Proctor
- Mucosal and Salivary Biology Division, Dental Institute, King's College London, Guy's Hospital, Floor 17, Tower Wing, London SE1 9RT, UK
| |
Collapse
|