1
|
Hsieh LL, Looney M, Figueroa A, Massaccesi G, Stavrakis G, Anaya EU, D'Alessio FR, Ordonez AA, Pekosz AS, DeFilippis VR, Karakousis PC, Karaba AH, Cox AL. Bystander monocytic cells drive infection-independent NLRP3 inflammasome response to SARS-CoV-2. mBio 2024; 15:e0081024. [PMID: 39240187 PMCID: PMC11481483 DOI: 10.1128/mbio.00810-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
The pathogenesis of COVID-19 is associated with a hyperinflammatory immune response. Monocytes and macrophages play a central role in this hyperinflammatory response to SARS-CoV-2. NLRP3 inflammasome activation has been observed in monocytes of patients with COVID-19, but the mechanism and consequences of inflammasome activation require further investigation. In this study, we inoculated a macrophage-like THP-1 cell line, primary differentiated human nasal epithelial cell (hNEC) cultures, and primary monocytes with SARS-CoV-2. We found that the activation of the NLRP3 inflammasome in macrophages does not rely on viral replication, receptor-mediated entry, or actin-dependent entry. SARS-CoV-2 productively infected hNEC cultures without triggering the production of inflammasome cytokines IL-18 and IL-1β. Importantly, these cytokines did not inhibit viral replication in hNEC cultures. SARS-CoV-2 inoculation of primary monocytes led to inflammasome activation and induced a macrophage phenotype in these cells. Monocytic cells from bronchoalveolar lavage (BAL) fluid, but not from peripheral blood, of patients with COVID-19, showed evidence of inflammasome activation, expressed the proinflammatory marker CD11b, and displayed oxidative burst. These findings highlight the central role of activated macrophages, as a result of direct viral sensing, in COVID-19 and support the inhibition of IL-1β and IL-18 as potential therapeutic strategies to reduce immunopathology without increasing viral replication. IMPORTANCE Inflammasome activation is associated with severe COVID-19. The impact of inflammasome activation on viral replication and mechanistic details of this activation are not clarified. This study advances our understanding of the role of inflammasome activation in macrophages by identifying TLR2, NLRP3, ASC, and caspase-1 as dependent factors in this activation. Further, it highlights that SARS-CoV-2 inflammasome activation is not a feature of nasal epithelial cells but rather activation of bystander macrophages in the airway. Finally, we demonstrate that two pro inflammatory cytokines produced by inflammasome activation, IL-18 and IL-1β, do not restrict viral replication and are potential targets to ameliorate pathological inflammation in severe COVID-19.
Collapse
Affiliation(s)
- Leon L. Hsieh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Monika Looney
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexis Figueroa
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Massaccesi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Georgia Stavrakis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Eduardo U. Anaya
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Franco R. D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alvaro A. Ordonez
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew S. Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Petros C. Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew H. Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L. Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Liu L, Zhang L, Hao X, Wang Y, Zhang X, Ge L, Wang P, Tian B, Zhang M. Coronavirus envelope protein activates TMED10-mediated unconventional secretion of inflammatory factors. Nat Commun 2024; 15:8708. [PMID: 39379362 PMCID: PMC11461611 DOI: 10.1038/s41467-024-52818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
The precise cellular mechanisms underlying heightened proinflammatory cytokine production during coronavirus infection remain incompletely understood. Here we identify the envelope (E) protein in severe coronaviruses (SARS-CoV-2, SARS, or MERS) as a potent inducer of interleukin-1 release, intensifying lung inflammation through the activation of TMED10-mediated unconventional protein secretion (UcPS). In contrast, the E protein of mild coronaviruses (229E, HKU1, or OC43) demonstrates a less pronounced effect. The E protein of severe coronaviruses contains an SS/DS motif, which is not present in milder strains and facilitates interaction with TMED10. This interaction enhances TMED10-oligomerization, facilitating UcPS cargo translocation into the ER-Golgi intermediate compartment (ERGIC)-a pivotal step in interleukin-1 UcPS. Progesterone analogues were identified as compounds inhibiting E-enhanced release of proinflammatory factors and lung inflammation in a Mouse Hepatitis Virus (MHV) infection model. These findings elucidate a molecular mechanism driving coronavirus-induced hyperinflammation, proposing the E-TMED10 interaction as a potential therapeutic target to counteract the adverse effects of coronavirus-induced inflammation.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Lijingyao Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinyan Hao
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaochun Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peihui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Boxue Tian
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Min Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China.
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
3
|
Tan X, Gao X, Zheng H, Yuan H, Liu H, Ran Q, Luo M. Platelet dysfunction caused by differentially expressed genes as key pathogenic mechanisms in COVID-19. Minerva Cardiol Angiol 2024; 72:517-534. [PMID: 38804627 DOI: 10.23736/s2724-5683.24.06501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
At the end of 2019, the novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became prevalent worldwide, which brought a heavy medical burden and tremendous economic losses to the world population. In addition to the common clinical respiratory symptoms such as fever, cough and headache, patients with COVID-19 often have hematological diseases, especially platelet dysfunction. Platelet dysfunction usually leads to multiple organ dysfunction, which is closely related to patient severity or mortality. In addition, studies have confirmed significant changes in the gene expression profile of circulating platelets under SARS-CoV-2 infection, which will further lead to changes in platelet function. At the same time, studies have shown that platelets may absorb SARS-COV-2 mRNA independently of ACE2, which further emphasizes the importance of the stability of platelet function in defense against SARS-CoV-2 infection. This study reviewed the relationship between COVID-19 and platelet and SARS-CoV-2 damage to the circulatory system, and further analyzed the significantly differentially expressed mRNA in platelets after infection with SARS-CoV-2 on the basis of previous studies. The top eight hub genes were identified as NLRP3, MT-CO1, CD86, ICAM1, MT-CYB, CASP8, CXCL8 and CXCR4. Subsequently, the effects of SARS-CoV-2 infection on platelet transcript abnormalities and platelet dysfunction were further explored on the basis of 8 hub genes. Finally, the treatment measures of complications caused by platelet dysfunction in patients with COVID-19 were discussed in detail, so as to provide reference for the prevention, diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Xiaoyong Tan
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Huanhuan Zheng
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Hui Yuan
- Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong Liu
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Qijun Ran
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China -
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Mao S, Liu X, Wu D, Zhang Z, Sun D, Ou X, Huang J, Wu Y, Yang Q, Tian B, Chen S, Liu M, Zhu D, Zhang S, Zhao X, He Y, Wu Z, Jia R, Wang M, Cheng A. Duck hepatitis A virus 1-encoded 2B protein disturbs ion and organelle homeostasis to promote NF-κB/NLRP3-mediated inflammatory response. Int J Biol Macromol 2024; 280:135876. [PMID: 39322136 DOI: 10.1016/j.ijbiomac.2024.135876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Previous studies by our group and others have highlighted the critical role of hyperinflammation in the pathogenicity of duck hepatitis A virus 1 (DHAV-1), an avian picornavirus that has caused significant devastation in the duck industry worldwide for decades. However, the precise mechanisms by which DHAV-1 infection regulates the inflammatory responses, particularly the production of IL-1β, remain poorly understood. In this study, we demonstrate that DHAV-1 infection triggers NF-κB- and NLRP3 inflammasome-mediated IL-1β production. Mechanistically, DHAV-1 infection, particularly its replication and translation, disrupts cellular homeostasis of Ca2+, K+, ROS and cathepsin, which act cooperatively as assembly signals for NLRP3 inflammasome activation. By screening DHAV-1-encoded proteins, we identified that the viroporin 2B dominates NF-κB as well as NLRP3 inflammasome activation. Mutation analysis revealed that I43 within the 2B protein is the key amino acid for Ca2+ mobilization and subsequent activation of NF-κB transcriptional activity and NLRP3 inflammasome. Moreover, DHAV-1 infection and the 2B protein activate the MAVS- and MyD88-NF-κB pathways by relay, providing the necessary priming signals for NLRP3 inflammasome activation. In summary, our findings elucidate a mechanism through which DHAV-1 triggers inflammatory responses via NF-κB/NLRP3 inflammasome activation, offering new perspectives on DHAV-1 pathogenesis and informing the development of targeted anti-DHAV-1 treatments.
Collapse
Affiliation(s)
- Sai Mao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Xinghong Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dandan Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhilong Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Di Sun
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Bin Tian
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Yu He
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Zhen Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China.
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
5
|
Xu Y, Qu X, Liang M, Huang D, Jin M, Sun L, Chen X, Liu F, Qiu Z. Focus on the role of calcium signaling in ferroptosis: a potential therapeutic strategy for sepsis-induced acute lung injury. Front Med (Lausanne) 2024; 11:1457882. [PMID: 39355841 PMCID: PMC11442327 DOI: 10.3389/fmed.2024.1457882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
By engaging in redox processes, ferroptosis plays a crucial role in sepsis-induced acute lung injury (ALI). Although iron stimulates calcium signaling through the stimulation of redox-sensitive calcium pathways, the function of calcium signals in the physiological process of ferroptosis in septic ALI remains unidentified. Iron homeostasis disequilibrium in ferroptosis is frequently accompanied by aberrant calcium signaling. Intracellular calcium overflow can be a symptom of dysregulation of the cellular redox state, which is characterized by iron overload during the early phase of ferroptosis. This can lead to disruptions in calcium homeostasis and calcium signaling. The mechanisms controlling iron homeostasis and ferroptosis are reviewed here, along with their significance in sepsis-induced acute lung injury, and the potential role of calcium signaling in these processes is clarified. We propose that the development of septic acute lung injury is a combined process involving the bidirectional interaction between iron homeostasis and calcium signaling. Our goal is to raise awareness about the pathophysiology of sepsis-induced acute lung injury and investigate the relationship between these mechanisms and ferroptosis. We also aimed to develop calcium-antagonistic therapies that target ferroptosis in septic ALI and improve the quality of survival for patients suffering from acute lung injury.
Collapse
Affiliation(s)
- Yifei Xu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintian Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghao Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minyan Jin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianhai Chen
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fen Liu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhanjun Qiu
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Somberg NH, Sučec I, Medeiros-Silva J, Jo H, Beresis R, Syed AM, Doudna JA, Hong M. Oligomeric State and Drug Binding of the SARS-CoV-2 Envelope Protein Are Sensitive to the Ectodomain. J Am Chem Soc 2024; 146:24537-24552. [PMID: 39167680 DOI: 10.1021/jacs.4c07686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The envelope (E) protein of SARS-CoV-2 is the smallest of the three structural membrane proteins of the virus. E mediates budding of the progeny virus in the endoplasmic reticulum Golgi intermediate compartment of the cell. It also conducts ions, and this channel activity is associated with the pathogenicity of SARS-CoV-2. The structural basis for these functions is still poorly understood. Biochemical studies of E in detergent micelles found a variety of oligomeric states, but recent 19F solid-state NMR data indicated that the transmembrane domain (ETM, residues 8-38) forms pentamers in lipid bilayers. Hexamethylene amiloride (HMA), an E inhibitor, binds the pentameric ETM at the lipid-exposed helix-helix interface. Here, we investigate the oligomeric structure and drug interaction of an ectodomain-containing E construct, ENTM (residues 1-41). Unexpectedly, 19F spin diffusion NMR data reveal that ENTM adopts an average oligomeric state of dimers instead of pentamers in lipid bilayers. A new amiloride inhibitor, AV-352, shows stronger inhibitory activity than HMA in virus-like particle assays. Distance measurements between 13C-labeled protein and a trifluoromethyl group of AV-352 indicate that the drug binds ENTM with a higher stoichiometry than ETM. We measured protein-drug contacts using a sensitivity-enhanced two-dimensional 13C-19F distance NMR technique. The results indicate that AV-352 binds the C-terminal half of the TM domain, similar to the binding region of HMA. These data provide evidence for the existence of multiple oligomeric states of E in lipid bilayers, which may carry out distinct functions and may be differentially targeted by antiviral drugs.
Collapse
Affiliation(s)
- Noah H Somberg
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - Iva Sučec
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - João Medeiros-Silva
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California San Francisco, 555 Mission Bay Blvd. South, San Francisco, California 94158, United States
| | - Richard Beresis
- Department of Pharmaceutical Chemistry, University of California San Francisco, 555 Mission Bay Blvd. South, San Francisco, California 94158, United States
| | - Abdullah M Syed
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California 94158, United States
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California 94720, United States
| | - Jennifer A Doudna
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California 94158, United States
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California 94720, United States
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720, United States
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Howard Hughes Medical Institute, University of California Berkeley, Berkeley, California 94720, United States
- Department of Chemistry, University of California Berkeley, Berkeley, California 94720, United States
- California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, California 94720, United States
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, California 94158, United States
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
7
|
Li Y, Qiang R, Cao Z, Wu Q, Wang J, Lyu W. NLRP3 Inflammasomes: Dual Function in Infectious Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:407-417. [PMID: 39102612 PMCID: PMC11299487 DOI: 10.4049/jimmunol.2300745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/11/2024] [Indexed: 08/07/2024]
Abstract
The Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome has been the most distinctive polymer protein complex. After recognizing the endogenous and exogenous danger signals, NLRP3 can cause inflammation by pyroptosis and secretion of mature, bioactive forms of IL-1β and IL-18. The NLRP3 inflammasome is essential in the genesis and progression of infectious illnesses. Herein, we provide a comprehensive review of the NLRP3 inflammasome in infectious diseases, focusing on its two-sided effects. As an essential part of host defense with a protective impact, abnormal NLRP3 inflammasome activation, however, result in a systemic high inflammatory response, leading to subsequent damage. In addition, scientific evidence of small molecules, biologics, and phytochemicals acting on the NLRP3 inflammasome has been reviewed. We believe that the NLRP3 inflammasome helps us understand the pathological mechanism of different stages of infectious diseases and that inhibitors targeting the NLRP3 inflammasome will become a new and valuable research direction for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Yanbo Li
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Rui Qiang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Shunyi Hospital, Beijing, China
| | - Zhengmin Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Qingjuan Wu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Jiuchong Wang
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Wenliang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| |
Collapse
|
8
|
Dutta M, Su Y, Plescia CB, Voth GA, Stahelin RV. The SARS-CoV-2 nucleoprotein associates with anionic lipid membranes. J Biol Chem 2024; 300:107456. [PMID: 38866325 PMCID: PMC11298601 DOI: 10.1016/j.jbc.2024.107456] [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: 09/27/2023] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/14/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a lipid-enveloped virus that acquires its lipid bilayer from the host cell it infects. SARS-CoV-2 can spread from cell to cell or from patient to patient by undergoing assembly and budding to form new virions. The assembly and budding of SARS-CoV-2 is mediated by several structural proteins known as envelope (E), membrane (M), nucleoprotein (N), and spike (S), which can form virus-like particles (VLPs) when co-expressed in mammalian cells. Assembly and budding of SARS-CoV-2 from the host ER-Golgi intermediate compartment is a critical step in the virus acquiring its lipid bilayer. To date, little information is available on how SARS-CoV-2 assembles and forms new viral particles from host membranes. In this study, we used several lipid binding assays and found the N protein can strongly associate with anionic lipids including phosphoinositides and phosphatidylserine. Moreover, we show lipid binding occurs in the N protein C-terminal domain, which is supported by extensive in silico analysis. We demonstrate anionic lipid binding occurs for both the free and the N oligomeric forms, suggesting N can associate with membranes in the nucleocapsid form. Based on these results, we present a lipid-dependent model based on in vitro, cellular, and in silico data for the recruitment of N to assembly sites in the lifecycle of SARS-CoV-2.
Collapse
Affiliation(s)
- Mandira Dutta
- Department of Chemistry, The University of Chicago, Chicago, Illinois, USA
| | - Yuan Su
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Caroline B Plescia
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Gregory A Voth
- Department of Chemistry, The University of Chicago, Chicago, Illinois, USA; Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Frank Institute, The University of Chicago, Chicago, Illinois, USA.
| | - Robert V Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
9
|
Mariappan V, Adla D, Jangili S, Ranganadin P, Green SR, Mohammed S, Mutheneni SR, Pillai AB. Understanding COVID-19 outcome: Exploring the prognostic value of soluble biomarkers indicative of endothelial impairment. Cytokine 2024; 180:156673. [PMID: 38857562 DOI: 10.1016/j.cyto.2024.156673] [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: 02/03/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Host proteins released by the activated endothelial cells during SARS-CoV-2 infection are implicated to be involved in coagulation and endothelial dysfunction. However, the underlying mechanism that governs the vascular dysfunction and disease severity in COVID-19 remains obscure. The study evaluated the serum levels of Bradykinin, Kallikrein, SERPIN A, and IL-18 in COVID-19 (N-42 with 20 moderate and 22 severe) patients compared to healthy controls (HC: N-10) using ELISA at the day of admission (DOA) and day 7 post-admission. The efficacy of the protein levels in predicting disease severity was further determined using machine learning models. The levels of bradykinins and SERPIN A were higher (P ≤ 0.001) in both severe and moderate cases on day 7 post-admission compared to DOA. All the soluble proteins studied were found to elevated (P ≤ 0.01) in severe compared to moderate in day 7 and were positively correlated (P ≤ 0.001) with D-dimer, a marker for coagulation. ROC analysis identified that SERPIN A, IL-18, and bradykinin could predict the clinical condition of COVID-19 with AUC values of 1, 0.979, and 1, respectively. Among the models trained using univariate model analysis, SERPIN A emerged as a strong prognostic biomarker for COVID-19 disease severity. The serum levels of SERPIN A in conjunction with the coagulation marker D-dimer, serve as a predictive indicator for COVID-19 clinical outcomes. However, studies are required to ascertain the role of these markers in disease virulence.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Deepthi Adla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Shraddha Jangili
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Pajanivel Ranganadin
- Department of Pulmonary Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Siva Ranaganthan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Salma Mohammed
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Srinivasa Rao Mutheneni
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| |
Collapse
|
10
|
Roohi A, Gharagozlou S. Vitamin D supplementation and calcium: Many-faced gods or nobody in fighting against Corona Virus Disease 2019. Clin Nutr ESPEN 2024; 62:172-184. [PMID: 38901939 DOI: 10.1016/j.clnesp.2024.05.015] [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: 11/15/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
In December 2019, Corona Virus Disease 2019 (COVID-19) was first identified and designated as a pandemic in March 2020 due to rapid spread of the virus globally. At the beginning of the pandemic, only a few treatment options, mainly focused on supportive care and repurposing medications, were available. Due to its effects on immune system, vitamin D was a topic of interest during the pandemic, and researchers investigated its potential impact on COVID-19 outcomes. However, the results of studies about the impact of vitamin D on the disease are inconclusive. In the present narrative review, different roles of vitamin D regarding the COVID-19 have been discussed to show that vitamin D supplementation should be recommended carefully.
Collapse
Affiliation(s)
- Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
11
|
Kononova PA, Selyutina OY, Fomenko VV, Salakhutdinov NF, Polyakov NE. The mutual lipid-mediated effect of the transmembrane domain of SARS-CoV-2 E-protein and glycyrrhizin nicotinate derivatives on the localization in the lipid bilayer. Arch Biochem Biophys 2024; 758:110080. [PMID: 38960345 DOI: 10.1016/j.abb.2024.110080] [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: 03/13/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Glycyrrhizinic acid (GA) is one of the active substances in licorice root. It exhibits antiviral activity against various enveloped viruses, for example, SARS-CoV-2. GA derivatives are promising biologically active compounds from perspective of developing broad-spectrum antiviral agents. Given that GA nicotinate derivatives (Glycyvir) demonstrate activity against various DNA- and RNA-viruses, a search for a possible mechanism of action of these compounds is required. In the present paper, the interaction of Glycyvir with the transmembrane domain of the SARS-CoV-2 E-protein (ETM) in a model lipid membrane was investigated by NMR spectroscopy and molecular dynamics simulation. The lipid-mediated influence on localization of the SARS-CoV-2 E-protein by Glycyvir was observed. The presence of Glycyvir leads to deeper immersion of the ETM in lipid bilayer. Taking into account that E-protein plays a significant role in virus production and takes part in virion assembly and budding, the data on the effect of potential antiviral agents on ETM localization and structure in the lipid environment may provide a basis for further studies of potential coronavirus E-protein inhibitors.
Collapse
Affiliation(s)
- Polina A Kononova
- V. V. Voevodsky Institute of Chemical Kinetics and Combustion, 3 Institutskaya St., 630090, Novosibirsk, Russia
| | - Olga Yu Selyutina
- V. V. Voevodsky Institute of Chemical Kinetics and Combustion, 3 Institutskaya St., 630090, Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, 18 Kutateladze St., 630128, Novosibirsk, Russia.
| | - Vladislav V Fomenko
- V. V. Voevodsky Institute of Chemical Kinetics and Combustion, 3 Institutskaya St., 630090, Novosibirsk, Russia; N. N. Vorozhtsov Institute of Organic Chemistry, 9 Lavrentiev Ave, 630090, Novosibirsk, Russia
| | - Nariman F Salakhutdinov
- N. N. Vorozhtsov Institute of Organic Chemistry, 9 Lavrentiev Ave, 630090, Novosibirsk, Russia
| | - Nikolay E Polyakov
- V. V. Voevodsky Institute of Chemical Kinetics and Combustion, 3 Institutskaya St., 630090, Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, 18 Kutateladze St., 630128, Novosibirsk, Russia
| |
Collapse
|
12
|
Máthé D, Szalay G, Cseri L, Kis Z, Pályi B, Földes G, Kovács N, Fülöp A, Szepesi Á, Hajdrik P, Csomos A, Zsembery Á, Kádár K, Katona G, Mucsi Z, Rózsa BJ, Kovács E. Monitoring correlates of SARS-CoV-2 infection in cell culture using a two-photon-active calcium-sensitive dye. Cell Mol Biol Lett 2024; 29:105. [PMID: 39030477 PMCID: PMC11264913 DOI: 10.1186/s11658-024-00619-0] [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: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The organism-wide effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral infection are well studied, but little is known about the dynamics of how the infection spreads in time among or within cells due to the scarcity of suitable high-resolution experimental systems. It has been reported that SARS-CoV-2 infection pathways converge at calcium influx and subcellular calcium distribution changes. Imaging combined with a proper staining technique is an effective tool for studying subcellular calcium-related infection and replication mechanisms at such resolutions. METHODS Using two-photon (2P) fluorescence imaging with our novel Ca-selective dye, automated image analysis and clustering analysis were applied to reveal titer and variant effects on SARS-CoV-2-infected Vero E6 cells. RESULTS The application of a new calcium sensor molecule is shown, combined with a high-end 2P technique for imaging and identifying the patterns associated with cellular infection damage within cells. Vero E6 cells infected with SARS-CoV-2 variants, D614G or B.1.1.7, exhibit elevated cytosolic calcium levels, allowing infection monitoring by tracking the cellular changes in calcium level by the internalized calcium sensor. The imaging provides valuable information on how the level and intracellular distribution of calcium are perturbed during the infection. Moreover, two-photon calcium sensing allowed the distinction of infections by two studied viral variants via cluster analysis of the image parameters. This approach will facilitate the study of cellular correlates of infection and their quantification depending on viral variants and viral load. CONCLUSIONS We propose a new two-photon microscopy-based method combined with a cell-internalized sensor to quantify the level of SARS-CoV-2 infection. We optimized the applied dye concentrations to not interfere with viral fusion and viral replication events. The presented method ensured the proper monitoring of viral infection, replication, and cell fate. It also enabled distinguishing intracellular details of cell damage, such as vacuole and apoptotic body formation. Using clustering analysis, 2P microscopy calcium fluorescence images were suitable to distinguish two different viral variants in cell cultures. Cellular harm levels read out by calcium imaging were quantitatively related to the initial viral multiplicity of infection numbers. Thus, 2P quantitative calcium imaging might be used as a correlate of infection or a correlate of activity in cellular antiviral studies.
Collapse
Affiliation(s)
- Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
- HUN-REN Physical Virology Research Group, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
| | - Gergely Szalay
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
| | - Levente Cseri
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
| | - Zoltán Kis
- National Center for Public Health, Albert Flórián út 2-6, 1097, Budapest, Hungary
| | - Bernadett Pályi
- National Center for Public Health, Albert Flórián út 2-6, 1097, Budapest, Hungary
| | - Gábor Földes
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Heart and Vascular Center, Semmelweis University, Városmajor utca. 68, 1122, Budapest, Hungary
| | - Noémi Kovács
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Tűzoltó utca 37-47, 1094, Budapest, Hungary
| | - Anna Fülöp
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
| | - Áron Szepesi
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
| | - Polett Hajdrik
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary
| | - Attila Csomos
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117, Budapest, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Kristóf Kádár
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Gergely Katona
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary
| | - Zoltán Mucsi
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary.
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary.
- Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Egyetem tér 1, 3515, Miskolc, Hungary.
| | - Balázs József Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary.
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary.
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary.
| | - Ervin Kovács
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary.
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok körútja 2, 1117, Budapest, Hungary.
| |
Collapse
|
13
|
de Antonellis P, Ferrucci V, Miceli M, Bibbo F, Asadzadeh F, Gorini F, Mattivi A, Boccia A, Russo R, Andolfo I, Lasorsa VA, Cantalupo S, Fusco G, Viscardi M, Brandi S, Cerino P, Monaco V, Choi DR, Cheong JH, Iolascon A, Amente S, Monti M, Fava LL, Capasso M, Kim HY, Zollo M. Targeting ATP2B1 impairs PI3K/Akt/FOXO signaling and reduces SARS-COV-2 infection and replication. EMBO Rep 2024; 25:2974-3007. [PMID: 38816514 PMCID: PMC11239940 DOI: 10.1038/s44319-024-00164-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: 09/01/2022] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024] Open
Abstract
ATP2B1 is a known regulator of calcium (Ca2+) cellular export and homeostasis. Diminished levels of intracellular Ca2+ content have been suggested to impair SARS-CoV-2 replication. Here, we demonstrate that a nontoxic caloxin-derivative compound (PI-7) reduces intracellular Ca2+ levels and impairs SARS-CoV-2 infection. Furthermore, a rare homozygous intronic variant of ATP2B1 is shown to be associated with the severity of COVID-19. The mechanism of action during SARS-CoV-2 infection involves the PI3K/Akt signaling pathway activation, inactivation of FOXO3 transcription factor function, and subsequent transcriptional inhibition of the membrane and reticulum Ca2+ pumps ATP2B1 and ATP2A1, respectively. The pharmacological action of compound PI-7 on sustaining both ATP2B1 and ATP2A1 expression reduces the intracellular cytoplasmic Ca2+ pool and thus negatively influences SARS-CoV-2 replication and propagation. As compound PI-7 lacks toxicity in vitro, its prophylactic use as a therapeutic agent against COVID-19 is envisioned here.
Collapse
Affiliation(s)
- Pasqualino de Antonellis
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Veronica Ferrucci
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
| | - Francesca Bibbo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Fatemeh Asadzadeh
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- European School of Molecular Medicine, SEMM, Naples, Italy
| | - Francesca Gorini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Alessia Mattivi
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | | | - Roberta Russo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Immacolata Andolfo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | | | | | - Giovanna Fusco
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Maurizio Viscardi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Sergio Brandi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Vittoria Monaco
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Department of Chemical Sciences, University 'Federico II' University of Naples, Naples, 80125, Italy
| | - Dong-Rac Choi
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Elysiumbio Inc., #2007, Samsung Cheil B/D, 309, Teheran-ro, Gangnam-gu, Seoul, 06151, Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Achille Iolascon
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Stefano Amente
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Maria Monti
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Department of Chemical Sciences, University 'Federico II' University of Naples, Naples, 80125, Italy
| | - Luca L Fava
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Mario Capasso
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Hong-Yeoul Kim
- Elysiumbio Inc., #2007, Samsung Cheil B/D, 309, Teheran-ro, Gangnam-gu, Seoul, 06151, Korea
| | - Massimo Zollo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy.
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy.
- European School of Molecular Medicine, SEMM, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, 'Federico II' University of Naples, 80131, Naples, Italy.
| |
Collapse
|
14
|
Li S, Wang J, Dai X, Li C, Li T, Chen L. The PDZ domain of the E protein in SARS-CoV induces carcinogenesis and poor prognosis in LUAD. Microbes Infect 2024:105381. [PMID: 38914369 DOI: 10.1016/j.micinf.2024.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND In both lung adenocarcinoma (LUAD) and severe acute respiratory syndrome (SARS), uncontrolled inflammation can be detected in lung tissue. The PDZ-binding motif (PBM) in the SARS-CoV-1 E protein has been demonstrated to be a virulence factor that induces a cytokine storm. METHODS To identify gene expression fluctuations induced by PBM, microarray sequencing data of lung tissue infected with wild-type (SARS-CoV-1-E-wt) or recombinant virus (SARS-CoV-1-E-mutPBM) were analyzed, followed by functional enrichment analysis. To understand the role of the screened genes in LUAD, overall survival and immune correlation were calculated. RESULTS A total of 12 genes might participate in the initial and developmental stages of LUAD through expression variation and mutation. Moreover, dysregulation of a total of 12 genes could lead to a poorer prognosis. In addition, the downregulation of MAMDC2 and ITGA8 by PBM could also affect patient prognosis. Although the conserved PBM (-D-L-L-V-) can be found at the end of the carboxyl terminus in multiple E proteins of coronaviruses, the specific function of each protein depends on the entire amino acid sequence. CONCLUSIONS In summary, PBM containing the SARS-CoV-1 E protein promoted the carcinogenesis of LUAD by dysregulating important gene expression profiles and subsequently influencing the immune response and overall prognosis.
Collapse
Affiliation(s)
- Shun Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China; Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Jinxuan Wang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Xiaozhen Dai
- School of Biosciences and Technology, Chengdu Medical College, Chengdu 610500, China
| | - Churong Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Tao Li
- Radiotherapy Center, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China; Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China.
| |
Collapse
|
15
|
Hoenigsperger H, Sivarajan R, Sparrer KM. Differences and similarities between innate immune evasion strategies of human coronaviruses. Curr Opin Microbiol 2024; 79:102466. [PMID: 38555743 DOI: 10.1016/j.mib.2024.102466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
So far, seven coronaviruses have emerged in humans. Four recurring endemic coronaviruses cause mild respiratory symptoms. Infections with epidemic Middle East respiratory syndrome-related coronavirus or severe acute respiratory syndrome coronavirus (SARS-CoV)-1 are associated with high mortality rates. SARS-CoV-2 is the causative agent of the coronavirus disease 2019 pandemic. To establish an infection, coronaviruses evade restriction by human innate immune defenses, such as the interferon system, autophagy and the inflammasome. Here, we review similar and distinct innate immune manipulation strategies employed by the seven human coronaviruses. We further discuss the impact on pathogenesis, zoonotic emergence and adaptation. Understanding the nature of the interplay between endemic/epidemic/pandemic coronaviruses and host defenses may help to better assess the pandemic potential of emerging coronaviruses.
Collapse
Affiliation(s)
- Helene Hoenigsperger
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rinu Sivarajan
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | | |
Collapse
|
16
|
Qu Y, Wang S, Jiang H, Wang Q, Liao Y, Qiu X, Tan L, Song C, Ding C, Sun Y, Yang Z. The Ca 2+-dependent phosphatase calcineurin dephosphorylates TBK1 to suppress antiviral innate immunity. J Virol 2024; 98:e0001624. [PMID: 38563732 PMCID: PMC11092360 DOI: 10.1128/jvi.00016-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Tumor necrosis factor receptor-associated factor family member-associated NF-κB activator-binding kinase 1 (TBK1) plays a key role in the induction of the type 1 interferon (IFN-I) response, which is an important component of innate antiviral defense. Viruses target calcium (Ca2+) signaling networks, which participate in the regulation of the viral life cycle, as well as mediate the host antiviral response. Although many studies have focused on the role of Ca2+ signaling in the regulation of IFN-I, the relationship between Ca2+ and TBK1 in different infection models requires further elucidation. Here, we examined the effects of the Newcastle disease virus (NDV)-induced increase in intracellular Ca2+ levels on the suppression of host antiviral responses. We demonstrated that intracellular Ca2+ increased significantly during NDV infection, leading to impaired IFN-I production and antiviral immunity through the activation of calcineurin (CaN). Depletion of Ca²+ was found to lead to a significant increase in virus-induced IFN-I production resulting in the inhibition of viral replication. Mechanistically, the accumulation of Ca2+ in response to viral infection increases the phosphatase activity of CaN, which in turn dephosphorylates and inactivates TBK1 in a Ca2+-dependent manner. Furthermore, the inhibition of CaN on viral replication was counteracted in TBK1 knockout cells. Together, our data demonstrate that NDV hijacks Ca2+ signaling networks to negatively regulate innate immunity via the CaN-TBK1 signaling axis. Thus, our findings not only identify the mechanism by which viruses exploit Ca2+ signaling to evade the host antiviral response but also, more importantly, highlight the potential role of Ca2+ homeostasis in the viral innate immune response.IMPORTANCEViral infections disrupt intracellular Ca2+ homeostasis, which affects the regulation of various host processes to create conditions that are conducive for their own proliferation, including the host immune response. The mechanism by which viruses trigger TBK1 activation and IFN-I induction through viral pathogen-associated molecular patterns has been well defined. However, the effects of virus-mediated Ca2+ imbalance on the IFN-I pathway requires further elucidation, especially with respect to TBK1 activation. Herein, we report that NDV infection causes an increase in intracellular free Ca2+ that leads to activation of the serine/threonine phosphatase CaN, which subsequently dephosphorylates TBK1 and negatively regulates IFN-I production. Furthermore, depletion of Ca2+ or inhibition of CaN activity exerts antiviral effects by promoting the production of IFN-I and inhibiting viral replication. Thus, our results reveal the potential role of Ca2+ in the innate immune response to viruses and provide a theoretical reference for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Yang Qu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Siyuan Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hui Jiang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Qingyi Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
17
|
Ambrożek-Latecka M, Kozlowski P, Hoser G, Bandyszewska M, Hanusek K, Nowis D, Gołąb J, Grzanka M, Piekiełko-Witkowska A, Schulz L, Hornung F, Deinhardt-Emmer S, Kozlowska E, Skirecki T. SARS-CoV-2 and its ORF3a, E and M viroporins activate inflammasome in human macrophages and induce of IL-1α in pulmonary epithelial and endothelial cells. Cell Death Discov 2024; 10:191. [PMID: 38664396 PMCID: PMC11045860 DOI: 10.1038/s41420-024-01966-9] [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: 02/23/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammasome assembly is a potent mechanism responsible for the host protection against pathogens, including viruses. When compromised, it can allow viral replication, while when disrupted, it can perpetuate pathological responses by IL-1 signaling and pyroptotic cell death. SARS-CoV-2 infection was shown to activate inflammasome in the lungs of COVID-19 patients, however, potential mechanisms responsible for this response are not fully elucidated. In this study, we investigated the effects of ORF3a, E and M SARS-CoV-2 viroporins in the inflammasome activation in major populations of alveolar sentinel cells: macrophages, epithelial and endothelial cells. We demonstrated that each viroporin is capable of activation of the inflammasome in macrophages to trigger pyroptosis-like cell death and IL-1α release from epithelial and endothelial cells. Small molecule NLRP3 inflammasome inhibitors reduced IL-1 release but weakly affected the pyroptosis. Importantly, we discovered that while SARS-CoV-2 could not infect the pulmonary microvascular endothelial cells it induced IL-1α and IL-33 release. Together, these findings highlight the essential role of macrophages as the major inflammasome-activating cell population in the lungs and point to endothelial cell expressed IL-1α as a potential novel component driving the pulmonary immunothromobosis in COVID-19.
Collapse
Affiliation(s)
- Magdalena Ambrożek-Latecka
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Piotr Kozlowski
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Magdalena Bandyszewska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Hanusek
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Faculty of Medicine, Medial University of Warsaw, Warsaw, Poland
| | - Jakub Gołąb
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Piekiełko-Witkowska
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Luise Schulz
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Franziska Hornung
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | | | - Ewa Kozlowska
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
18
|
Steiner S, Kratzel A, Barut GT, Lang RM, Aguiar Moreira E, Thomann L, Kelly JN, Thiel V. SARS-CoV-2 biology and host interactions. Nat Rev Microbiol 2024; 22:206-225. [PMID: 38225365 DOI: 10.1038/s41579-023-01003-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/17/2024]
Abstract
The zoonotic emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease 2019 (COVID-19) pandemic have profoundly affected our society. The rapid spread and continuous evolution of new SARS-CoV-2 variants continue to threaten global public health. Recent scientific advances have dissected many of the molecular and cellular mechanisms involved in coronavirus infections, and large-scale screens have uncovered novel host-cell factors that are vitally important for the virus life cycle. In this Review, we provide an updated summary of the SARS-CoV-2 life cycle, gene function and virus-host interactions, including recent landmark findings on general aspects of coronavirus biology and newly discovered host factors necessary for virus replication.
Collapse
Affiliation(s)
- Silvio Steiner
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Annika Kratzel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G Tuba Barut
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto M Lang
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Etori Aguiar Moreira
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Lisa Thomann
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jenna N Kelly
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
- European Virus Bioinformatics Center, Jena, Germany
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland.
- European Virus Bioinformatics Center, Jena, Germany.
| |
Collapse
|
19
|
Sučec I, Pankratova Y, Parasar M, Hong M. Transmembrane conformation of the envelope protein of an alpha coronavirus, NL63. Protein Sci 2024; 33:e4923. [PMID: 38501465 PMCID: PMC10949323 DOI: 10.1002/pro.4923] [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: 12/28/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 03/20/2024]
Abstract
The envelope (E) proteins of coronaviruses (CoVs) form cation-conducting channels that are associated with the pathogenicity of these viruses. To date, high-resolution structural information about these viroporins is limited to the SARS-CoV E protein. To broaden our structural knowledge of other members of this family of viroporins, we now investigate the conformation of the E protein of the human coronavirus (hCoV), NL63. Using two- and three-dimensional magic-angle-spinning NMR, we have measured 13 C and 15 N chemical shifts of the transmembrane domain of E (ETM), which yielded backbone (ϕ, ψ) torsion angles. We further measured the water accessibility of NL63 ETM at neutral pH versus acidic pH in the presence of Ca2+ ions. These data show that NL63 ETM adopts a regular α-helical conformation that is unaffected by pH and the N-terminal ectodomain. Interestingly, the water accessibility of NL63 ETM increases only modestly at acidic pH in the presence of Ca2+ compared to neutral pH, in contrast to SARS ETM, which becomes much more hydrated at acidic pH. This difference suggests a structural basis for the weaker channel conductance of α-CoV compared to β-CoV E proteins. The weaker E channel activity may in turn contribute to the reduced virulence of hCoV-NL63 compared to SARS-CoV viruses.
Collapse
Affiliation(s)
- Iva Sučec
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yanina Pankratova
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Mriganka Parasar
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Mei Hong
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
20
|
Wallace HL, Russell RS. Inflammatory Consequences: Hepatitis C Virus-Induced Inflammasome Activation and Pyroptosis. Viral Immunol 2024; 37:126-138. [PMID: 38593460 DOI: 10.1089/vim.2023.0138] [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] [Indexed: 04/11/2024] Open
Abstract
Hepatitis C virus (HCV), despite the availability of effective direct-acting antivirals (DAAs) that clear the virus from >95% of individuals treated, continues to cause significant health care burden due to disease progression that can lead to fibrosis, cirrhosis, and/or hepatocellular carcinoma. The fact that some people who are treated with DAAs still go on to develop worsening liver disease warrants further study into the immunopathogenesis of HCV. Many viral infections, including HCV, have been associated with activation of the inflammasome/pyroptosis pathway. This inflammatory cell death pathway ultimately results in cell lysis and release of inflammatory cytokines, IL-18 and IL-1β. This review will report on studies that investigated HCV and inflammasome activation/pyroptosis. This includes clinical in vivo data showing elevated pyroptosis-associated cytokines in the blood of individuals living with HCV, studies of genetic associations of pyroptosis-related genes and development of liver disease, and in vitro studies aimed at understanding the mechanism of pyroptosis induced by HCV. Finally, we discuss major gaps in understanding and outstanding questions that remain in the field of HCV-induced pyroptosis.
Collapse
Affiliation(s)
- Hannah L Wallace
- Immunology and Infectious Diseases Group, Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St John's, Canada
| | - Rodney S Russell
- Immunology and Infectious Diseases Group, Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St John's, Canada
| |
Collapse
|
21
|
Cubisino SAM, Milenkovic S, Conti-Nibali S, Musso N, Bonacci P, De Pinto V, Ceccarelli M, Reina S. Electrophysiological properties and structural prediction of the SARS-CoV-2 viroprotein E. Front Mol Biosci 2024; 11:1334819. [PMID: 38606285 PMCID: PMC11007222 DOI: 10.3389/fmolb.2024.1334819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/15/2024] [Indexed: 04/13/2024] Open
Abstract
COVID-19, the infectious disease caused by the most recently discovered coronavirus SARS- CoV-2, has caused millions of sick people and thousands of deaths all over the world. The viral positive-sense single-stranded RNA encodes 31 proteins among which the spike (S) is undoubtedly the best known. Recently, protein E has been reputed as a potential pharmacological target as well. It is essential for the assembly and release of the virions in the cell. Literature describes protein E as a voltage-dependent channel with preference towards monovalent cations whose intracellular expression, though, alters Ca2+ homeostasis and promotes the activation of the proinflammatory cascades. Due to the extremely high sequence identity of SARS-CoV-2 protein E (E-2) with the previously characterized E-1 (i.e., protein E from SARS-CoV) many data obtained for E-1 were simply adapted to the other. Recent solid state NMR structure revealed that the transmembrane domain (TMD) of E-2 self-assembles into a homo-pentamer, albeit the oligomeric status has not been validated with the full-length protein. Prompted by the lack of a common agreement on the proper structural and functional features of E-2, we investigated the specific mechanism/s of pore-gating and the detailed molecular structure of the most cryptic protein of SARS-CoV-2 by means of MD simulations of the E-2 structure and by expressing, refolding and analyzing the electrophysiological activity of the transmembrane moiety of the protein E-2, in its full length. Our results show a clear agreement between experimental and predictive studies and foresee a mechanism of activity based on Ca2+ affinity.
Collapse
Affiliation(s)
| | | | - Stefano Conti-Nibali
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paolo Bonacci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Vito De Pinto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- We.MitoBiotech S.R.L, Catania, Italy
| | | | - Simona Reina
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- We.MitoBiotech S.R.L, Catania, Italy
| |
Collapse
|
22
|
Xu JB, Guan WJ, Zhang YL, Qiu ZE, Chen L, Hou XC, Yue J, Zhou YY, Sheng J, Zhao L, Zhu YX, Sun J, Zhao J, Zhou WL, Zhong NS. SARS-CoV-2 envelope protein impairs airway epithelial barrier function and exacerbates airway inflammation via increased intracellular Cl - concentration. Signal Transduct Target Ther 2024; 9:74. [PMID: 38528022 DOI: 10.1038/s41392-024-01753-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/02/2024] [Accepted: 01/19/2024] [Indexed: 03/27/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection disrupts the epithelial barrier and triggers airway inflammation. The envelope (E) protein, a core virulence structural component of coronaviruses, may play a role in this process. Pathogens could interfere with transepithelial Cl- transport via impairment of the cystic fibrosis transmembrane conductance regulator (CFTR), which modulates nuclear factor κB (NF-κB) signaling. However, the pathological effects of SARS-CoV-2 E protein on airway epithelial barrier function, Cl- transport and the robust inflammatory response remain to be elucidated. Here, we have demonstrated that E protein down-regulated the expression of tight junctional proteins, leading to the disruption of the airway epithelial barrier. In addition, E protein triggered the activation of Toll-like receptor (TLR) 2/4 and downstream c-Jun N-terminal kinase (JNK) signaling, resulting in an increased intracellular Cl- concentration ([Cl-]i) via up-regulating phosphodiesterase 4D (PDE4D) expression in airway epithelial cells. This elevated [Cl-]i contributed to the heightened airway inflammation through promoting the phosphorylation of serum/glucocorticoid regulated kinase 1 (SGK1). Moreover, blockade of SGK1 or PDE4 alleviated the robust inflammatory response induced by E protein. Overall, these findings provide novel insights into the pathogenic role of SARS-CoV-2 E protein in airway epithelial damage and the ongoing airway inflammation during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jian-Bang Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China.
- Department of Thoracic Surgery, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China.
- Guangzhou National Laboratory, Guangzhou, P. R. China.
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Junqing Yue
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China
- Guangzhou National Laboratory, Guangzhou, P. R. China
| | - Yu-Yun Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jie Sheng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Lei Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, P. R. China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China
- Guangzhou National Laboratory, Guangzhou, P. R. China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China
- Guangzhou National Laboratory, Guangzhou, P. R. China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China.
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China.
- Guangzhou National Laboratory, Guangzhou, P. R. China.
| |
Collapse
|
23
|
Huang W, Chen X, Yin M, Li J, Luo M, Ai Y, Xie L, Li W, Liu Y, Xie X, Chen Y, Zhang X, He J. Protection effects of mice liver and lung injury induced by coronavirus infection of Qingfei Paidu decoction involve inhibition of the NLRP3 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117512. [PMID: 38040130 DOI: 10.1016/j.jep.2023.117512] [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: 07/30/2023] [Revised: 11/12/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coronavirus Disease 2019 (COVID-19) is a grave and pervasive global infectious malady brought about by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), posing a significant menace to human well-being. Qingfei Paidu decoction (QFPD) represents a pioneering formulation derived from four classical Chinese medicine prescriptions. Substantiated evidence attests to its efficacy in alleviating clinical manifestations, mitigating the incidence of severe and critical conditions, and reducing mortality rates among COVID-19 patients. AIM OF THE STUDY This study aims to investigate the protection effects of QFPD in mice afflicted with a coronavirus infection, with a particular focus on determining whether its mechanism involves the NLRP3 signaling pathway. MATERIALS AND METHODS The coronavirus mice model was established through intranasal infection of Kunming mice with Hepatic Mouse Virus A59 (MHV-A59). In the dose-effect experiment, normal saline, ribavirin (80 mg/kg), or QFPD (5, 10, 20 g/kg) were administered to the mice 2 h following MHV-A59 infection. In the time-effect experiment, normal saline or QFPD (20 g/kg) was administered to mice 2 h post MHV-A59 infection. Following the assessment of mouse body weights, food consumption, and water intake, intragastric administration was conducted once daily at consistent intervals over a span of 5 days. The impact of QFPD on pathological alterations in the livers and lungs of MHV-A59-infected mice was evaluated through H&E staining. The viral loads of MHV-A59 in both the liver and lung were determined using qPCR. The expression levels of genes and proteins related to the NLRP3 pathway in the liver and lung were assessed through qPCR, Western Blot analysis, and immunofluorescence. RESULTS The administration of QFPD was shown to ameliorate the reduced weight gain, decline in food consumption, and diminished water intake, all of which were repercussions of MHV-A59 infection in mice. QFPD treatment exhibited notable efficacy in safeguarding tissue integrity. The extent of hepatic and pulmonary injury, when coupled with QFPD treatment, demonstrated not only a reduction with higher treatment dosages but also a decline with prolonged treatment duration. In the dose-effect experiment, there was a notable, dose-dependent reduction in the viral loads, as well as the expression levels of IL-1β, NLRP3, ASC, Caspase 1, Caspase-1 p20, GSDMD, GSDMD-N, and NF-κB within the liver of the QFPD-treated groups. Additionally, in the time-effects experiments, the viral loads and the expression levels of genes and proteins linked to the NLRP3 pathway were consistently lower in the QFPD-treated groups compared with the model control groups, particularly during the periods when their expressions reached their zenith in the model group. Notably, IL-18 showed only a modest elevation relative to the blank control group following QFPD treatment. CONCLUSIONS To sum up, our current study demonstrated that QFPD treatment has the capacity to alleviate infection-related symptoms, mitigate tissue damage in infected organs, and suppress viral replication in coronavirus-infected mice. The protective attributes of QFPD in coronavirus-infected mice are plausibly associated with its modulation of the NLRP3 signaling pathway. We further infer that QFPD holds substantial promise in the context of coronavirus infection therapy.
Collapse
Affiliation(s)
- Wenguan Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiuyun Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mingyu Yin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junlin Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Minyi Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Ai
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wanxi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yatian Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinyuan Xie
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuan Chen
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinyu Zhang
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation. Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinyang He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Manfrini N, Notarbartolo S, Grifantini R, Pesce E. SARS-CoV-2: A Glance at the Innate Immune Response Elicited by Infection and Vaccination. Antibodies (Basel) 2024; 13:13. [PMID: 38390874 PMCID: PMC10885122 DOI: 10.3390/antib13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has led to almost seven million deaths worldwide. SARS-CoV-2 causes infection through respiratory transmission and can occur either without any symptoms or with clinical manifestations which can be mild, severe or, in some cases, even fatal. Innate immunity provides the initial defense against the virus by sensing pathogen-associated molecular patterns and triggering signaling pathways that activate the antiviral and inflammatory responses, which limit viral replication and help the identification and removal of infected cells. However, temporally dysregulated and excessive activation of the innate immune response is deleterious for the host and associates with severe COVID-19. In addition to its defensive role, innate immunity is pivotal in priming the adaptive immune response and polarizing its effector function. This capacity is relevant in the context of both SARS-CoV-2 natural infection and COVID-19 vaccination. Here, we provide an overview of the current knowledge of the innate immune responses to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Nicola Manfrini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Samuele Notarbartolo
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- CheckmAb Srl, 20122 Milan, Italy
| | - Elisa Pesce
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
25
|
Wölk C, Shen C, Hause G, Surya W, Torres J, Harvey RD, Bello G. Membrane Condensation and Curvature Induced by SARS-CoV-2 Envelope Protein. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2646-2655. [PMID: 38258382 PMCID: PMC10851660 DOI: 10.1021/acs.langmuir.3c03079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
The envelope (E) protein of SARS-CoV-2 participates in virion encapsulation and budding at the membrane of the endoplasmic reticulum Golgi intermediate compartment (ERGIC). The positively curved membrane topology required to fit an 80 nm viral particle is energetically unfavorable; therefore, viral proteins must facilitate ERGIC membrane curvature alteration. To study the possible role of the E protein in this mechanism, we examined the structural modification of the host lipid membrane by the SARS-CoV-2 E protein using synchrotron-based X-ray methods. Our reflectometry results on solid-supported planar bilayers show that E protein markedly condenses the surrounding lipid bilayer. For vesicles, this condensation effect differs between the two leaflets such that the membrane becomes asymmetric and increases its curvature. The formation of such a curved and condensed membrane is consistent with the requirements to stably encapsulate a viral core and supports a role for E protein in budding during SARS-CoV-2 virion assembly.
Collapse
Affiliation(s)
- Christian Wölk
- Pharmaceutical
Technology, Medical Faculty, University
Leipzig, Eilenburger
Straße 15a, 04317 Leipzig, Germany
| | - Chen Shen
- Deutsches
Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Gerd Hause
- Biocenter, Martin-Luther University Halle-Wittenberg, Weinbergweg 22, 06120 Halle (Saale), Germany
| | - Wahyu Surya
- School
of Biological Sciences, Nanyang Technological
University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jaume Torres
- School
of Biological Sciences, Nanyang Technological
University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Richard D. Harvey
- Division
of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, UZA 2, Vienna 1090, Austria
| | - Gianluca Bello
- Division
of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, UZA 2, Vienna 1090, Austria
| |
Collapse
|
26
|
Chiarini A, Armato U, Gui L, Dal Prà I. "Other Than NLRP3" Inflammasomes: Multiple Roles in Brain Disease. Neuroscientist 2024; 30:23-48. [PMID: 35815856 DOI: 10.1177/10738584221106114] [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] [Indexed: 11/15/2022]
Abstract
Human neuroinflammatory and neurodegenerative diseases, whose prevalence keeps rising, are still unsolved pathobiological/therapeutical problems. Among others, recent etiology hypotheses stressed as their main driver a chronic neuroinflammation, which is mediated by innate immunity-related protein oligomers: the inflammasomes. A panoply of exogenous and/or endogenous harmful agents activates inflammasomes' assembly, signaling, and IL-1β/IL-18 production and neural cells' pyroptotic death. The underlying concept is that inflammasomes' chronic activation advances neurodegeneration while their short-lasting operation restores tissue homeostasis. Hence, from a therapeutic standpoint, it is crucial to understand inflammasomes' regulatory mechanisms. About this, a deluge of recent studies focused on the NLRP3 inflammasome with suggestions that its pharmacologic block would hinder neurodegeneration. Yet hitherto no evidence proves this view. Moreover, known inflammasomes are numerous, and the mechanisms regulating their expression and function may vary with the involved animal species and strains, as well as organs and cells, and the harmful factors triggered as a result. Therefore, while presently leaving out some little-studied inflammasomes, this review focuses on the "other than NLRP3" inflammasomes that participate in neuroinflammation's complex mechanisms: NLRP1, NLRP2, NLRC4, and AIM2. Although human-specific data about them are relatively scant, we stress that only a holistic view including several human brain inflammasomes and other potential pathogenetic drivers will lead to successful therapies for neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Li Gui
- Department of Neurology, Southwest Hospital, Chongqing, China
| | - Ilaria Dal Prà
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
27
|
Liu MH, Lin XL, Xiao LL. SARS-CoV-2 nucleocapsid protein promotes TMAO-induced NLRP3 inflammasome activation by SCAP-SREBP signaling pathway. Tissue Cell 2024; 86:102276. [PMID: 37979395 DOI: 10.1016/j.tice.2023.102276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 11/12/2023] [Accepted: 11/12/2023] [Indexed: 11/20/2023]
Abstract
The sterol regulatory element-binding protein (SREBP) activation and cytokine level were significantly increased in coronavirus disease-19. The NLRP3 inflammasome is an amplifier for cellular inflammation. This study aimed to elucidate the modulatory effect of SARS-CoV-2 nucleocapsid protein (SARS-CoV-2 NP) on trimethylamine N-oxide (TMAO)-induced lipogenesis and NLRP3 inflammasome activation and the underlying mechanisms in vascular smooth muscle cells (VSMCs). Our data indicated that SARS-CoV-2 NP activates the dissociation of the SREBP cleavage activating protein (SCAP) from the endoplasmic reticulum, resulting in SREBP activation, increased lipogenic gene expression, and NLRP3 inflammasome activation. TMAO was applied to VSMC-induced NLRP3 inflammasome by promoting the SCAP-SREBP complex endoplasmic reticulum-to-Golgi translocation, which facilitates directly binding of SARS-CoV-2 NP to the NLRP3 protein for NLRP3 inflammasome assembly. SARS-CoV-2 NP amplified the TMAO-induced lipogenic gene expression and NLRP3 inflammasome. Knockdown of SCAP-SREBP2 can effectively reduce lipogenic gene expression and alleviate NLRP3 inflammasome-mediated systemic inflammation in VSMCs stimulated with TMAO and SARS-CoV-2 NP. These results reveal that SARS-CoV-2 NP amplified TMAO-induced lipogenesis and NLRP3 inflammasome activation via priming the SCAP-SREBP signaling pathway.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, People's Republic of China.
| | - Xiao-Long Lin
- Department of Pathology, Hui Zhou Third People's Hospital, Guangzhou Medical University, Huizhou City, Guangdong 516002, People's Republic of China
| | - Le-Le Xiao
- Intensive Care Unit, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, People's Republic of China.
| |
Collapse
|
28
|
Zhang J, Hom K, Zhang C, Nasr M, Gerzanich V, Zhang Y, Tang Q, Xue F, Simard JM, Zhao RY. SARS-CoV-2 ORF3a Protein as a Therapeutic Target against COVID-19 and Long-Term Post-Infection Effects. Pathogens 2024; 13:75. [PMID: 38251382 PMCID: PMC10819734 DOI: 10.3390/pathogens13010075] [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: 12/19/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has posed unparalleled challenges due to its rapid transmission, ability to mutate, high mortality and morbidity, and enduring health complications. Vaccines have exhibited effectiveness, but their efficacy diminishes over time while new variants continue to emerge. Antiviral medications offer a viable alternative, but their success has been inconsistent. Therefore, there remains an ongoing need to identify innovative antiviral drugs for treating COVID-19 and its post-infection complications. The ORF3a (open reading frame 3a) protein found in SARS-CoV-2, represents a promising target for antiviral treatment due to its multifaceted role in viral pathogenesis, cytokine storms, disease severity, and mortality. ORF3a contributes significantly to viral pathogenesis by facilitating viral assembly and release, essential processes in the viral life cycle, while also suppressing the body's antiviral responses, thus aiding viral replication. ORF3a also has been implicated in triggering excessive inflammation, characterized by NF-κB-mediated cytokine production, ultimately leading to apoptotic cell death and tissue damage in the lungs, kidneys, and the central nervous system. Additionally, ORF3a triggers the activation of the NLRP3 inflammasome, inciting a cytokine storm, which is a major contributor to the severity of the disease and subsequent mortality. As with the spike protein, ORF3a also undergoes mutations, and certain mutant variants correlate with heightened disease severity in COVID-19. These mutations may influence viral replication and host cellular inflammatory responses. While establishing a direct link between ORF3a and mortality is difficult, its involvement in promoting inflammation and exacerbating disease severity likely contributes to higher mortality rates in severe COVID-19 cases. This review offers a comprehensive and detailed exploration of ORF3a's potential as an innovative antiviral drug target. Additionally, we outline potential strategies for discovering and developing ORF3a inhibitor drugs to counteract its harmful effects, alleviate tissue damage, and reduce the severity of COVID-19 and its lingering complications.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Mohamed Nasr
- Drug Development and Clinical Sciences Branch, Division of AIDS, NIAID, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA;
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA;
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
- Department of Microbiology-Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
29
|
Radwan E, Abdelaziz A, Mandour MAM, Meki ARMA, El-Kholy MM, Mohamed MN. MBOAT7 expression is associated with disease progression in COVID-19 patients. Mol Biol Rep 2024; 51:79. [PMID: 38183501 PMCID: PMC10771377 DOI: 10.1007/s11033-023-09009-9] [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: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND AND AIM The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019 caused a pandemic of acute respiratory disease, named coronavirus disease 2019 (COVID-19). COVID-19 became one of the most challenging health emergencies, hence the necessity to find different prognostic factors for disease progression, and severity. Membrane bound O-acyltransferase domain containing 7 (MBOAT7) demonstrates anti-inflammatory effects through acting as a fine-tune regulator of the amount of cellular free arachidonic acid. We aimed in this study to evaluate MBOAT7 expression in COVID-19 patients and to correlate it with disease severity and outcomes. METHODS This case-control study included 56 patients with confirmed SARS-CoV-2 diagnosis and 28 control subjects. Patients were further classified into moderate (n = 28) and severe (n = 28) cases. MBOAT7, tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) mRNA levels were evaluated in peripheral blood mononuclear cells (PBMC) samples isolated from patients and control subjects by real time quantitative polymerase chain reaction (RT-qPCR). In addition, circulating MBOAT7 protein levels were assayed by enzyme-linked immunosorbent assay (ELISA). RESULTS Significant lower levels of circulating MBOAT7 mRNA and protein were observed in COVID-19 patients compared to control subjects with severe COVID-19 cases showing significant lower levels compared to moderate cases. Moreover, severe cases showed a significant upregulation of TNF-α and IL-1ß mRNA. MBOAT7 mRNA and protein levels were significantly correlated with inflammatory markers (TNF-α, IL-1ß, C-reactive protein (CRP), and ferritin), liver enzymes, severity, and oxygen saturation levels. CONCLUSION COVID-19 is associated with downregulation of MBAOT7, which correlates with disease severity.
Collapse
Affiliation(s)
- Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt.
| | - Ahmed Abdelaziz
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt
| | - Manal A M Mandour
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Abdel-Raheim M A Meki
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt
| | - Maha M El-Kholy
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwan N Mohamed
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
30
|
Yue Z, Zhang X, Gu Y, Liu Y, Lan LM, Liu Y, Li Y, Yang G, Wan P, Chen X. Regulation and functions of the NLRP3 inflammasome in RNA virus infection. Front Cell Infect Microbiol 2024; 13:1309128. [PMID: 38249297 PMCID: PMC10796458 DOI: 10.3389/fcimb.2023.1309128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Virus infection is one of the greatest threats to human life and health. In response to viral infection, the host's innate immune system triggers an antiviral immune response mostly mediated by inflammatory processes. Among the many pathways involved, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome has received wide attention in the context of viral infection. The NLRP3 inflammasome is an intracellular sensor composed of three components, including the innate immune receptor NLRP3, adaptor apoptosis-associated speck-like protein containing CARD (ASC), and the cysteine protease caspase-1. After being assembled, the NLRP3 inflammasome can trigger caspase-1 to induce gasdermin D (GSDMD)-dependent pyroptosis, promoting the maturation and secretion of proinflammatory cytokines such as interleukin-1 (IL-1β) and interleukin-18 (IL-18). Recent studies have revealed that a variety of viruses activate or inhibit the NLRP3 inflammasome via viral particles, proteins, and nucleic acids. In this review, we present a variety of regulatory mechanisms and functions of the NLRP3 inflammasome upon RNA viral infection and demonstrate multiple therapeutic strategies that target the NLRP3 inflammasome for anti-inflammatory effects in viral infection.
Collapse
Affiliation(s)
- Zhaoyang Yue
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Xuelong Zhang
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yu Gu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ying Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Lin-Miaoshen Lan
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yilin Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yongkui Li
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xin Chen
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| |
Collapse
|
31
|
Singh Dagur H, Behmard E, Rajakumara E, Barzegari E. Identifying potent inhibitory phytocompounds from Lagerstroemia speciosa against SARS-Coronavirus-2: structure-based virtual screening. J Biomol Struct Dyn 2024; 42:806-818. [PMID: 37170794 DOI: 10.1080/07391102.2023.2205942] [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: 09/30/2022] [Accepted: 03/20/2023] [Indexed: 05/13/2023]
Abstract
The ongoing spillover of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) calls for expedited countermeasure through developing therapeutics from natural reservoirs and/or the use of less time-consuming drug discovery methodologies. This study aims to apply these approaches to identify potential blockers of the virus from the longstanding medicinal herb, Lagerstroemia speciosa, through comprehensive computational-based screening. Nineteen out of 22 L. speciosa phytochemicals were selected on the basis of their pharmacokinetic properties. SARS-CoV-2 Main protease (Mpro), RNA-directed RNA polymerase (RdRp), Envelope viroporin protein (Evp) and receptor-binding domain of Spike glycoprotein (S-RBD), as well as the human receptor Angiotensin-converting enzyme-2 (hACE2) were chosen as targets. The screening was performed by molecular docking, followed by 100-ns molecular dynamic simulations and free energy calculations. 24-Methylene cycloartanol acetate (24MCA) was found as the best inhibitor for both Evp and RdRp, and sitosterol acetate (SA) as the best hit for Mpro, S-RBD and hACE2. Dynamic simulations, binding mode analyses, free energy terms and share of key amino acids in protein-drug interactions confirmed the stable binding of these phytocompounds to the hotspot sites on the target proteins. With their possible multi-targeting capability, the introduced phytoligands might offer promising lead compounds for persistent fight with the rapidly evolving coronavirus. Therefore, experimental verification of their safety and efficacy is recommended.
Collapse
Affiliation(s)
- Hanuman Singh Dagur
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eerappa Rajakumara
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Ebrahim Barzegari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
32
|
Gao H, Wang S, Duan H, Wang Y, Zhu H. Biological analysis of the potential pathogenic mechanisms of Infectious COVID-19 and Guillain-Barré syndrome. Front Immunol 2023; 14:1290578. [PMID: 38115996 PMCID: PMC10728822 DOI: 10.3389/fimmu.2023.1290578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023] Open
Abstract
Background Guillain-Barré syndrome (GBS) is a medical condition characterized by the immune system of the body attacking the peripheral nerves, including those in the spinal nerve roots, peripheral nerves, and cranial nerves. It can cause limb weakness, abnormal sensations, and facial nerve paralysis. Some studies have reported clinical cases associated with the severe coronavirus disease 2019 (COVID-19) and GBS, but how COVID-19 affects GBS is unclear. Methods We utilized bioinformatics techniques to explore the potential genetic connection between COVID-19 and GBS. Differential expression of genes (DEGs) related to COVID-19 and GBS was collected from the Gene Expression Omnibus (GEO) database. By taking the intersection, we obtained shared DEGs for COVID-19 and GBS. Subsequently, we utilized bioinformatics analysis tools to analyze common DEGs, conducting functional enrichment analysis and constructing Protein-protein interaction networks (PPI), Transcription factors (TF) -gene networks, and TF-miRNA networks. Finally, we validated our findings by constructing the Receiver Operating Characteristic (ROC) curves. Results This study utilizes bioinformatics tools for the first time to investigate the close genetic relationship between COVID-19 and GBS. CAMP, LTF, DEFA1B, SAMD9, GBP1, DDX60, DEFA4, and OAS3 are identified as the most significant interacting genes between COVID-19 and GBS. In addition, the signaling pathway of NOD-like receptors is believed to be essential in the link between COVID-19 and GBS.
Collapse
Affiliation(s)
| | | | | | | | - Hui Zhu
- Department of Neurology, The First Teaching Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
33
|
Baral B, Saini V, Tandon A, Singh S, Rele S, Dixit AK, Parmar HS, Meena AK, Jha HC. SARS-CoV-2 envelope protein induces necroptosis and mediates inflammatory response in lung and colon cells through receptor interacting protein kinase 1. Apoptosis 2023; 28:1596-1617. [PMID: 37658919 DOI: 10.1007/s10495-023-01883-9] [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] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
SARS-CoV-2 Envelope protein (E) is one of the crucial components in virus assembly and pathogenesis. The current study investigated its role in the SARS-CoV-2-mediated cell death and inflammation in lung and gastrointestinal epithelium and its effect on the gastrointestinal-lung axis. We observed that transfection of E protein increases the lysosomal pH and induces inflammation in the cell. The study utilizing Ethidium bromide/Acridine orange and Hoechst/Propidium iodide staining demonstrated necrotic cell death in E protein transfected cells. Our study revealed the role of the necroptotic marker RIPK1 in cell death. Additionally, inhibition of RIPK1 by its specific inhibitor Nec-1s exhibits recovery from cell death and inflammation manifested by reduced phosphorylation of NFκB. The E-transfected cells' conditioned media induced inflammation with differential expression of inflammatory markers compared to direct transfection in the gastrointestinal-lung axis. In conclusion, SARS-CoV-2 E mediates inflammation and necroptosis through RIPK1, and the E-expressing cells' secretion can modulate the gastrointestinal-lung axis. Based on the data of the present study, we believe that during severe COVID-19, necroptosis is an alternate mechanism of cell death besides ferroptosis, especially when the disease is not associated with drastic increase in serum ferritin.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Akrati Tandon
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Siddharth Singh
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Samiksha Rele
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector-V, Bidhannagar, Kolkata, 700091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh, 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Amkhoh, Gwalior, Madhya Pradesh, 474001, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India.
| |
Collapse
|
34
|
Ghafouri E, Bigdeli M, Khalafiyan A, Amirkhani Z, Ghanbari R, Hasan A, Khanahmad H, Boshtam M, Makvandi P. Unmasking the complex roles of hypocalcemia in cancer, COVID-19, and sepsis: Engineered nanodelivery and diagnosis. ENVIRONMENTAL RESEARCH 2023; 238:116979. [PMID: 37660871 DOI: 10.1016/j.envres.2023.116979] [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: 07/10/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Calcium (Ca2+) homeostasis is essential for maintaining physiological processes in the body. Disruptions in Ca2+ signaling can lead to various pathological conditions including inflammation, fibrosis, impaired immune function, and accelerated senescence. Hypocalcemia, a common symptom in diseases such as acute respiratory distress syndrome (ARDS), cancer, septic shock, and COVID-19, can have both potential protective and detrimental effects. This article explores the multifaceted role of Ca2+ dysregulation in inflammation, fibrosis, impaired immune function, and accelerated senescence, contributing to disease severity. Targeting Ca2+ signaling pathways may provide opportunities to develop novel therapeutics for age-related diseases and combat viral infections. However, the role of Ca2+ in viral infections is complex, and evidence suggests that hypocalcemia may have a protective effect against certain viruses, while changes in Ca2+ homeostasis can influence susceptibility to viral infections. The effectiveness and safety of Ca2+ supplements in COVID-19 patients remain a subject of ongoing research and debate. Further investigations are needed to understand the intricate interplay between Ca2+ signaling and disease pathogenesis.
Collapse
Affiliation(s)
- Elham Ghafouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Anis Khalafiyan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohre Amirkhani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roham Ghanbari
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
35
|
Medeiros-Silva J, Dregni AJ, Somberg NH, Duan P, Hong M. Atomic structure of the open SARS-CoV-2 E viroporin. SCIENCE ADVANCES 2023; 9:eadi9007. [PMID: 37831764 PMCID: PMC10575589 DOI: 10.1126/sciadv.adi9007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/08/2023] [Indexed: 10/15/2023]
Abstract
The envelope (E) protein of the SARS-CoV-2 virus forms cation-conducting channels in the endoplasmic reticulum Golgi intermediate compartment (ERGIC) of infected cells. The calcium channel activity of E is associated with the inflammatory responses of COVID-19. Using solid-state NMR (ssNMR) spectroscopy, we have determined the open-state structure of E's transmembrane domain (ETM) in lipid bilayers. Compared to the closed state, open ETM has an expansive water-filled amino-terminal chamber capped by key glutamate and threonine residues, a loose phenylalanine aromatic belt in the middle, and a constricted polar carboxyl-terminal pore filled with an arginine and a threonine residue. This structure gives insights into how protons and calcium ions are selected by ETM and how they permeate across the hydrophobic gate of this viroporin.
Collapse
Affiliation(s)
| | - Aurelio J. Dregni
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Pu Duan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
36
|
Somberg NH, Medeiros-Silva J, Jo H, Wang J, DeGrado WF, Hong M. Hexamethylene amiloride binds the SARS-CoV-2 envelope protein at the protein-lipid interface. Protein Sci 2023; 32:e4755. [PMID: 37632140 PMCID: PMC10503410 DOI: 10.1002/pro.4755] [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: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023]
Abstract
The SARS-CoV-2 envelope (E) protein forms a five-helix bundle in lipid bilayers whose cation-conducting activity is associated with the inflammatory response and respiratory distress symptoms of COVID-19. E channel activity is inhibited by the drug 5-(N,N-hexamethylene) amiloride (HMA). However, the binding site of HMA in E has not been determined. Here we use solid-state NMR to measure distances between HMA and the E transmembrane domain (ETM) in lipid bilayers. 13 C, 15 N-labeled HMA is combined with fluorinated or 13 C-labeled ETM. Conversely, fluorinated HMA is combined with 13 C, 15 N-labeled ETM. These orthogonal isotopic labeling patterns allow us to conduct dipolar recoupling NMR experiments to determine the HMA binding stoichiometry to ETM as well as HMA-protein distances. We find that HMA binds ETM with a stoichiometry of one drug per pentamer. Unexpectedly, the bound HMA is not centrally located within the channel pore, but lies on the lipid-facing surface in the middle of the TM domain. This result suggests that HMA may inhibit the E channel activity by interfering with the gating function of an aromatic network. These distance data are obtained under much lower drug concentrations than in previous chemical shift perturbation data, which showed the largest perturbation for N-terminal residues. This difference suggests that HMA has higher affinity for the protein-lipid interface than the channel pore. These results give insight into the inhibition mechanism of HMA for SARS-CoV-2 E.
Collapse
Affiliation(s)
- Noah H Somberg
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - João Medeiros-Silva
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Jun Wang
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
37
|
Chen L, Yang L, Li Y, Liu T, Yang B, Liu L, Wu R. Autophagy and Inflammation: Regulatory Roles in Viral Infections. Biomolecules 2023; 13:1454. [PMID: 37892135 PMCID: PMC10604974 DOI: 10.3390/biom13101454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Autophagy is a highly conserved intracellular degradation pathway in eukaryotic organisms, playing an adaptive role in various pathophysiological processes throughout evolution. Inflammation is the immune system's response to external stimuli and tissue damage. However, persistent inflammatory reactions can lead to a range of inflammatory diseases and cancers. The interaction between autophagy and inflammation is particularly evident during viral infections. As a crucial regulator of inflammation, autophagy can either promote or inhibit the occurrence of inflammatory responses. In turn, inflammation can establish negative feedback loops by modulating autophagy to suppress excessive inflammatory reactions. This interaction is pivotal in the pathogenesis of viral diseases. Therefore, elucidating the regulatory roles of autophagy and inflammation in viral infections will significantly enhance our understanding of the mechanisms underlying related diseases. Furthermore, it will provide new insights and theoretical foundations for disease prevention, treatment, and drug development.
Collapse
Affiliation(s)
- Li Chen
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| | - Limin Yang
- School of Medicine, Dalian University, Dalian 116622, China;
| | - Yingyu Li
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| | - Tianrun Liu
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| | - Bolun Yang
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| | - Lei Liu
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| | - Rui Wu
- School of Medicine, Jiamusi University, Jiamusi 154007, China; (L.C.); (Y.L.); (T.L.); (B.Y.)
| |
Collapse
|
38
|
Wells EW, Parker MT. Regulating Select Agent Chimeras: Defining the Problem(s) Through the Lens of SARS-CoV-1/SARS-CoV-2 Chimeric Viruses. Health Secur 2023; 21:392-406. [PMID: 37703547 DOI: 10.1089/hs.2023.0008] [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] [Indexed: 09/15/2023] Open
Abstract
In late 2021, the US Centers for Disease Control and Prevention (CDC) posted an interim final rule (86 FR 64075) to the federal register regulating the possession, use, and transfer of SARS-CoV-1/SARS-CoV-2 chimeric viruses. In doing so, the CDC provided the reasoning that viral chimeras combining the transmissibility of SARS-CoV-2 with the pathogenicity and lethality of SARS-CoV-1 pose a significant risk to public health and should thus be placed on the select agents and toxins list. However, 86 FR 64075 lacked clarity in its definitions and scope, some of which the CDC addressed in response to public comments in the final rule, 88 FR 13322, in early 2023. To evaluate these regulatory actions, we reviewed the existing select agent regulations to understand the landscape of chimeric virus regulation. Based on our findings, we first present clear definitions for the terms "chimeric virus," "viral chimera," and "virulence factor" and provide a list of SARS-CoV-1 virulence factors in an effort to aid researchers and federal rulemaking for these agents moving forward. We then provide suggestions for a combination of similarity and functional characteristic cutoffs that the government could use to enable researchers to distinguish between regulated and nonregulated chimeras. Finally, we discuss current select agent regulations and their overlaps with 86 FR 64075 and 88 FR 13322 and make suggestions for how to address chimera concerns within and/or without these regulations. Collectively, we believe that our findings fill important gaps in current federal regulations and provide forward-looking philosophical and practical analysis that can guide future decisionmaking.
Collapse
Affiliation(s)
- Elizabeth W Wells
- Elizabeth W. Wells is a Student, Department of Biology, Georgetown College of Arts & Sciences, Georgetown University, Washington, DC
| | - Michael T Parker
- Michael T. Parker, PhD, is Assistant Dean, Georgetown College of Arts & Sciences, Georgetown University, Washington, DC
| |
Collapse
|
39
|
Zhang X, Chen G, Yin J, Li L, Huang K, Du Q, Tong D, Huang Y. Pseudorabies virus infection activates the NLRP3 and IFI16 inflammasomes to trigger pyroptosis. Vet Microbiol 2023; 284:109826. [PMID: 37421928 DOI: 10.1016/j.vetmic.2023.109826] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/10/2023]
Abstract
Pseudorabies virus (PRV) preferably invades neural tissue and various organs, whereupon may result in multisystemic lesions. Pyroptosis mediated by proteolytic cleavage of gasdermin D (GSDMD) by inflammatory caspases (caspase-1/4/5/11), is closely associated with the activation of inflammasomes, a multiprotein proinflammatory complex. However, further studies on the mechanisms regarding PRV-induced pyroptosis in its natural host are required. Herein, it is demonstrated that PRV infection triggered GSDMD- not GSDME-mediated pyroptosis in porcine alveolar macrophage cells, resulting in increased secretion of IL-1β and LDH. During this process, caspase-1 was activated and participated in the cleaving of GSDMD. Interestingly, we found that the viral replication process or protein production is required to induce pyroptotic cell death. Also, our findings showed that PRV triggered NLRP3 inflammasome activation, which was associated with the production of reactive oxygen species (ROS) and potassium efflux. In addition to the NLRP3 inflammasome, the IFI16 inflammasome was also activated. Importantly, the NLRP3- and IFI16- inflammasomes were both involved in pyroptosis during PRV infection. Finally, we observed that the cleaved GSDMD, activated caspase-1, increased IFI16 levels, and elevated NLRP3 protein in PRV-infected tissues (brain and lung), supporting the occurrence of pyroptosis and the activation of NLRP3- and IFI16- inflammasome in PRV-infected pigs. This research advances our understanding of the PRV-mediated inflammatory response and cell death pathways, providing a deeper knowledge of effective treatments for pseudorabies.
Collapse
Affiliation(s)
- Xiaohua Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Guiyuan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Junqing Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Linghao Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kai Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| |
Collapse
|
40
|
Sučec I, Mammeri NE, Dregni AJ, Hong M. Rapid Determination of the Topology of Oligomeric α-Helical Membrane Proteins by Water- and Lipid-Edited Methyl NMR. J Phys Chem B 2023; 127:7518-7530. [PMID: 37606918 PMCID: PMC10893779 DOI: 10.1021/acs.jpcb.3c05295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Single-span oligomeric α-helical transmembrane proteins are common in virus ion channels, which are targets of antiviral drugs. Knowledge about the high-resolution structures of these oligomeric α-helical bundles is so far scarce. Structure determination of these membrane proteins by solid-state NMR traditionally requires resolving and assigning protein chemical shifts and measuring many interhelical distances, which are time-consuming. To accelerate experimental structure determination, here we introduce a simple solid-state NMR approach that uses magnetization transfer from water and lipid protons to the protein. By detecting the water- and lipid-transferred intensities of the high-sensitivity methyl 13C signals of Leu, Val, and Ile residues, which are highly enriched in these membrane proteins, we can derive models of the topology of these homo-oligomeric helical bundles. The topology is specified by the positions of amino acid residues in heptad repeats and the orientations of residues relative to the channel pore, lipids, and the helical interface. We demonstrate this water- and lipid-edited methyl NMR approach on the envelope (E) protein of SARS-CoV-2, the causative agent of the COVID-19 pandemic. We show that water-edited and lipid-edited 2D 13C-13C correlation spectra can be measured with sufficient sensitivity. Even without resolving multiple residues of the same type in the NMR spectra, we can obtain the helical bundle topology. We apply these experiments to the structurally unknown E proteins of the MERS coronavirus and the human coronavirus NL63. The resulting structural topologies show interesting differences in the positions of the aromatic residues in these three E proteins, suggesting that these viroporins may have different mechanisms of activation and ion conduction.
Collapse
Affiliation(s)
- Iva Sučec
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Nadia El Mammeri
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Aurelio J. Dregni
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| |
Collapse
|
41
|
Ávila-Flores A, Sánchez-Cabezón JJ, Ochoa-Echeverría A, Checa AI, Rosas-García J, Téllez-Araiza M, Casado S, Liébana R, Santos-Mendoza T, Mérida I. Identification of Host PDZ-Based Interactions with the SARS-CoV-2 E Protein in Human Monocytes. Int J Mol Sci 2023; 24:12793. [PMID: 37628973 PMCID: PMC10454406 DOI: 10.3390/ijms241612793] [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: 07/01/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Proteins containing PDZ (post-synaptic density, PSD-95/disc large, Dlg/zonula occludens, ZO-1) domains assemble signaling complexes that orchestrate cell responses. Viral pathogens target host PDZ proteins by coding proteins containing a PDZ-binding motif (PBM). The presence of a PBM in the SARS-CoV-2 E protein contributes to the virus's pathogenicity. SARS-CoV-2 infects epithelia, but also cells from the innate immune response, including monocytes and alveolar macrophages. This process is critical for alterations of the immune response that are related to the deaths caused by SARS-CoV-2. Identification of E-protein targets in immune cells might offer clues to understanding how SARS-CoV-2 alters the immune response. We analyzed the interactome of the SARS-CoV-2 E protein in human monocytes. The E protein was expressed fused to a GFP tag at the amino terminal in THP-1 monocytes, and associated proteins were identified using a proteomic approach. The E-protein interactome provided 372 partners; only 8 of these harbored PDZ domains, including the cell polarity protein ZO-2, the chemoattractant IL-16, and syntenin. We addressed the expression and localization of the identified PDZ proteins along the differentiation of primary and THP-1 monocytes towards macrophages and dendritic cells. Our data highlight the importance of identifying the functions of PDZ proteins in the maintenance of immune fitness and the viral alteration of inflammatory response.
Collapse
Affiliation(s)
- Antonia Ávila-Flores
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Juan José Sánchez-Cabezón
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Ana I. Checa
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Jorge Rosas-García
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Mariana Téllez-Araiza
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Sara Casado
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Rosa Liébana
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Teresa Santos-Mendoza
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Isabel Mérida
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| |
Collapse
|
42
|
Santoro L, Zaccone V, Falsetti L, Ruggieri V, Danese M, Miro C, Di Giorgio A, Nesci A, D’Alessandro A, Moroncini G, Santoliquido A. Role of Endothelium in Cardiovascular Sequelae of Long COVID. Biomedicines 2023; 11:2239. [PMID: 37626735 PMCID: PMC10452509 DOI: 10.3390/biomedicines11082239] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The global action against coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, shed light on endothelial dysfunction. Although SARS-CoV-2 primarily affects the pulmonary system, multiple studies have documented pan-vascular involvement in COVID-19. The virus is able to penetrate the endothelial barrier, damaging it directly or indirectly and causing endotheliitis and multi-organ injury. Several mechanisms cooperate to development of endothelial dysfunction, including endothelial cell injury and pyroptosis, hyperinflammation and cytokine storm syndrome, oxidative stress and reduced nitric oxide bioavailability, glycocalyx disruption, hypercoagulability, and thrombosis. After acute-phase infection, some patients reported signs and symptoms of a systemic disorder known as long COVID, in which a broad range of cardiovascular (CV) disorders emerged. To date, the exact pathophysiology of long COVID remains unclear: in addition to the persistence of acute-phase infection mechanisms, specific pathways of CV damage have been postulated, such as persistent viral reservoirs in the heart or an autoimmune response to cardiac antigens through molecular mimicry. The aim of this review is to provide an overview of the main molecular patterns of enduring endothelial activation following SARS-CoV-2 infection and to offer the latest summary of CV complications in long COVID.
Collapse
Affiliation(s)
- Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Vincenzo Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vittorio Ruggieri
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Martina Danese
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Chiara Miro
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Angela Di Giorgio
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Antonio Nesci
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Alessia D’Alessandro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
43
|
Shi W, Jin M, Chen H, Wu Z, Yuan L, Liang S, Wang X, Memon FU, Eldemery F, Si H, Ou C. Inflammasome activation by viral infection: mechanisms of activation and regulation. Front Microbiol 2023; 14:1247377. [PMID: 37608944 PMCID: PMC10440708 DOI: 10.3389/fmicb.2023.1247377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023] Open
Abstract
Viral diseases are the most common problems threatening human health, livestock, and poultry industries worldwide. Viral infection is a complex and competitive dynamic biological process between a virus and a host/target cell. During viral infection, inflammasomes play important roles in the host and confer defense mechanisms against the virus. Inflammasomes are polymeric protein complexes and are considered important components of the innate immune system. These immune factors recognize the signals of cell damage or pathogenic microbial infection after activation by the canonical pathway or non-canonical pathway and transmit signals to the immune system to initiate the inflammatory responses. However, some viruses inhibit the activation of the inflammasomes in order to replicate and proliferate in the host. In recent years, the role of inflammasome activation and/or inhibition during viral infection has been increasingly recognized. Therefore, in this review, we describe the biological properties of the inflammasome associated with viral infection, discuss the potential mechanisms that activate and/or inhibit NLRP1, NLRP3, and AIM2 inflammasomes by different viruses, and summarize the reciprocal regulatory effects of viral infection on the NLRP3 inflammasome in order to explore the relationship between viral infection and inflammasomes. This review will pave the way for future studies on the activation mechanisms of inflammasomes and provide novel insights for the development of antiviral therapies.
Collapse
Affiliation(s)
- Wen Shi
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Mengyun Jin
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hao Chen
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | | | - Liuyang Yuan
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Si Liang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaohan Wang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Fareed Uddin Memon
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Fatma Eldemery
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| | - Changbo Ou
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| |
Collapse
|
44
|
Surya W, Tavares-Neto E, Sanchis A, Queralt-Martín M, Alcaraz A, Torres J, Aguilella VM. The Complex Proteolipidic Behavior of the SARS-CoV-2 Envelope Protein Channel: Weak Selectivity and Heterogeneous Oligomerization. Int J Mol Sci 2023; 24:12454. [PMID: 37569828 PMCID: PMC10420310 DOI: 10.3390/ijms241512454] [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: 07/07/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
The envelope (E) protein is a small polypeptide that can form ion channels in coronaviruses. In SARS coronavirus 2 (SARS-CoV-2), the agent that caused the recent COVID-19 pandemic, and its predecessor SARS-CoV-1, E protein is found in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), where virion budding takes place. Several reports claim that E protein promotes the formation of "cation-selective channels". However, whether this term represents specificity to certain ions (e.g., potassium or calcium) or the partial or total exclusion of anions is debatable. Herein, we discuss this claim based on the available data for SARS-CoV-1 and -2 E and on new experiments performed using the untagged full-length E protein from SARS-CoV-2 in planar lipid membranes of different types, including those that closely mimic the ERGIC membrane composition. We provide evidence that the selectivity of the E-induced channels is very mild and depends strongly on lipid environment. Thus, despite past and recent claims, we found no indication that the E protein forms cation-selective channels that prevent anion transport, and even less that E protein forms bona fide specific calcium channels. In fact, the E channel maintains its multi-ionic non-specific neutral character even in concentrated solutions of Ca2+ ions. Also, in contrast to previous studies, we found no evidence that SARS-CoV-2 E channel activation requires a particular voltage, high calcium concentrations or low pH, in agreement with available data from SARS-CoV-1 E. In addition, sedimentation velocity experiments suggest that the E channel population is mostly pentameric, but very dynamic and probably heterogeneous, consistent with the broad distribution of conductance values typically found in electrophysiological experiments. The latter has been explained by the presence of proteolipidic channel structures.
Collapse
Affiliation(s)
- Wahyu Surya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Ernesto Tavares-Neto
- Laboratory of Molecular Biophysics, Department of Physics, Universitat Jaume I, 12080 Castellon, Spain; (E.T.-N.); (M.Q.-M.); (A.A.)
| | - Andrea Sanchis
- Laboratory of Molecular Biophysics, Department of Physics, Universitat Jaume I, 12080 Castellon, Spain; (E.T.-N.); (M.Q.-M.); (A.A.)
| | - María Queralt-Martín
- Laboratory of Molecular Biophysics, Department of Physics, Universitat Jaume I, 12080 Castellon, Spain; (E.T.-N.); (M.Q.-M.); (A.A.)
| | - Antonio Alcaraz
- Laboratory of Molecular Biophysics, Department of Physics, Universitat Jaume I, 12080 Castellon, Spain; (E.T.-N.); (M.Q.-M.); (A.A.)
| | - Jaume Torres
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Vicente M. Aguilella
- Laboratory of Molecular Biophysics, Department of Physics, Universitat Jaume I, 12080 Castellon, Spain; (E.T.-N.); (M.Q.-M.); (A.A.)
| |
Collapse
|
45
|
Huang H, Li X, Zha D, Lin H, Yang L, Wang Y, Xu L, Wang L, Lei T, Zhou Z, Xiao YF, Xin HB, Fu M, Qian Y. SARS-CoV-2 E protein-induced THP-1 pyroptosis is reversed by Ruscogenin. Biochem Cell Biol 2023; 101:303-312. [PMID: 36927169 DOI: 10.1139/bcb-2022-0359] [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] [Indexed: 03/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an emerging pathogenic coronavirus, has been reported to cause excessive inflammation and dysfunction in multiple cells and organs, but the underlying mechanisms remain largely unknown. Here we showed exogenous addition of SARS-CoV-2 envelop protein (E protein) potently induced cell death in cultured cell lines, including THP-1 monocytic leukemia cells, endothelial cells, and bronchial epithelial cells, in a time- and concentration-dependent manner. SARS-CoV-2 E protein caused pyroptosis-like cell death in THP-1 and led to GSDMD cleavage. In addition, SARS-CoV-2 E protein upregulated the expression of multiple pro-inflammatory cytokines that may be attributed to activation of NF-κB, JNK and p38 signal pathways. Notably, we identified a natural compound, Ruscogenin, effectively reversed E protein-induced THP-1 death via inhibition of NLRP3 activation and GSDMD cleavage. In conclusion, these findings suggested that Ruscogenin may have beneficial effects on preventing SARS-CoV-2 E protein-induced cell death and might be a promising treatment for the complications of COVID-19.
Collapse
Affiliation(s)
- Houda Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Xiuzhen Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Duoduo Zha
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Hongru Lin
- Department of Scientific Research, Hainan General Hospital, Haikou, 570311, China
| | - Lingyi Yang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Yihan Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Luyan Xu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Linsiqi Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Tianhua Lei
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Zhou Zhou
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Yun-Fei Xiao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Mingui Fu
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| |
Collapse
|
46
|
Fani M, Moossavi M, Bakhshi H, Jahrodi AN, Khazdair MR, Zardast AH, Ghafari S. Targeting host calcium channels and viroporins: a promising strategy for SARS-CoV-2 therapy. Future Virol 2023:10.2217/fvl-2022-0203. [PMID: 37700758 PMCID: PMC10494978 DOI: 10.2217/fvl-2022-0203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 08/18/2023] [Indexed: 09/14/2023]
Abstract
Despite passing the pandemic phase of the COVID-19, researchers are still investigating various drugs. Previous evidence suggests that blocking the calcium channels may be a suitable treatment option. Ca2+ is required to enhance the fusion process of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Also, some important inflammatory factors during SARS-CoV-2 infection are dependent on Ca2+ level. On the other hand, viroporins have emerged as attractive targets for antiviral therapy due to their essential role in viral replication and pathogenesis. By inhibiting the host calcium channels and viroporins, it is possible to limit the spread of infection. Therefore, calcium channel blockers (CCBs) and drugs targeting Viroporins can be considered an effective option in the fight against SARS-CoV-2.
Collapse
Affiliation(s)
- Mona Fani
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
- North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Maryam Moossavi
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran
| | - Hasan Bakhshi
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Mohammad Reza Khazdair
- Pharmaceutical Science & Clinical Physiology, Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Shokouh Ghafari
- Cellular & Molecular Research Center, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran
- Department of Microbiology & Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran
| |
Collapse
|
47
|
Chen M, He Y, Hu X, Dong X, Yan Z, Zhao Q, Li J, Xiang D, Lin Y, Song H, Bian X. Vitamin D3 attenuates SARS-CoV-2 nucleocapsid protein-caused hyperinflammation by inactivating the NLRP3 inflammasome through the VDR-BRCC3 signaling pathway in vitro and in vivo. MedComm (Beijing) 2023; 4:e318. [PMID: 37361896 PMCID: PMC10285036 DOI: 10.1002/mco2.318] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection-caused coronavirus disease 2019 (COVID-19) is a global crisis with no satisfactory therapies. Vitamin D3 (VD3) is considered a potential candidate for COVID-19 treatment; however, little information is available regarding the exact effects of VD3 on SARS-CoV-2 infection and the underlying mechanism. Herein, we confirmed that VD3 reduced SARS-CoV-2 nucleocapsid (N) protein-caused hyperinflammation in human bronchial epithelial (HBE) cells. Meanwhile, VD3 inhibited the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation in N protein-overexpressed HBE (HBE-N) cells. Notably, the inhibitors of caspase-1, NLRP3, and NLRP3 or caspase-1 small interference RNA (siRNA) enhanced VD3-induced NLRP3 inflammasome inactivation, with subsequent suppression of interleukin-6 (IL6) and IL1β release in HBE-N cells, which were abolished by the NLRP3 agonist. Moreover, VD3 increased NLRP3 ubiquitination (Ub-NLRP3) expression and the binding of the VDR with NLRP3, with decreased BRCA1/BRCA2-containing complex subunit 3 (BRCC3) expression and NLRP3-BRCC3 association. VD3-induced Ub-NLRP3 expression, NLRP3 inflammasome inactivation, and hyperinflammation inhibition were improved by the BRCC3 inhibitor or BRCC3 siRNA, which were attenuated by the vitamin D receptor (VDR) antagonist or VDR siRNA in HBE-N cells. Finally, the results of the in vivo study in AAV-Lung-enhanced green fluorescent protein-N-infected lungs were consistent with the findings of the in vitro experiment. In conclusion, VD3 attenuated N protein-caused hyperinflammation by inactivating the NLRP3 inflammasome partially through the VDR-BRCC3 signaling pathway.
Collapse
Affiliation(s)
- Mingliang Chen
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
- Institute of ToxicologySchool of Military Preventive MedicineArmy Medical UniversityChongqingChina
| | - Ying He
- Department of UltrasoundXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Xiaofeng Hu
- Department of Health Supervision and SurveillanceChinese PLA Center for Disease Control and PreventionBeijingChina
| | - Xunhu Dong
- Institute of ToxicologySchool of Military Preventive MedicineArmy Medical UniversityChongqingChina
| | - Zexuan Yan
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Qingning Zhao
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Jingyuan Li
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Dongfang Xiang
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Yong Lin
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Hongbin Song
- Department of Health Supervision and SurveillanceChinese PLA Center for Disease Control and PreventionBeijingChina
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer CentreSouthwest HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|
48
|
Ewart G, Bobardt M, Bentzen BH, Yan Y, Thomson A, Klumpp K, Becker S, Rosenkilde MM, Miller M, Gallay P. Post-infection treatment with the E protein inhibitor BIT225 reduces disease severity and increases survival of K18-hACE2 transgenic mice infected with a lethal dose of SARS-CoV-2. PLoS Pathog 2023; 19:e1011328. [PMID: 37549173 PMCID: PMC10434922 DOI: 10.1371/journal.ppat.1011328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/17/2023] [Accepted: 07/06/2023] [Indexed: 08/09/2023] Open
Abstract
The Coronavirus envelope (E) protein is a small structural protein with ion channel activity that plays an important role in virus assembly, budding, immunopathogenesis and disease severity. The viroporin E is also located in Golgi and ER membranes of infected cells and is associated with inflammasome activation and immune dysregulation. Here we evaluated in vitro antiviral activity, mechanism of action and in vivo efficacy of BIT225 for the treatment of SARS-CoV-2 infection. BIT225 showed broad-spectrum direct-acting antiviral activity against SARS-CoV-2 in Calu3 and Vero cells with similar potency across 6 different virus strains. BIT225 inhibited ion channel activity of E protein but did not inhibit endogenous currents or calcium-induced ion channel activity of TMEM16A in Xenopus oocytes. BIT225 administered by oral gavage for 12 days starting 12 hours before infection completely prevented body weight loss and mortality in SARS-CoV-2 infected K18 mice (100% survival, n = 12), while all vehicle-dosed animals reached a mortality endpoint by Day 9 across two studies (n = 12). When treatment started at 24 hours after infection, body weight loss, and mortality were also prevented (100% survival, n = 5), while 4 of 5 mice maintained and increased body weight and survived when treatment started 48 hours after infection. Treatment efficacy was dependent on BIT225 dose and was associated with significant reductions in lung viral load (3.5 log10), virus titer (4000 pfu/ml) and lung and serum cytokine levels. These results validate viroporin E as a viable antiviral target and support the clinical study of BIT225 for treatment and prophylaxis of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Gary Ewart
- Biotron Limited, North Ryde, New South Wales, Australia
| | - Michael Bobardt
- The Scripps Institute, Immunology and Microbiology, La Jolla, California, United States of America
| | - Bo Hjorth Bentzen
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen, Denmark
| | - Yannan Yan
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen, Denmark
| | | | - Klaus Klumpp
- Biotron Limited, North Ryde, New South Wales, Australia
| | | | - Mette M. Rosenkilde
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen, Denmark
| | | | - Philippe Gallay
- The Scripps Institute, Immunology and Microbiology, La Jolla, California, United States of America
| |
Collapse
|
49
|
Dinda B, Dinda M, Dinda S, De UC. An overview of anti-SARS-CoV-2 and anti-inflammatory potential of baicalein and its metabolite baicalin: Insights into molecular mechanisms. Eur J Med Chem 2023; 258:115629. [PMID: 37437351 DOI: 10.1016/j.ejmech.2023.115629] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
The current Coronavirus Disease 2019 (COVID-19) pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is highly contagious infection that breaks the healthcare systems of several countries worldwide. Till to date, no effective antiviral drugs against COVID-19 infection have reached the market, and some repurposed drugs and vaccines are prescribed for the treatment and prevention of this disease. The currently prescribed COVID-19 vaccines are less effective against the newly emergent variants of concern of SARS-CoV-2 due to several mutations in viral spike protein and obviously there is an urgency to develop new antiviral drugs against this disease. In this review article, we systematically discussed the anti-SARS-CoV-2 and anti-inflammatory efficacy of two flavonoids, baicalein and its 7-O-glucuronide, baicalin, isolated from Scutellaria baicalensis, Oroxylum indicum, and other plants as well as their pharmacokinetics and oral bioavailability, for development of safe and effective drugs for COVID-19 treatment. Both baicalein and baicalin target the activities of viral S-, 3CL-, PL-, RdRp- and nsp13-proteins, and host mitochondrial OXPHOS for suppression of viral infection. Moreover, these compounds prevent sepsis-related inflammation and organ injury by modulation of host innate immune responses. Several nanoformulated and inclusion complexes of baicalein and baicalin have been reported to increase oral bioavailability, but their safety and efficacy in SARS-CoV-2-infected transgenic animals are not yet evaluated. Future studies on these compounds are required for use in clinical trials of COVID-19 patients.
Collapse
Affiliation(s)
- Biswanath Dinda
- Department of Chemistry, Tripura University, Suryamaninagar, Agartala, Tripura, India.
| | - Manikarna Dinda
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Subhajit Dinda
- Department of Chemistry, Government Degree College, Kamalpur, Dhalai, Tripura, India
| | - Utpal Chandra De
- Department of Chemistry, Tripura University, Suryamaninagar, Agartala, Tripura, India
| |
Collapse
|
50
|
Cerato JA, da Silva EF, Porto BN. Breaking Bad: Inflammasome Activation by Respiratory Viruses. BIOLOGY 2023; 12:943. [PMID: 37508374 PMCID: PMC10376673 DOI: 10.3390/biology12070943] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023]
Abstract
The nucleotide-binding domain leucine-rich repeat-containing receptor (NLR) family is a group of intracellular sensors activated in response to harmful stimuli, such as invading pathogens. Some NLR family members form large multiprotein complexes known as inflammasomes, acting as a platform for activating the caspase-1-induced canonical inflammatory pathway. The canonical inflammasome pathway triggers the secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 by the rapid rupture of the plasma cell membrane, subsequently causing an inflammatory cell death program known as pyroptosis, thereby halting viral replication and removing infected cells. Recent studies have highlighted the importance of inflammasome activation in the response against respiratory viral infections, such as influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While inflammasome activity can contribute to the resolution of respiratory virus infections, dysregulated inflammasome activity can also exacerbate immunopathology, leading to tissue damage and hyperinflammation. In this review, we summarize how different respiratory viruses trigger inflammasome pathways and what harmful effects the inflammasome exerts along with its antiviral immune response during viral infection in the lungs. By understanding the crosstalk between invading pathogens and inflammasome regulation, new therapeutic strategies can be exploited to improve the outcomes of respiratory viral infections.
Collapse
Affiliation(s)
- Julia A. Cerato
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
| | - Emanuelle F. da Silva
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
| | - Barbara N. Porto
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|