1
|
Rodrigues TS, Zamboni DS. Inflammasome Activation by RNA Respiratory Viruses: Mechanisms, Viral Manipulation, and Therapeutic Insights. Immunol Rev 2025; 330:e70003. [PMID: 39891396 DOI: 10.1111/imr.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 02/03/2025]
Abstract
Respiratory viruses, including SARS-CoV-2, influenza, parainfluenza, rhinovirus, and respiratory syncytial virus (RSV), are pathogens responsible for lower respiratory tract infections, particularly in vulnerable populations such as children and the elderly. Upon infection, these viruses are recognized by pattern recognition receptors, leading to the activation of inflammasomes, which are essential for mediating inflammatory responses. This review discusses the mechanisms by which these RNA respiratory viruses activate inflammasomes, emphasizing the roles of various signaling pathways and components involved in this process. Additionally, we highlight the specific interactions between viral proteins and inflammasome sensors, elucidating how these viruses manipulate the host immune response to facilitate infection. Understanding the dynamics of inflammasome activation in response to respiratory viruses provides critical insights for developing immunomodulatory therapeutic strategies aimed at mitigating inflammation and improving outcomes in respiratory tract infections.
Collapse
Affiliation(s)
- Tamara S Rodrigues
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Dario S Zamboni
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
2
|
Lu Y, Liang X, Song J, Guan Y, Yang L, Shen R, Niu Y, Guo Z, Zhu N. Niclosamide modulates phenotypic switch and inflammatory responses in human pulmonary arterial smooth muscle cells. Mol Cell Biochem 2025; 480:1583-1593. [PMID: 38980591 DOI: 10.1007/s11010-024-05061-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 06/29/2024] [Indexed: 07/10/2024]
Abstract
Excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) represent key steps of pulmonary vascular remodeling, leading to the development of pulmonary arterial hypertension (PAH) and right ventricular failure. Niclosamide (NCL), an FDA-approved anthelmintic, has been shown to regulate cell proliferation, migration, invasion, and apoptosis through a variety of signaling pathways. However, its role on modulating the phenotypic switch and inflammatory responses in PASMCs remains unclear. In this study, cell proliferation assay showed that NCL inhibited PDGF-BB induced proliferation of human PASMCs in a dose-dependent manner. Western blot analysis further confirmed a notable reduction in the expression of cyclin D1 and PCNA proteins. Subsequently, flow cytometry analysis demonstrated that NCL induced an increased percentage of cells in the G1 phase while promoting apoptosis in PASMCs. Moreover, both scratch wound assay and transwell assay confirmed that NCL decreased PDGF-BB-induced migration of PASMCs. Mechanistically, western blot revealed that pretreatment of PASMCs with NCL markedly restored the protein levels of SMA, SM22, and calponin, while reducing phosphorylation of P38/STAT3 signaling in the presence of PDGF-BB. Interestingly, macrophages adhesion assay showed that NCL markedly reduced recruitment of Calcein-AM labeled RAW264.7 by TNFα-stimulated PASMCs. Western blot revealed that NCL suppressed TNFα-induced expression of both of VCAM-1 and ICAM-1 proteins. Furthermore, pretreatment of PASMCs with NCL significantly inhibited NLRP3 inflammasome activity through reducing NLRP3, AIM2, mature interleukin-1β (IL-β), and cleaved Caspase-1 proteins expression. Together, these results suggested versatile effects of NCL on controlling of proliferation, migration, and inflammatory responses in PASMCs through modulating different pathways, indicating that repurposing of NCL may emerge as a highly effective drug for PAH treatment.
Collapse
Affiliation(s)
- Yuwen Lu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaogan Liang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jingwen Song
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yugen Guan
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Liang Yang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Rongrong Shen
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yunpu Niu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhifu Guo
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Ni Zhu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
3
|
Ling J, Lundkvist Å, Guerrini M, Ferro V, Li J, Li J. A Heparan Sulfate Mimetic RAFT Copolymer Inhibits SARS-CoV-2 Infection and Ameliorates Viral-Induced Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411737. [PMID: 39679877 PMCID: PMC11809384 DOI: 10.1002/advs.202411737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/29/2024] [Indexed: 12/17/2024]
Abstract
The high transmissibility and mutation ability of coronaviruses enable them to easily escape existing immune protection and also pose a challenge to existing antiviral drugs. Moreover, drugs only targeting viruses cannot always attenuate the "cytokine storm". Herein, a synthetic heparan sulfate (HS) mimetic, HMSA-06 is reported, that exhibited antiviral activities against both the SARS-CoV-2 prototype and Omicron strains by targeting viral entry and replication. Of particular note, HMSA-06 demonstrated more potent anti-SARS-CoV-2 effects than PG545 and Roneparstat. SARS-CoV-2 is reported to hijack autophagy to facilitate its replication, therefore boosting autophagy can attenuate SARS-CoV-2 infection. It is revealed that HMSA-06, but not a similar HS mimetic that failed to inhibit SARS-CoV-2, can upregulate cellular autophagy flux. In addition, HMSA-06 was found to robustly block the NLRP3-mediated inflammatory reaction in SARS-CoV-2 infected THP-1 derived macrophages as evidenced by a reduction in inflammasome formation and the subsequent decreased secretion of mature caspase-1 and IL-1β. The HMSA-06's inflammation inhibitory function is further confirmed using a LPS/ATP-stimulated THP-1 macrophage model. Altogether, this study has identified a promising HS mimetic to combat SARS-CoV-2-associated diseases by inhibiting viral infection and attenuating viral-induced inflammatory reaction, providing insights into the development of novel anti-coronavirus drugs in the future.
Collapse
Affiliation(s)
- Jiaxin Ling
- Department of Medical Biochemistry and MicrobiologyThe Biomedical CenterUppsala UniversityUppsala75123Sweden
- Zoonosis Science CenterUppsala UniversityUppsala75123Sweden
| | - Åke Lundkvist
- Department of Medical Biochemistry and MicrobiologyThe Biomedical CenterUppsala UniversityUppsala75123Sweden
- Zoonosis Science CenterUppsala UniversityUppsala75123Sweden
| | - Marco Guerrini
- Istituto Di Ricerche Chimiche e Biochimiche G. RonzoniMilan20133Italy
| | - Vito Ferro
- School of Chemistry and Molecular BiosciencesThe University of QueenslandBrisbaneQLD4072Australia
- Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQLD4072Australia
| | - Jin‐Ping Li
- Department of Medical Biochemistry and MicrobiologyThe Biomedical CenterUppsala UniversityUppsala75123Sweden
- SciLifeLab UppsalaUppsala UniversityUppsala75123Sweden
| | - Jinlin Li
- Department of Medical Biochemistry and MicrobiologyThe Biomedical CenterUppsala UniversityUppsala75123Sweden
- Zoonosis Science CenterUppsala UniversityUppsala75123Sweden
| |
Collapse
|
4
|
Hiti L, Markovič T, Lainscak M, Farkaš Lainščak J, Pal E, Mlinarič-Raščan I. The immunopathogenesis of a cytokine storm: The key mechanisms underlying severe COVID-19. Cytokine Growth Factor Rev 2025:S1359-6101(24)00104-7. [PMID: 39884914 DOI: 10.1016/j.cytogfr.2024.12.003] [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: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025]
Abstract
A cytokine storm is marked by excessive pro-inflammatory cytokine release, and has emerged as a key factor in severe COVID-19 cases - making it a critical therapeutic target. However, its pathophysiology was poorly understood, which hindered effective treatment. SARS-CoV-2 initially disrupts angiotensin signalling, promoting inflammation through ACE-2 downregulation. Some patients' immune systems then fail to shift from innate to adaptive immunity, suppressing interferon responses and leading to excessive pyroptosis and neutrophil activation. This amplifies tissue damage and inflammation, creating a pro-inflammatory loop. The result is the disruption of Th1/Th2 and Th17/Treg balances, lymphocyte exhaustion, and extensive blood clotting. Cytokine storm treatments include glucocorticoids to suppress the immune system, monoclonal antibodies to neutralize specific cytokines, and JAK inhibitors to block cytokine receptor signalling. However, the most effective treatment options for mitigating SARS-CoV-2 infection remain vaccines as a preventive measure and antiviral drugs for the early stages of infection. This article synthesizes insights into immune dysregulation in COVID-19, offering a framework to better understand cytokine storms and to improve monitoring, biomarker discovery, and treatment strategies for COVID-19 and other conditions involving cytokine storms.
Collapse
Affiliation(s)
- Luka Hiti
- Faculty of Pharmacy, University of Ljubljana, Slovenia
| | | | - Mitja Lainscak
- General Hospital Murska Sobota, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Emil Pal
- General Hospital Murska Sobota, Slovenia
| | | |
Collapse
|
5
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [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: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
6
|
Khan A, Ling J, Li J. Is Autophagy a Friend or Foe in SARS-CoV-2 Infection? Viruses 2024; 16:1491. [PMID: 39339967 PMCID: PMC11437447 DOI: 10.3390/v16091491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
As obligate parasites, viruses need to hijack resources from infected cells to complete their lifecycle. The interaction between the virus and host determines the viral infection process, including viral propagation and the disease's outcome. Understanding the interaction between the virus and host factors is a basis for unraveling the intricate biological processes in the infected cells and thereby developing more efficient and targeted antivirals. Among the various fundamental virus-host interactions, autophagy plays vital and also complicated roles by directly engaging in the viral lifecycle and functioning as an anti- and/or pro-viral factor. Autophagy thus becomes a promising target against virus infection. Since the COVID-19 pandemic, there has been an accumulation of studies aiming to investigate the roles of autophagy in SARS-CoV-2 infection by using different models and from distinct angles, providing valuable information for systematically and comprehensively dissecting the interplay between autophagy and SARS-CoV-2. In this review, we summarize the advancements in the studies of the interaction between SARS-CoV-2 and autophagy, as well as detailed molecular mechanisms. We also update the current knowledge on the pharmacological strategies used to suppress SARS-CoV-2 replication through remodeling autophagy. These extensive studies on SARS-CoV-2 and autophagy can advance our understanding of virus-autophagy interaction and provide insights into developing efficient antiviral therapeutics by regulating autophagy.
Collapse
Affiliation(s)
- Asifa Khan
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Jiaxin Ling
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
- Zoonosis Science Center, Uppsala University, 751 23 Uppsala, Sweden
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
| |
Collapse
|
7
|
Shukla N, Shamim U, Agarwal P, Pandey R, Narayan J. From bench to bedside: potential of translational research in COVID-19 and beyond. Brief Funct Genomics 2024; 23:349-362. [PMID: 37986554 DOI: 10.1093/bfgp/elad051] [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: 09/07/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 2019 (COVID-19) have been around for more than 3 years now. However, due to constant viral evolution, novel variants are emerging, leaving old treatment protocols redundant. As treatment options dwindle, infection rates continue to rise and seasonal infection surges become progressively common across the world, rapid solutions are required. With genomic and proteomic methods generating enormous amounts of data to expand our understanding of SARS-CoV-2 biology, there is an urgent requirement for the development of novel therapeutic methods that can allow translational research to flourish. In this review, we highlight the current state of COVID-19 in the world and the effects of post-infection sequelae. We present the contribution of translational research in COVID-19, with various current and novel therapeutic approaches, including antivirals, monoclonal antibodies and vaccines, as well as alternate treatment methods such as immunomodulators, currently being studied and reiterate the importance of translational research in the development of various strategies to contain COVID-19.
Collapse
Affiliation(s)
- Nityendra Shukla
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Uzma Shamim
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Preeti Agarwal
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Rajesh Pandey
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Jitendra Narayan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| |
Collapse
|
8
|
Tsukalov I, Sánchez-Cerrillo I, Rajas O, Avalos E, Iturricastillo G, Esparcia L, Buzón MJ, Genescà M, Scagnetti C, Popova O, Martin-Cófreces N, Calvet-Mirabent M, Marcos-Jimenez A, Martínez-Fleta P, Delgado-Arévalo C, de Los Santos I, Muñoz-Calleja C, Calzada MJ, González Álvaro I, Palacios-Calvo J, Alfranca A, Ancochea J, Sánchez-Madrid F, Martin-Gayo E. NFκB and NLRP3/NLRC4 inflammasomes regulate differentiation, activation and functional properties of monocytes in response to distinct SARS-CoV-2 proteins. Nat Commun 2024; 15:2100. [PMID: 38453949 PMCID: PMC10920883 DOI: 10.1038/s41467-024-46322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Increased recruitment of transitional and non-classical monocytes in the lung during SARS-CoV-2 infection is associated with COVID-19 severity. However, whether specific innate sensors mediate the activation or differentiation of monocytes in response to different SARS-CoV-2 proteins remain poorly characterized. Here, we show that SARS-CoV-2 Spike 1 but not nucleoprotein induce differentiation of monocytes into transitional or non-classical subsets from both peripheral blood and COVID-19 bronchoalveolar lavage samples in a NFκB-dependent manner, but this process does not require inflammasome activation. However, NLRP3 and NLRC4 differentially regulated CD86 expression in monocytes in response to Spike 1 and Nucleoprotein, respectively. Moreover, monocytes exposed to Spike 1 induce significantly higher proportions of Th1 and Th17 CD4 + T cells. In contrast, monocytes exposed to Nucleoprotein reduce the degranulation of CD8 + T cells from severe COVID-19 patients. Our study provides insights in the differential impact of innate sensors in regulating monocytes in response to different SARS-CoV-2 proteins, which might be useful to better understand COVID-19 immunopathology and identify therapeutic targets.
Collapse
Affiliation(s)
- Ilya Tsukalov
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ildefonso Sánchez-Cerrillo
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Rajas
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Elena Avalos
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | | | - Laura Esparcia
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - María José Buzón
- Infectious Diseases Department, Institut de Recerca Hospital Univesritari Vall d'Hebrón (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Institut de Recerca Hospital Univesritari Vall d'Hebrón (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Camila Scagnetti
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Olga Popova
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Noa Martin-Cófreces
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Marta Calvet-Mirabent
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Ana Marcos-Jimenez
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Pedro Martínez-Fleta
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Cristina Delgado-Arévalo
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Ignacio de Los Santos
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
| | - María José Calzada
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Isidoro González Álvaro
- Rheumatology Department from Hospital Universitario La Princesa. Instituto de Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - José Palacios-Calvo
- Department of Pathology, Hospital Universitario Ramón y Cajal. Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Universidad de Alcalá. Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Ancochea
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Martin-Gayo
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain.
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain.
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Rodrigues TS, Zamboni DS. Inflammasome activation by SARS-CoV-2 and its participation in COVID-19 exacerbation. Curr Opin Immunol 2023; 84:102387. [PMID: 37703588 DOI: 10.1016/j.coi.2023.102387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
COVID-19 is an infectious and inflammatory disease caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome-Coronavirus-2) that might progress to severe illness in humans, characterized by excessive pulmonary and systemic inflammation. Exacerbated production of inflammatory cytokines and cell death contributes to disease aggravation and the inflammasomes take a central stage in this process. Activation of the NLRP3 has been demonstrated in macrophages and monocytes infected in vitro, in mouse models of infection, and in cells and lungs of severe cases of COVID-19. It is still not clear how SARS-CoV-2 activates the NLRP3 inflammasome, and recent reports suggest that the virus engages the CASP4/11 (Caspase 4/11)-mediated noncanonical activation of NLRP3. In this review, we discuss the recent data regarding the activation of NLRP3 inflammasome by SARS-CoV-2 and their participation in the development of severe cases of COVID-19.
Collapse
Affiliation(s)
- Tamara S Rodrigues
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14049-900, Brazil.
| |
Collapse
|
10
|
Li X, Chen S, Zhao L, Zeng X, Liu Y, Li C, Yang Q. Effect of lactic acid bacteria by different concentrations of copper based on non-target metabolomic analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:107568-107579. [PMID: 37737949 DOI: 10.1007/s11356-023-29925-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Copper (Cu) is an essential element for mammals, but excess intake can have detrimental health consequences. However, Cu is no longer present in the "Limit of Contaminants in Foods" promulgated in 2022. The potential impact of different Cu (II) concentrations on human health remains unclear. In this study, a strain of lactic acid bacteria (LAB), namely, Lactiplantibacillus plantarum CICC 23121 (L23121), was selected as a prebiotic indicator strain to indirectly assess the effects of food-limited Cu (II) concentrations (issued by Tolerance limit of copper in foods in 1994) on the functions of intestinal microbes. We used non-target metabolomics, automatic growth curve detector, scanning electron microscopy (SEM), and Fourier transform infrared spectroscopy (FTIR) to investigate the effects of Cu (II) on L23121. The study revealed shows that the 50% minimum inhibitory concentration (MIC50) of Cu (II) for most lactic acid bacteria was 4 mg/L. At low Cu (II) concentrations (≤ 4 mg/L), the pentose phosphate pathway and pyrimidine metabolism of the lactic acid bacteria were affected, resulting in a decrease in the content of beneficial secondary metabolites and a significant decrease in the cell activity. As Cu (II) concentrations increase (≥ 6 mg/L), the key amino acid and lipid metabolisms were affected, leading to the inhibition of growth and primary metabolite production of the bacteria. Under high concentration of Cu (II) (6 mg/L), the surface adhesion of the bacteria was distorted and covered with significantly large particles, and the functional groups of the cells were significantly shifted. As a probiotic, the abundance of lactic acid bacteria in the intestine is significantly reduced, which will inevitably seriously damage intestinal homeostasis. Thus, to protect human intestinal microbes' health, it is recommended to limit the concentration of Cu in food to less than 4 mg/L.
Collapse
Affiliation(s)
- Xinlei Li
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Shiyue Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Lili Zhao
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Xiangpeng Zeng
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Yanyan Liu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Chaochuang Li
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Qingxiang Yang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China.
- Henan International Joint Laboratory of Agricultural Microbial Ecology and Technology, Henan Normal University, Xinxiang, 453007, China.
| |
Collapse
|
11
|
Wang Y, Li P, Xu L, de Vries AC, Rottier RJ, Wang W, Crombag MRB, Peppelenbosch MP, Kainov DE, Pan Q. Combating pan-coronavirus infection by indomethacin through simultaneously inhibiting viral replication and inflammatory response. iScience 2023; 26:107631. [PMID: 37664584 PMCID: PMC10474465 DOI: 10.1016/j.isci.2023.107631] [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: 06/25/2023] [Revised: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023] Open
Abstract
Severe infections with coronaviruses are often accompanied with hyperinflammation, requiring therapeutic strategies to simultaneously tackle the virus and inflammation. By screening a safe-in-human broad-spectrum antiviral agents library, we identified that indomethacin can inhibit pan-coronavirus infection in human cell and airway organoids models. Combining indomethacin with oral antiviral drugs authorized for treating COVID-19 results in synergistic anti-coronavirus activity. Coincidentally, screening a library of FDA-approved drugs identified indomethacin as the most potent potentiator of interferon response through increasing STAT1 phosphorylation. Combining indomethacin with interferon-alpha exerted synergistic antiviral effects against multiple coronaviruses. The anti-coronavirus activity of indomethacin is associated with activating interferon response. In a co-culture system of lung epithelial cells with macrophages, indomethacin inhibited both viral replication and inflammatory response. Collectively, indomethacin is a pan-coronavirus inhibitor that can simultaneously inhibit virus-triggered inflammatory response. The therapeutic potential of indomethacin can be further augmented by combining it with oral antiviral drugs or interferon-alpha.
Collapse
Affiliation(s)
- Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Lei Xu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Annemarie C. de Vries
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Robbert J. Rottier
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands
- Department of Cell Biology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Marie-Rose B.S. Crombag
- Department of Hospital Pharmacy, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Maikel P. Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Denis E. Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7028 Trondheim, Norway
- Institute of Technology, University of Tartu, 50090 Tartu, Estonia
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Yang Z, Zhang Q, Wu X, Hao S, Hao X, Jones E, Zhang Y, Qiu J, Xu L. Repurposing Niclosamide as a Novel Anti-SARS-CoV-2 Drug by Restricting Entry Protein CD147. Biomedicines 2023; 11:2019. [PMID: 37509657 PMCID: PMC10377517 DOI: 10.3390/biomedicines11072019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/29/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the global coronavirus disease 2019 (COVID-19) pandemic, and the search for effective treatments has been limited. Furthermore, the rapid mutations of SARS-CoV-2 have posed challenges to existing vaccines and neutralizing antibodies, as they struggle to keep up with the increased viral transmissibility and immune evasion. However, there is hope in targeting the CD147-spike protein, which serves as an alternative point for the entry of SARS-CoV-2 into host cells. This protein has emerged as a promising therapeutic target for the development of drugs against COVID-19. Here, we demonstrate that the RNA-binding protein Human-antigen R (HuR) plays a crucial role in the post-transcriptional regulation of CD147 by directly binding to its 3'-untranslated region (UTR). We observed a decrease in CD147 levels across multiple cell lines upon HuR depletion. Furthermore, we identified that niclosamide can reduce CD147 by lowering the cytoplasmic translocation of HuR and reducing CD147 glycosylation. Moreover, our investigation revealed that SARS-CoV-2 infection induces an upregulation of CD147 in ACE2-expressing A549 cells, which can be effectively neutralized by niclosamide in a dose-dependent manner. Overall, our study unveils a novel regulatory mechanism of regulating CD147 through HuR and suggests niclosamide as a promising therapeutic option against COVID-19.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Qi Zhang
- Higuchi Biosciences Center, The University of Kansas, Lawrence, KS 66045, USA
| | - Xiaoqing Wu
- Higuchi Biosciences Center, The University of Kansas, Lawrence, KS 66045, USA
- The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xinbao Hao
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Liang Xu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
- The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Radiation Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
13
|
Li Y, Wang Y, Li Y, de Vries AC, Li P, Peppelenbosch MP, Pan Q. Seasonal coronavirus infections trigger NLRP3 inflammasome activation in macrophages but is therapeutically targetable. Antiviral Res 2023; 216:105674. [PMID: 37459896 DOI: 10.1016/j.antiviral.2023.105674] [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: 05/07/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Seasonal coronaviruses widely circulate in the global population, and severe complications can occur in specific vulnerable populations. Little is known on their pathogenic mechanisms and no approved treatment is available. Here, we present anecdotal evidence that the level of IL-1β, a hallmark of inflammasome activation, appears elevated in a subset of seasonal coronavirus infected patients. We found that cultured human macrophages support the full life cycle of three cultivatable seasonal coronaviruses. Their infections effectively activate NLRP3 inflammasome activation through TLR4 ligation and NF-κB activation. This activation can be attenuated by specific pharmacological inhibitors and clinically used medications including dexamethasone and flufenamic acid. Interestingly, combination of antiviral and anti-inflammatory drugs simultaneously inhibit seasonal coronavirus-triggered inflammatory response and viral replication. Collectively, these findings show that the TLR4/NF-κB/NLRP3 axis drives seasonal coronavirus triggered-inflammatory response, which in turn represents a viable therapeutic target.
Collapse
Affiliation(s)
- Yang Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Yunlong Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Annemarie C de Vries
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
14
|
Wu N, Zheng C, Xu J, Ma S, Jia H, Yan M, An F, Zhou Y, Qi J, Bian H. Race between virus and inflammasomes: inhibition or escape, intervention and therapy. Front Cell Infect Microbiol 2023; 13:1173505. [PMID: 37465759 PMCID: PMC10351387 DOI: 10.3389/fcimb.2023.1173505] [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: 02/24/2023] [Accepted: 05/17/2023] [Indexed: 07/20/2023] Open
Abstract
The inflammasome is a multiprotein complex that further regulates cell pyroptosis and inflammation by activating caspase-1. The assembly and activation of inflammasome are associated with a variety of diseases. Accumulative studies have shown that inflammasome is a key modulator of the host's defense response to viral infection. Indeed, it has been established that activation of inflammasome occurs during viral infection. At the same time, the host has evolved a variety of corresponding mechanisms to inhibit unnecessary inflammasome activation. Therefore, here, we review and summarize the latest research progress on the interaction between inflammosomes and viruses, highlight the assembly and activation of inflammosome in related cells after viral infection, as well as the corresponding molecular regulatory mechanisms, and elucidate the effects of this activation on virus immune escape and host innate and adaptive immune defenses. Finally, we also discuss the potential therapeutic strategies to prevent and/or ameliorate viral infection-related diseases via targeting inflammasomes and its products.
Collapse
Affiliation(s)
- Nijin Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chunzhi Zheng
- Shandong Provincial Hospital for Skin Diseases and Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiarui Xu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shujun Ma
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huimin Jia
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Meizhu Yan
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Fuxiang An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yi Zhou
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jianni Qi
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongjun Bian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Deng CH, Li TQ, Zhang W, Zhao Q, Wang Y. Targeting Inflammasome Activation in Viral Infection: A Therapeutic Solution? Viruses 2023; 15:1451. [PMID: 37515138 PMCID: PMC10384481 DOI: 10.3390/v15071451] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammasome activation is exclusively involved in sensing activation of innate immunity and inflammatory response during viral infection. Accumulating evidence suggests that the manipulation of inflammasome assembly or its interaction with viral proteins are critical factors in viral pathogenesis. Results from pilot clinical trials show encouraging results of NLRP3 inflammasome suppression in reducing mortality and morbidity in SARS-CoV-2-infected patients. In this article, we summarize the up-to-date understanding of inflammasomes, including NLRP3, AIM2, NLRP1, NLRP6, and NLRC4 in various viral infections, with particular focus on RNA viruses such as SARS-CoV-2, HIV, IAV, and Zika virus and DNA viruses such as herpes simplex virus 1. We also discuss the current achievement of the mechanisms involved in viral infection-induced inflammatory response, host defense, and possible therapeutic solutions.
Collapse
Affiliation(s)
- Chuan-Han Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Tian-Qi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Av. Wai Long, Taipa, Macao 999078, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- Minister of Education Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
17
|
Zhang Y, Duan Z, Guan Y, Xu T, Fu Y, Li G. Identification of 3 key genes as novel diagnostic and therapeutic targets for OA and COVID-19. Front Immunol 2023; 14:1167639. [PMID: 37283761 PMCID: PMC10239847 DOI: 10.3389/fimmu.2023.1167639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Background Corona Virus Disease 2019 (COVID-19) and Osteoarthritis (OA) are diseases that seriously affect the physical and mental health and life quality of patients, particularly elderly patients. However, the association between COVID-19 and osteoarthritis at the genetic level has not been investigated. This study is intended to analyze the pathogenesis shared by OA and COVID-19 and to identify drugs that could be used to treat SARS-CoV-2-infected OA patients. Methods The four datasets of OA and COVID-19 (GSE114007, GSE55235, GSE147507, and GSE17111) used for the analysis in this paper were obtained from the GEO database. Common genes of OA and COVID-19 were identified through Weighted Gene Co-Expression Network Analysis (WGCNA) and differential gene expression analysis. The least absolute shrinkage and selection operator (LASSO) algorithm was used to screen key genes, which were analyzed for expression patterns by single-cell analysis. Finally, drug prediction and molecular docking were carried out using the Drug Signatures Database (DSigDB) and AutoDockTools. Results Firstly, WGCNA identified a total of 26 genes common between OA and COVID-19, and functional analysis of the common genes revealed the common pathological processes and molecular changes between OA and COVID-19 are mainly related to immune dysfunction. In addition, we screened 3 key genes, DDIT3, MAFF, and PNRC1, and uncovered that key genes are possibly involved in the pathogenesis of OA and COVID-19 through high expression in neutrophils. Finally, we established a regulatory network of common genes between OA and COVID-19, and the free energy of binding estimation was used to identify suitable medicines for the treatment of OA patients infected with SARS-CoV-2. Conclusion In the present study, we succeeded in identifying 3 key genes, DDIT3, MAFF, and PNRC1, which are possibly involved in the development of both OA and COVID-19 and have high diagnostic value for OA and COVID-19. In addition, niclosamide, ciclopirox, and ticlopidine were found to be potentially useful for the treatment of OA patients infected with SARS-CoV-2.
Collapse
|
18
|
STARK RYAN. Protein-mediated interactions in the dynamic regulation of acute inflammation. BIOCELL 2023; 47:1191-1198. [PMID: 37261220 PMCID: PMC10231872 DOI: 10.32604/biocell.2023.027838] [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: 11/18/2022] [Accepted: 02/09/2023] [Indexed: 06/02/2023]
Abstract
Protein-mediated interactions are the fundamental mechanism through which cells regulate health and disease. These interactions require physical contact between proteins and their respective targets of interest. These targets include not only other proteins but also nucleic acids and other important molecules as well. These proteins are often involved in multibody complexes that work dynamically to regulate cellular health and function. Various techniques have been adapted to study these important interactions, such as affinity-based assays, mass spectrometry, and fluorescent detection. The application of these techniques has led to a greater understanding of how protein interactions are responsible for both the instigation and resolution of acute inflammatory diseases. These pursuits aim to provide opportunities to target specific protein interactions to alleviate acute inflammation.
Collapse
Affiliation(s)
- RYAN STARK
- Department of Pediatric Critical Care Medicine, Vanderbilt University Medical Center, 2200 Children’s Way, 5121 Doctors’ Office Tower, Nashville, TN 37232-9075
| |
Collapse
|
19
|
Wang M, Yu F, Chang W, Zhang Y, Zhang L, Li P. Inflammasomes: a rising star on the horizon of COVID-19 pathophysiology. Front Immunol 2023; 14:1185233. [PMID: 37251383 PMCID: PMC10213254 DOI: 10.3389/fimmu.2023.1185233] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.
Collapse
Affiliation(s)
- Man Wang
- *Correspondence: Man Wang, ; Peifeng Li,
| | | | | | | | | | - Peifeng Li
- *Correspondence: Man Wang, ; Peifeng Li,
| |
Collapse
|
20
|
Wang RS, Loscalzo J. Repurposing Drugs for the Treatment of COVID-19 and Its Cardiovascular Manifestations. Circ Res 2023; 132:1374-1386. [PMID: 37167362 PMCID: PMC10171294 DOI: 10.1161/circresaha.122.321879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
COVID-19 is an infectious disease caused by SARS-CoV-2 leading to the ongoing global pandemic. Infected patients developed a range of respiratory symptoms, including respiratory failure, as well as other extrapulmonary complications. Multiple comorbidities, including hypertension, diabetes, cardiovascular diseases, and chronic kidney diseases, are associated with the severity and increased mortality of COVID-19. SARS-CoV-2 infection also causes a range of cardiovascular complications, including myocarditis, myocardial injury, heart failure, arrhythmias, acute coronary syndrome, and venous thromboembolism. Although a variety of methods have been developed and many clinical trials have been launched for drug repositioning for COVID-19, treatments that consider cardiovascular manifestations and cardiovascular disease comorbidities specifically are limited. In this review, we summarize recent advances in drug repositioning for COVID-19, including experimental drug repositioning, high-throughput drug screening, omics data-based, and network medicine-based computational drug repositioning, with particular attention on those drug treatments that consider cardiovascular manifestations of COVID-19. We discuss prospective opportunities and potential methods for repurposing drugs to treat cardiovascular complications of COVID-19.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Channing Division of Network Medicine (R.-S.W., J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
| | - Joseph Loscalzo
- Channing Division of Network Medicine (R.-S.W., J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
- Division of Cardiovascular Medicine (J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
| |
Collapse
|
21
|
Yang Z, Zhang Q, Wu X, Hao S, Hao X, Jones E, Zhang Y, Qiu J, Xu L. Repurposing niclosamide as a novel anti-SARS-Cov-2 drug by restricting entry protein CD147. RESEARCH SQUARE 2023:rs.3.rs-2763207. [PMID: 37090542 PMCID: PMC10120763 DOI: 10.21203/rs.3.rs-2763207/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Background The burst of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing the global COVID-19 pandemic. But until today only limited numbers of drugs are discovered to treat COVID-19 patients. Even worse, the rapid mutations of SARS-CoV-2 compromise the effectiveness of existing vaccines and neutralizing antibodies due to the increased viral transmissibility and immune escape. CD147-spike protein, one of the entries of SRAR-CoV-2 into host cells, has been reported as a promising therapeutic target for developing drugs against COVID-19. Methods CRISPR-Cas9 induced gene knockout, western blotting, tet-off protein overexpression, ribonucleoprotein IP and RNA-IP were used to confirm the regulation of HuR on mRNA of CD147. Regulation of niclosamide on HuR nucleo-translocation was assessed by immunofluorescence staining of cell lines, IHC staining of tissue of mouse model and western blotting. Finally, the suppression of niclosamide on SARS-CoV-2 infection induced CD147 was evaluated by ACE2-expressing A549 cells and western blotting. Results We first discovered a novel regulation mechanism of CD147 via the RNA-binding protein HuR. We found that HuR regulates CD147 post-transcription by directly bound to its 3'-UTR. The loss of HuR reduced CD147 in multiple cell lines. Niclosamide inhibited CD147 function by blocking HuR cytoplasmic translocation and diminishing CD147 glycosylation. SARS-CoV-2 infection induced CD147 in ACE2-expressing A549 cells, which could be neutralized by niclosamide in a dose-dependent manner. Conclusion Together, our study reveals a novel regulation mechanism of CD147 and niclosamide can be repurposed as an effective COVID-19 drug by targeting the virus entry, CD147-spike protein.
Collapse
|
22
|
Leal VNC, Andrade MMS, Teixeira FME, Cambui RAG, Roa MEGV, Marra LG, Yamada SM, Alberca RW, Gozzi-Silva SC, Yendo TM, Netto LC, Duarte AJS, Sato MN, Pontillo A. Severe COVID-19 patients show a dysregulation of the NLRP3 inflammasome in circulating neutrophils. Scand J Immunol 2023; 97:e13247. [PMID: 36541819 DOI: 10.1111/sji.13247] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
SARS-CoV-2 triggers inflammasome-dependent release of pro-inflammatory cytokine IL-1β and pyroptosis, therefore, contributes to the huge inflammatory response observed in severe COVID-19 patients. Less is known about the engagement of inflammasome in neutrophils, main players in tissue injury and severe infection. We studied the activation of the inflammasome in neutrophils from severe COVID-19 patients and assessed its consequence in term of cells contribution to disease pathogenesis. We demonstrated that NLRP3 inflammasome is dramatically activated in neutrophils from severe COVID-19 patients and that the specific inhibition of NLRP3 reverts neutrophils' activation. Next, the stimulation of severe patients' neutrophils with common NLRP3 stimuli was not able to further activate the inflammasome, possibly due to exhaustion or increased percentage of circulating immature neutrophils. Collectively, our results demonstrate that the NLRP3 inflammasome is hyperactivated in severe COVID-19 neutrophils and its exhaustion may be responsible for the increased susceptibility to subsequent (and possibly lethal) infections. Our findings thus include a novel piece in the complex puzzle of COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Vinicius N C Leal
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| | - Milena M S Andrade
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Franciane M E Teixeira
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Raylane A G Cambui
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| | - Mariela E G V Roa
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| | - Letícia G Marra
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| | - Suemy M Yamada
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| | - Ricardo W Alberca
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Sarah C Gozzi-Silva
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Tatiana M Yendo
- Departamento de Dermatologia, Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lucas C Netto
- Unidade Terapia Intensiva, Hospital das Clínicas/FMUSP, São Paulo, Brazil
| | - Alberto J S Duarte
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Maria N Sato
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Hospital das Clínicas e Faculdade de Medicina/HCFMUSP, São Paulo, Brazil
| | - Alessandra Pontillo
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, Brazil
| |
Collapse
|
23
|
Wang Y, Huang H, Li D, Zhao C, Li S, Qin P, Li Y, Yang X, Du W, Li W, Li Y. Identification of niclosamide as a novel antiviral agent against porcine epidemic diarrhea virus infection by targeting viral internalization. Virol Sin 2023; 38:296-308. [PMID: 36702255 PMCID: PMC10176444 DOI: 10.1016/j.virs.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), an enteropathogenic coronavirus, has catastrophic impacts on the global pig industry. However, there remain no effective drugs against PEDV infection. In this study, we utilized a recombinant PEDV expressing renilla luciferase (PEDV-Rluc) to screen potential anti-PEDV agents from an FDA-approved drug library in Vero cells. Four compounds were identified that significantly decreased luciferase activity of PEDV-Rluc. Among them, niclosamide was further characterized because it exhibited the most potent antiviral activity with the highest selectivity index. It can efficiently inhibit viral RNA synthesis, protein expression and viral progeny production of classical and variant PEDV strains in a dose-dependent manner. Time of addition assay showed that niclosamide exhibited potent anti-PEDV activity when added simultaneously with or after virus infection. Furthermore, niclosamide significantly inhibited the entry stage of PEDV infection by affecting viral internalization rather than viral attachment to cells. In addition, a combination with other small molecule inhibitors of endosomal acidification enhanced the anti-PEDV effect of niclosamide in vitro. Taken together, these findings suggested that niclosamide is a novel antiviral agent that might provide a basis for the development of novel drug therapies against PEDV and other related pathogenic coronavirus infections.
Collapse
Affiliation(s)
- Yue Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Huimin Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Dongliang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Shuai Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Panpan Qin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yaqin Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xia Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wenjuan Du
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China.
| | - Yongtao Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China; Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands.
| |
Collapse
|
24
|
Yang Z, Cai X, Ye Q, Zhao Y, Li X, Zhang S, Zhang L. High-Throughput Screening for the Potential Inhibitors of SARS-CoV-2 with Essential Dynamic Behavior. Curr Drug Targets 2023; 24:532-545. [PMID: 36876836 DOI: 10.2174/1389450124666230306141725] [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: 06/18/2022] [Revised: 11/09/2022] [Accepted: 01/11/2023] [Indexed: 03/07/2023]
Abstract
Global health security has been challenged by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic. Due to the lengthy process of generating vaccinations, it is vital to reposition currently available drugs in order to relieve anti-epidemic tensions and accelerate the development of therapies for Coronavirus Disease 2019 (COVID-19), the public threat caused by SARS-CoV-2. High throughput screening techniques have established their roles in the evaluation of already available medications and the search for novel potential agents with desirable chemical space and more cost-effectiveness. Here, we present the architectural aspects of highthroughput screening for SARS-CoV-2 inhibitors, especially three generations of virtual screening methodologies with structural dynamics: ligand-based screening, receptor-based screening, and machine learning (ML)-based scoring functions (SFs). By outlining the benefits and drawbacks, we hope that researchers will be motivated to adopt these methods in the development of novel anti- SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Xinhui Cai
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Qiushi Ye
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| |
Collapse
|