1
|
Valente Coronel PM, Luiz Soares Basilio DC, Teixeira Espinoça I, Souza de Souza KF, Miranda Campos N, Seiji Nakano Ota R, Paredes-Gamero EJ, Wilhelm Filho D, Coimbra Motta-Castro AR, Trentin Perdomo R, Parisotto EB. Involvement of oxidative stress in post-acute sequelae of COVID-19: clinical implications. Redox Rep 2025; 30:2471738. [PMID: 40032512 PMCID: PMC11878172 DOI: 10.1080/13510002.2025.2471738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Oxidative stress (OS) plays a key role in the pathophysiology of COVID-19 and may be associated with sequelae after severe SARS-CoV-2 infection. This study evaluated OS and inflammation biomarkers in blood from individuals with post-acute sequelae of COVID-19 (PASC). 64 male and female participants were distributed into three groups: healthy individuals (n = 20), acute COVID-19 patients (symptoms for <3 weeks, n = 15), and PASC patients (symptoms for >12 weeks, n = 29). Analyses included inflammatory cytokines, myeloperoxidase (MPO) activity, and OS markers, such as superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), gamma-glutamyl transferase (GGT), reduced glutathione (GSH), uric acid (UA), thiobarbituric acid reactive substances (TBARS), and protein carbonyls (PC). Individuals with PASC showed increased IL-6 and IL-8. Both COVID-19 groups exhibited decreased SOD and CAT. GST decreased only in the acute group. Elevated GGT and GSH were found in the PASC group. High UA levels were observed in PASC individuals. There were no changes in TBARS values in the PASC group. However, PC concentrations were elevated only in this group. Correlations were identified between inflammatory markers and OS parameters. These findings suggest that individuals with PASC pronounced OS, which potentially exacerbates disease complications. Monitoring OS biomarkers could aid in patient prognosis and management.
Collapse
Affiliation(s)
- Paola Mayara Valente Coronel
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Denise Caroline Luiz Soares Basilio
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Isabelly Teixeira Espinoça
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Nathalia Miranda Campos
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Rafael Seiji Nakano Ota
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Edgar Julian Paredes-Gamero
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Danilo Wilhelm Filho
- Departamento de Ecologia e Zoologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ana Rita Coimbra Motta-Castro
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Renata Trentin Perdomo
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Eduardo Benedetti Parisotto
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| |
Collapse
|
2
|
Ebrahim N, Al Saihati HA, Alali Z, Mahmoud SYM, Rabaan AA, Dessouky AA, Salim RF, Shamaa AA, Abdallah AN, Elsherbiny NM, Othman G, Badawy AA, Di Leva G, Badr OA. Lyophilized MSC-EVs attenuates COVID-19 pathogenesis by regulating the JAK/STAT pathway. Stem Cell Res Ther 2025; 16:244. [PMID: 40369583 PMCID: PMC12079845 DOI: 10.1186/s13287-025-04284-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/19/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND The JAK/STAT signaling pathway plays a crucial role in the release of interferons (IFNs) and the proinflammatory response during SARS-CoV-2 infection, contributing to the cytokine storm characteristic of severe COVID-19 cases. STAT3, a key protein in this pathway, has been implicated in promoting inflammation, making its inhibition a potential therapeutic strategy to mitigate disease severity. Mesenchymal Stem Cell-derived Extracellular Vesicles (MSC-EVs), enriched with immunomodulatory and antiviral miRNAs, offer a promising therapeutic approach by modulating gene expression and regulating inflammatory responses. This study investigates the ability of Lyophilized MSC-EVs to inhibit the JAK/STAT pathway, highlighting their potential application in COVID-19 management. METHODS Male Syrian hamsters were used as an experimental model, housed under controlled laboratory conditions. SARS-CoV-2 (hCoV-19/Egypt/NRC-03/2020) was propagated in Vero E6 cells, and viral titers were determined using plaque assays. Hamsters were intranasally challenged with the virus and treated intraperitoneally with 0.5 mL of lyophilized human Wharton's jelly-derived MSC-extracellular vesicles (MSC-EVs). Histopathological evaluations were performed on lung tissues using H&E, Masson's trichrome, and immunohistochemical staining. Morphometric analyses were conducted to assess lung injury and fibrosis. Western blotting was employed to evaluate protein expression. All procedures adhered to ethical and biosafety guidelines. RESULTS The administration of MSC-EVs significantly upregulated the expression levels of miRNA-146a, miRNA-124, miRNA-155, miRNA-29b, miRNA-7, miRNA-145 and miRNA-18a compared to their levels in the COVID-19 group, suggesting a targeted release of miRNA-cargo from the MSC-EVs into the lung tissue of the animals. MSC-EVs impaired the activation of the STAT3/STAT1 signaling pathway and reduced the cytokine storm and coagulopathy associated with COVID-19. CONCLUSIONS These findings suggest that MSC-EVs have the potential to effectively mitigate the pathogenesis of COVID-19 by targeting the JAK/STAT signaling pathway. Further research is needed to fully understand the mechanisms underlying the therapeutic effects of MSC-EVs and their clinical application in combating the COVID-19 pandemic.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Medical Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt
- Stem Cell Unit, Faculty of Medicine, Benha University, Benha, Egypt
- Faculty of Medicine, Benha National University, Obour, Egypt
- Keele University, Keele, UK
| | - Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Hafar Al-Batin, Saudi Arabia.
| | - Zahraa Alali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, P.O Box 1803, 31991, Hafr Al Batin, Saudi Arabia
| | - Sabry Younis Mohamed Mahmoud
- Biology Department, College of Sciences, University of Hafr Al Batin, P. O. Box 1803, 31991, Hafar Al Batin, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, 31311, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Banha, Egypt
| | - Ashraf A Shamaa
- Anesthesiology & Radiology, Faculty of Vet. Men, Cairo University, P. O. Box 12211, Giza, Egypt
| | - Ahmed N Abdallah
- Hormones Department, National Research Centre, Medical Research and Clinical Studies Institute, Cairo, Egypt
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Gamal Othman
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, Saudi Arabia
| | - Abdelnaser A Badawy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Gianpiero Di Leva
- School of Life Sciences, Keele University Staffordshire, Keele, ST5 5BG, UK
| | - Omnia A Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt.
| |
Collapse
|
3
|
Tian L, Zhao Z, Gao W, Liu Z, Li X, Zhang W, Li Z. SARS-CoV-2 nsp16 is regulated by host E3 ubiquitin ligases, UBR5 and MARCHF7. eLife 2025; 13:RP102277. [PMID: 40358464 PMCID: PMC12074641 DOI: 10.7554/elife.102277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), remains a global public health threat with considerable economic consequences. The nonstructural protein 16 (nsp16), in complex with nsp10, facilitates the final viral mRNA capping step through its 2'-O-methylase activity, helping the virus to evade host immunity and prevent mRNA degradation. However, nsp16 regulation by host factors remains poorly understood. While various E3 ubiquitin ligases interact with SARS-CoV-2 proteins, their roles in targeting nsp16 for degradation remain unclear. In this study, we demonstrate that nsp16 undergoes ubiquitination and proteasomal degradation mediated by the host E3 ligases UBR5 and MARCHF7. UBR5 induces K48-linked ubiquitination, whereas MARCHF7 promotes K27-linked ubiquitination, independently suppressing SARS-CoV-2 replication in cell cultures and in mice. Notably, UBR5 and MARCHF7 also degrade nsp16 variants from different viral strains, exhibiting broad-spectrum antiviral activity. Our findings reveal novel antiviral mechanisms of the ubiquitin-proteasome system (UPS) and highlight their potential therapeutic targets against COVID-19.
Collapse
Affiliation(s)
- Li Tian
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zongzheng Zhao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenying Gao
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zirui Liu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Xiao Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenyan Zhang
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zhaolong Li
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
4
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
5
|
Gambari R, Papi C, Gasparello J, Agostinelli E, Finotti A. Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 2025; 29:85. [PMID: 40084194 PMCID: PMC11904878 DOI: 10.3892/etm.2025.12835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences and to develop novel therapeutic approaches. Interleukin-6 (IL-6) and IL-8 are considered significant therapeutic targets for COVID-19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19-25 nucleotides in length, non-coding RNAs that regulate gene expression at the post-transcriptional level through the sequence-selective recognition of the 3'-untranslated region (3'-UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID-19 'cytokine storm', miR-93-5p can inhibit IL-8 gene expression by directly targeting the 3'-UTR of IL-8 mRNA. In addition, miR-93-5p can regulate Toll-like receptor-4 (TLR4) and interleukin-1 receptor-associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor-κB (NF-κB) pathway and the expression of NF-κB-regulated genes, such as IL-6, IL-1β and other hyper-expressed genes during the COVID-19 'cytokine storm'. In the present study, the results provided preliminary evidence suggesting that the miR-93-5p-based miRNA therapeutics could be combined with the anti-inflammatory aged garlic extract (AGE) to more effectively inhibit IL-8 gene expression. The human bronchial epithelial IB3-1 cell line was employed as experimental model system. IB3-1 cells were stimulated with the BNT162b2 COVID-19 vaccine and transfected with pre-miR-93-5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2-induced expression of the IL-8 gene. The accumulation of IL-8 mRNA was assessed by RT-qPCR; the release of IL-8 protein was determined by Bio-Plex assay. In addition, the possible applications of TLR4/NF-κB inhibitory agents (such as miR-93-5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID-19, cystic fibrosis and other respiratory diseases, were summarized.
Collapse
Affiliation(s)
- Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
6
|
Ricci-Junior E, Rosa AS, do Nascimento T, Santos-Oliveira R, da Silva MAN, Barreto-Vieira DF, Batista LT, da Conceição GB, Quintão TAN, Ferreira VNS, Miranda MD. Nanotechnology-Driven Strategy Against SARS-CoV-2: Pluronic F127-Based Nanomicelles with or Without Atazanavir Reduce Viral Replication in Calu-3 Cells. Viruses 2025; 17:518. [PMID: 40284961 PMCID: PMC12031194 DOI: 10.3390/v17040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Despite extensive efforts, no highly effective antiviral molecule exists for treating moderate and severe COVID-19. Nanotechnology has emerged as a promising approach for developing novel drug delivery systems to enhance antiviral efficacy. Among these, polymeric nanomicelles improve the solubility, bioavailability, and cellular uptake of therapeutic agents. In this study, Pluronic F127-based nanomicelles were developed and evaluated for their antiviral activity against SARS-CoV-2. The nanomicelles, formulated using the direct dissolution method, exhibited an average size of 37.4 ± 8.01 nm and a polydispersity index (PDI) of 0.427 ± 0.01. Their antiviral efficacy was assessed in SARS-CoV-2-infected Vero E6 and Calu-3 cell models, where treatment with a 1:2 dilution inhibited viral replication by more than 90%. Cytotoxicity assays confirmed the nanomicelles were non-toxic to both cell lines after 72 h. In SARS-CoV-2-infected Calu-3 cells (human type II pneumocyte model), treatment with Pluronic F127-based nanomicelles containing atazanavir (ATV) significantly reduced viral replication, even under high MOI (2) and after 48 h, while also preventing IL-6 upregulation. To investigate their mechanism, viral pretreatment with nanomicelles showed no inhibitory effect. However, pre-exposure of Calu-3 cells led to significant viral replication reduction (>85% and >75% for 1:2 and 1:4 dilutions, respectively), as confirmed by transmission electron microscopy. These findings highlight Pluronic F127-based nanomicelles as a promising nanotechnology-driven strategy against SARS-CoV-2, reinforcing their potential for future antiviral therapies.
Collapse
Affiliation(s)
- Eduardo Ricci-Junior
- Galenic Development Laboratory, University Pharmacy, Universidade Federal de Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Alice Santos Rosa
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Tatielle do Nascimento
- Galenic Development Laboratory, University Pharmacy, Universidade Federal de Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmacy and Synthesis of Novel Radiopharmaceuticals, Nuclear Engineering Institute, Rio de Janeiro 21941-906, Brazil;
- Laboratory of Nanoradiopharmacy and Radiopharmaceuticals, Zona Oeste State University, Rio de Janeiro 21941-906, Brazil
| | - Marcos Alexandre Nunes da Silva
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Medicina Tropical, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Debora Ferreira Barreto-Vieira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
- Programa de Pós-graduação em Medicina Tropical, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Luísa Tozatto Batista
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Giovanna Barbosa da Conceição
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Tayane Alvites Nunes Quintão
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Vivian Neuza Santos Ferreira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Milene Dias Miranda
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
7
|
Lemos FFB, Lopes LW, Brito GC, Viana AIS, de Castro CT, Luz MS, Gonçalves AP, Dórea RSDM, da Silva FAF, de Brito BB, Santos MLC, Júnior GMS, de Lorenzo Barcia MTA, de Amorim Marques R, Botelho AB, Dantas ACS, Pinheiro FD, Teixeira AF, Souza CL, Oliveira MV, de Magalhães Queiroz DM, de Melo FF. Prognostic significance of cytokine dysregulation in critically ill COVID-19 patients. Cytokine 2025; 187:156867. [PMID: 39874939 DOI: 10.1016/j.cyto.2025.156867] [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: 07/01/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Understanding the immunopathogenesis of COVID-19 has yielded valuable insights into predicting adverse outcomes-particularly mortality. However, significant gaps persist in our comprehension of the complex interplay among the proposed pathophysiological mechanisms. Here, we aim to investigate the immunological factors associated with mortality in critically ill, unvaccinated COVID-19 patients admitted to the intensive care unit (ICU). METHODS We conducted a single-center, prospective study involving 56 unvaccinated COVID-19 patients admitted to the ICU. Plasma cytokine levels at admission were quantified using enzyme-linked immunosorbent assay (ELISA). Continuous variables were presented as median (IQR), and categorical variables as frequencies and percentages. Non-parametric tests assessed group differences. Logistic regression and receiver operating characteristic (ROC) curve analyses identified predictors of mortality, with bootstrapping (1000 re-samplings; 95 % BCa CI) applied for model validation. RESULTS Deceased patients exhibited significantly higher levels of interleukin (IL)-1β, IL-2, IL-6, transforming growth factor (TGF)-β, and interferon (IFN)-γ compared to survivors. Conversely, IL-10 and IL-27 were associated with favorable outcomes. Logistic regression modeling identified elevated IL-2 and IFN-γ levels as significant predictors of mortality. Notably, individual ROC curve analyses demonstrated that IL-1β and TGF-β had excellent discriminatory ability for mortality, while IFN-γ, IL-2, and IL-27 showed very good to excellent discriminatory capacity. CONCLUSION Our results indicate that distinct cytokine profiles differentiate survivors from non-survivors in critically ill, unvaccinated COVID-19 patients. These findings highlight the importance of cytokine dysregulation in severe COVID-19 cases and suggest potential targets for prognostic approaches. Further research is warranted to validate these results and translate them into effective clinical management strategies.
Collapse
Affiliation(s)
- Fabian Fellipe Bueno Lemos
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Luana Weber Lopes
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Carvalho Brito
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Airton Idalecio Sousa Viana
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Marcel Silva Luz
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - André Pereira Gonçalves
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | - Breno Bittencourt de Brito
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Maria Luísa Cordeiro Santos
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - André Bezerra Botelho
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Anna Carolina Saúde Dantas
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fillipe Dantas Pinheiro
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Adriano Fernandes Teixeira
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Cláudio Lima Souza
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Márcio Vasconcelos Oliveira
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Fabrício Freire de Melo
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil.
| |
Collapse
|
8
|
Liu A, Ruetalo N, Raja Xavier JP, Lankapalli AK, Admard J, Camarena-Sainz M, Brucker SY, Singh Y, Schindler M, Salker MS. Harnessing 2D and 3D human endometrial cell culture models to investigate SARS-CoV-2 infection in early pregnancy. Clin Sci (Lond) 2025; 139:CS20241215. [PMID: 39666439 DOI: 10.1042/cs20241215] [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: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/14/2024]
Abstract
Vertical transmission of SARS-CoV-2 during human pregnancy remains highly controversial as most studies have focused on the third trimester or the peripartum period. Given the lack of early trimester data, determining the prevalence of vertical transmission during early pregnancy and assessing the potential risks for fetal morbidity and mortality pose a challenge. Therefore, we analysed the impact of SARS-CoV-2 infection on an endometrial 3D spheroid model system. The 3D spheroids are capable of decidualization and express angiotensin-converting enzyme 2 (ACE2) as well as transmembrane protease serine 2 (TMPRSS2), rendering them susceptible to SARS-CoV-2 infection. Employing this 3D cell model, we identified that SARS-CoV-2 can infect both non-decidualized and decidualized endometrial spheroids. Infection significantly increased the chemokine Monocyte chemoattractant protein-1 (MCP-1) compared to non-infected spheroids. Decidualized spheroids exhibited upregulated Interleukin (IL)-8 levels. Furthermore, RNA sequencing revealed dysregulation of several genes involved in tissue-specific immune response, Fc receptor signalling, angiotensin-activated signalling and actin function. Gene expression changes varied between SARS-CoV-2 infected non-decidualized and decidualized spheroids and genes associated with the innate immune system (CD38, LCN2 and NR4A3) were dysregulated as a potential mechanism for immune evasion of SARS-CoV-2. Altogether, our study demonstrates that endometrial spheroids are a useful model to examine the clinical implications of SARS-CoV-2 vertical transmission, warranting further investigations.
Collapse
Affiliation(s)
- Anna Liu
- Department of Women's Health, Eberhard-Karls University Tuebingen, Calwerstr. 7/6, 72076 Tuebingen, Germany
| | - Natalia Ruetalo
- Institute for Medical Virology, Department of Molecular Virology, University Hospital Tuebingen, Elfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| | - Janet P Raja Xavier
- Department of Women's Health, Eberhard-Karls University Tuebingen, Calwerstr. 7/6, 72076 Tuebingen, Germany
| | - Aditya Kumar Lankapalli
- Ineos Oxford Institute for Antimicrobial Research and Department of Biology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Miguel Camarena-Sainz
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Sara Y Brucker
- Department of Women's Health, Eberhard-Karls University Tuebingen, Calwerstr. 7/6, 72076 Tuebingen, Germany
| | - Yogesh Singh
- Department of Women's Health, Eberhard-Karls University Tuebingen, Calwerstr. 7/6, 72076 Tuebingen, Germany
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Michael Schindler
- Institute for Medical Virology, Department of Molecular Virology, University Hospital Tuebingen, Elfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| | - Madhuri S Salker
- Department of Women's Health, Eberhard-Karls University Tuebingen, Calwerstr. 7/6, 72076 Tuebingen, Germany
| |
Collapse
|
9
|
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
|
10
|
Rosolen V, Beorchia Y, Castriotta L, Fanizza C, Profili F, Floridia M, Giuliano M, Pricci F, Villa M, Grisetti T, Grassi T, Tiple D, Silenzi A, Francesconi P, Bisceglia L, Barbone F, Brusaferro S, Onder G. Insight into potential long COVID effects: Antidepressant use in post SARS-CoV-2 Infection scenarios. A multiregional nested case-control study. Psychiatry Res 2025; 344:116290. [PMID: 39644590 DOI: 10.1016/j.psychres.2024.116290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
This study aimed to investigate the impact of previous SARS-CoV-2 infection and the role of vaccination in the onset of neuropsychiatric conditions, evaluated through antidepressant prescriptions. This case-control study evaluated the risk of new antidepressant prescriptions in relation to previous exposure to SARS-CoV-2 infection and vaccination. It was conducted in three Italian Regions on adults who did not receive antidepressant prescriptions in the year preceding the study period. Individuals with newly prescribed antidepressants (cases) were matched by sex and age to non-users of antidepressants (controls). Pooled estimates of regional Odds Ratios (ORs) were obtained through a meta-analysis. Findings showed that individuals previously infected with SARS-CoV-2 had a higher risk of receiving a new prescription of antidepressants. Moreover, this association was stronger among subjects hospitalized due to infection, and SARS-CoV-2 vaccination may have acted as an effect modifier by reducing that risk. In fact, the pooled OR for receiving a new antidepressant prescription was higher for unvaccinated individuals than for those vaccinated. The results of this study confirm the role of SARS-CoV-2 infection as a risk factor for the onset of neuropsychiatric symptoms. Antidepressant treatment initiation was much more likely after severe COVID-19 infection but vaccination reduced such a risk.
Collapse
Affiliation(s)
- Valentina Rosolen
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Trieste, Italy
| | - Yvonne Beorchia
- Institute of Hygiene and Evaluative Epidemiology, Friuli Centrale University Health Authority, Udine, Italy
| | - Luigi Castriotta
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Trieste, Italy; Institute of Hygiene and Evaluative Epidemiology, Friuli Centrale University Health Authority, Udine, Italy.
| | - Caterina Fanizza
- Strategic Regional Agency for Health and Social Affairs, Apulia Region, Bari, Italy
| | | | | | | | | | - Marika Villa
- Italian National Institute of Health, Rome, Italy
| | | | | | - Dorina Tiple
- Italian National Institute of Health, Rome, Italy
| | | | | | - Lucia Bisceglia
- Strategic Regional Agency for Health and Social Affairs, Apulia Region, Bari, Italy
| | - Fabio Barbone
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Silvio Brusaferro
- Accreditation, Clinical Risk Management, and Health Care Performance Evaluation Udine, Friuli Centrale University Health Authority, Udine, Italy
| | - Graziano Onder
- Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| |
Collapse
|
11
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
12
|
Thakur N, Chakraborty P, Tufariello JM, Basler CF. SARS-CoV-2 Nsp14 binds Tollip and activates pro-inflammatory pathways while downregulating interferon-α and interferon-γ receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628214. [PMID: 39713296 PMCID: PMC11661139 DOI: 10.1101/2024.12.12.628214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
SARS coronavirus 2 (SARS-CoV-2) non-structural protein 14 (Nsp14) possesses an N-terminal exonuclease (ExoN) domain that provides a proofreading function for the viral RNA-dependent RNA polymerase and a C-terminal N7-methyltransferase (N7-MTase) domain that methylates viral mRNA caps. Nsp14 also modulates host functions. This includes the activation of NF-κB and downregulation of interferon alpha/beta receptor 1 (IFNAR1). Here we demonstrate that Nsp14 exerts broader effects, activating not only NF-κB responses but also ERK, p38 and JNK MAP kinase (MAPK) signaling, promoting cytokine production. Further, Nsp14 downregulates not only IFNAR1 but also IFN-γ receptor 1 (IFNGR1), impairing cellular responses to both IFNα and IFNγ. IFNAR1 and IFNGR1 downregulation is via a lysosomal pathway and also occurs in SARS-CoV-2 infected cells. Analysis of a panel of Nsp14 mutants reveals a consistent pattern. Mutants that disable ExoN function remain active, whereas N7-MTase mutations impair both pro-inflammatory pathway activation and IFN receptor downregulation. Innate immune modulating functions also require the presence of both the ExoN and N7-MTase domains likely reflecting the need for the ExoN domain for N7-MTase activity. We further identify multi-functional host protein Tollip as an Nsp14 interactor. Interaction requires the phosphoinositide-binding C2 domain of Tollip and sequences C-terminal to the C2 domain. Full length Tollip or regions encompassing the Nsp14 interaction domain are sufficient to counteract both Nsp14-mediated and Nsp14-independent activation of NF-κB. Knockdown of Tollip partially reverses IFNAR1 and IFNGR1 downregulation in SARS-CoV-2 infected cells, suggesting relevance of Nsp14-Tollip interaction for Nsp14 innate immune evasion functions.
Collapse
Affiliation(s)
- Naveen Thakur
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Poushali Chakraborty
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - JoAnn M. Tufariello
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Christopher F. Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
13
|
Camici M, Del Duca G, Brita AC, Antinori A. Connecting dots of long COVID-19 pathogenesis: a vagus nerve- hypothalamic-pituitary- adrenal-mitochondrial axis dysfunction. Front Cell Infect Microbiol 2024; 14:1501949. [PMID: 39735263 PMCID: PMC11671747 DOI: 10.3389/fcimb.2024.1501949] [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: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
The pathogenesis of long COVID (LC) still presents many areas of uncertainty. This leads to difficulties in finding an effective specific therapy. We hypothesize that the key to LC pathogenesis lies in the presence of chronic functional damage to the main anti-inflammatory mechanisms of our body: the three reflexes mediated by the vagus nerve, the hypothalamic-pituitary-adrenal (HPA) hormonal axis, and the mitochondrial redox status. We will illustrate that this neuro-endocrine-metabolic axis is closely interconnected and how the SARS-CoV-2 can damage it at all stages through direct, immune-inflammatory, epigenetic damage mechanisms, as well as through the reactivation of neurotropic viruses. According to our theory, the direct mitochondrial damage carried out by the virus, which replicates within these organelles, and the cellular oxidative imbalance, cannot be countered in patients who develop LC. This is because their anti-inflammatory mechanisms are inconsistent due to reduced vagal tone and direct damage to the endocrine glands of the HPA axis. We will illustrate how acetylcholine (ACh) and cortisol, with its cytoplasmatic and cellular receptors respectively, are fundamental players in the LC process. Both Ach and cortisol play multifaceted and synergistic roles in reducing inflammation. They achieve this by modulating the activity of innate and cell-mediated immunity, attenuating endothelial and platelet activation, and modulating mitochondrial function, which is crucial for cellular energy production and anti-inflammatory mechanisms. In our opinion, it is essential to study the sensitivity of the glucocorticoids receptor in people who develop LC and whether SARS-CoV-2 can cause long-term epigenetic variations in its expression and function.
Collapse
Affiliation(s)
- Marta Camici
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Del Duca
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Clelia Brita
- Department of Clinical Psychology, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Antinori
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
14
|
Peng Y, Wang Q, Jin F, Tao T, Qin Q. Assessment of urine CCL2 as a potential diagnostic biomarker for acute kidney injury and septic acute kidney injury in intensive care unit patients. Ren Fail 2024; 46:2313171. [PMID: 38345000 PMCID: PMC10863526 DOI: 10.1080/0886022x.2024.2313171] [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: 07/28/2023] [Accepted: 01/27/2024] [Indexed: 02/15/2024] Open
Abstract
Acute kidney injury (AKI) is a prevalent and serious condition in the intensive care unit (ICU), associated with significant morbidity and mortality. Septic acute kidney injury (SAKI) contributes substantially to AKI cases in the ICU. However, current diagnostic methods have limitations, necessitating the exploration of novel biomarkers. In this study, we investigated the potential of plasma and urine CCL2 levels as diagnostic markers for AKI and SAKI in 216 ICU patients. Our findings revealed significant differences in plasma (p < 0.01) and urine CCL2 (p < 0.0001) levels between AKI and non-AKI patients in the ICU. Notably, urine CCL2 demonstrated promising predictive value for AKI, exhibiting high specificity and sensitivity (AUC = 0.8976; p < 0.0001). Furthermore, we observed higher urine CCL2 levels in SAKI compared to non-septic AKI (p < 0.001) and urine CCL2 could also differentiate SAKI from non-septic AKI (AUC = 0.7597; p < 0.0001). These results suggest that urine CCL2 levels hold promise as early biomarkers for AKI and SAKI, offering valuable insights for timely intervention and improved management of ICU patients.
Collapse
Affiliation(s)
- Yuan Peng
- Intensive Care Unit, The First People’s Hospital of Kunshan Affiliated to Jiangsu University, Kunshan, PR China
| | - Qin Wang
- Intensive Care Unit, The First People’s Hospital of Kunshan Affiliated to Jiangsu University, Kunshan, PR China
| | - Fang Jin
- Intensive Care Unit, The First People’s Hospital of Kunshan Affiliated to Jiangsu University, Kunshan, PR China
| | - Tao Tao
- Intensive Care Unit, The First People’s Hospital of Kunshan Affiliated to Jiangsu University, Kunshan, PR China
| | - Qihong Qin
- Department of Emergency, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, PR China
| |
Collapse
|
15
|
Paranga TG, Mitu I, Pavel-Tanasa M, Rosu MF, Miftode IL, Constantinescu D, Obreja M, Plesca CE, Miftode E. Cytokine Storm in COVID-19: Exploring IL-6 Signaling and Cytokine-Microbiome Interactions as Emerging Therapeutic Approaches. Int J Mol Sci 2024; 25:11411. [PMID: 39518964 PMCID: PMC11547016 DOI: 10.3390/ijms252111411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
IL-6 remains a key molecule of the cytokine storms characterizing COVID-19, exerting both proinflammatory and anti-inflammatory effects. Emerging research underscores the significance of IL-6 trans-signaling over classical signaling pathways, which has shifted the focus of therapeutic strategies. Additionally, the synergistic action of TNF-α and IFN-γ has been found to induce inflammatory cell death through PANoptosis, further amplifying the severity of cytokine storms. Long COVID-19 patients, as well as those with cytokine storms triggered by other conditions, exhibit distinct laboratory profiles, indicating the need for targeted approaches to diagnosis and management. Growing evidence also highlights the gut microbiota's crucial role in modulating the immune response during COVID-19 by affecting cytokine production, adding further complexity to the disease's immunological landscape. Targeted intervention strategies should focus on specific cytokine cutoffs, though accurate cytokine quantification remains a clinical challenge. Current treatment strategies are increasingly focused on inhibiting IL-6 trans-signaling, which offers promise for more precise therapeutic approaches to manage hyperinflammatory responses in COVID-19. In light of recent discoveries, this review summarizes key research findings on cytokine storms, particularly their role in COVID-19 and other inflammatory conditions. It explores emerging therapeutic strategies targeting cytokines like IL-6, TNF-α, and IFN-γ, while also addressing open questions, such as the need for better biomarkers to detect and manage cytokine storms. Additionally, the review highlights ongoing challenges in developing targeted treatments that mitigate hyperinflammation without compromising immune function, emphasizing the importance of continued research in this field.
Collapse
Affiliation(s)
- Tudorita Gabriela Paranga
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Ivona Mitu
- Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mariana Pavel-Tanasa
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, 700101 Iasi, Romania
| | - Manuel Florin Rosu
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
- Department of Preventive Medicine and Interdisciplinarity, Faculty of Medicine, University of Medicine and Pharmacy Grigore. T. Popa, 700115 Iasi, Romania
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Daniela Constantinescu
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, 700101 Iasi, Romania
| | - Maria Obreja
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Claudia Elena Plesca
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Egidia Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| |
Collapse
|
16
|
Serwaa A, Oyawoye F, Owusu IA, Dosoo D, Manu AA, Sobo AK, Fosu K, Olwal CO, Quashie PK, Aikins AR. In vitro analysis suggests that SARS-CoV-2 infection differentially modulates cancer-like phenotypes and cytokine expression in colorectal and prostate cancer cells. Sci Rep 2024; 14:24625. [PMID: 39427065 PMCID: PMC11490510 DOI: 10.1038/s41598-024-75718-1] [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: 05/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) reportedly exacerbates cancer outcomes. However, how COVID-19 influences cancer prognosis and development remains poorly understood. Here, we investigated the effect of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), the etiological agent of COVID-19, on cellular cancer phenotypes the expression of cancer-related markers, and various proinflammatory cytokines. We infected prostate (22RV1) and colorectal (DLD-1) cancer cell lines, which express angiotensin-converting enzyme 2 (ACE2), with spike pseudovirus (sPV) and laboratory stocks of live SARS-CoV-2 viruses. After infection, we quantified changes in the cellular cancer phenotypes, the gene expression levels of some cancer markers, including Ki-67, BCL-2, VIM, MMP9, and VEGF, and proinflammatory cytokines. Phenotypic analysis was performed using MTT and wound healing assays, whereas gene expression analysis was carried out using real-time quantitative PCR (RT-qPCR). We show that SARS-CoV-2 infection impacts several key cellular pathways involved in cell growth, apoptosis, and migration, in prostate and colorectal cancer cells. Our results suggest that SARS-CoV-2 infection does influence various cancer cellular phenotypes and expression of molecular cancer markers and proinflammatory cytokines, albeit in a cell-type-specific manner. Our findings hint at the need for further studies and could have implications for evaluating the impact of other viruses on cancer progression.
Collapse
Affiliation(s)
- Alberta Serwaa
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Fatima Oyawoye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Irene Amoakoh Owusu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Aaron Adom Manu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Augustine Kojo Sobo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Fosu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles Ochieng Olwal
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Anastasia Rosebud Aikins
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
17
|
Adonias GL, Siljak H, Balasubramaniam S, Barros MT. In silico modelling of neuron signal impact of cytokine storm-induced demyelination. Open Biol 2024; 14:240138. [PMID: 39226928 PMCID: PMC11371429 DOI: 10.1098/rsob.240138] [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: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/05/2024] Open
Abstract
In this study, we develop an in silico model of a neuron's behaviour under demyelination caused by a cytokine storm to investigate the effects of viral infections in the brain. We use a comprehensive model to measure how cytokine-induced demyelination affects the propagation of action potential (AP) signals within a neuron. We analysed the effects of neuron-neuron communications by applying information and communication theory at different levels of demyelination. Our simulations demonstrate that virus-induced degeneration can play a role in the signal power and spiking rate, which compromise the propagation and processing of information between neurons. We propose a transfer function to model the weakening effects on the AP. Our results show that demyelination induced by a cytokine storm not only degrades the signal but also impairs its propagation within the axon. Our proposed in silico model can analyse virus-induced neurodegeneration and enhance our understanding of virus-induced demyelination.
Collapse
Affiliation(s)
- Geoflly L. Adonias
- Walton Institute for Information and Communication Systems Science, South East Technology University, Waterford, Ireland
| | - Harun Siljak
- Department of Electronic and Electrical Engineering, Trinity College Dublin, Dublin, Ireland
| | | | - Michael Taynnan Barros
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK
| |
Collapse
|
18
|
Liu D, Leung KY, Zhang R, Lam HY, Fan Y, Xie X, Chan KH, Hung IFN. Efficacy of molnupiravir and interferon for the treatment of SARS-CoV-2 in golden Syrian hamster. J Med Virol 2024; 96:e29901. [PMID: 39210614 DOI: 10.1002/jmv.29901] [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: 06/17/2024] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The mortality and hospitalization rate by COVID-19 dropped significantly currently, but its seasonal outbreaks make antiviral treatment still vital. The mortality and hospitazation rate by COVID-19 dropped significantly currently, but its seasonal ourbreaks make antiviral treatment still vital. In our study, syrian golden hamsters were treated with molnupiravir and interferons (IFNs) after SARS-CoV-2 infection. Their weight changes, pathological changes, virus replication and inflammation levels were evaluated. In the IFNs single treatment, only IFN-α group reduced viral load (p < 0.05) and virus titer in hamster lungs. The TNF-α expression decreased significantly in both IFNs treatment at 2dpi. Histological and immunofluorescence results showed lung damage in the IFNs groups were milder at 4dpi. In the molnupiravir/IFN-α combination treatment, weight loss and virus replication in lung were significantly decreased in the mono-molnupiravir group and combination group (p < 0.05), the expression of IL-6, TNF-α, IL-1β and MIP-1α also decreased significantly (p < 0.05), but the combination treatment was not more effective than the mono-molnupiravir treatment. Histological and immunofluorescence results showed the lung damage and inflammation in mono-molnupiravir and combination groups were milder. In summary, IFNs treatment had anti-inflammatory effect against SARS-CoV-2, only IFN-α showed a weak antiviral effect. Molnupiravir/IFN-α combination treatment was effective against SARS-CoV-2 but was not superior to mono-molnupiravir treatment. IFN-α could be considered for immunocompromised patients to stimulate and activate early immune responses.
Collapse
Affiliation(s)
- Danlei Liu
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ka-Yi Leung
- Department of Microbiology, Li Ka Shing faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Hoi-Yan Lam
- Department of Microbiology, Li Ka Shing faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Yujing Fan
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Xiaochun Xie
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing faculty of Medicine, University of Hong Kong, Hong Kong, China
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, Li Ka Shing faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, Li Ka Shing faculty of Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
20
|
Barroso-Arévalo S, Sánchez-Morales L, Porras N, Díaz-Frutos M, Barasona JA, Isla J, López D, Gortázar C, Domínguez L, Sánchez-Vizcaíno JM. Comparative SARS-CoV-2 Omicron BA.5 variant and D614G-Wuhan strain infections in ferrets: insights into attenuation and disease progression during subclinical to mild COVID-19. Front Vet Sci 2024; 11:1435464. [PMID: 39211479 PMCID: PMC11358085 DOI: 10.3389/fvets.2024.1435464] [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: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction As the SARS-CoV-2 virus continues to evolve and new variants emerge, it becomes crucial to understand the comparative pathological and immunological responses elicited by different strains. This study focuses on the original Wuhan strain and the Omicron variant, which have demonstrated significant differences in clinical outcomes and immune responses. Methods We employed ferrets as an experimental model to assess the D614G variant (a derivative of the Wuhan strain) and the Omicron BA.5 variant. Each variant was inoculated into separate groups of ferrets to compare disease severity, viral dissemination, and immune responses. Results The D614G variant induced more severe disease and greater viral spread than the Omicron variant. Notably, ferrets infected with the D614G variant exhibited a robust neutralizing antibody response, whereas those infected with the Omicron variant failed to produce a detectable neutralizing antibody response. Despite the clearance of the virus from nearly all tissues by 7 days post-infection, an increase in pathological lesions was observed from 14 to 21 days, particularly in those infected with the D614G variant, suggesting a sustained immune response even after viral clearance. Discussion These findings underscore the adaptability of SARS-CoV-2 and illuminate how susceptibility and clinical manifestations vary across different strains and species. The results emphasize the necessity of considering both the direct effects of viral infection and the indirect, often prolonged, impacts of the immune response in evaluating the outcomes of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Sandra Barroso-Arévalo
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Lidia Sánchez-Morales
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Néstor Porras
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Marta Díaz-Frutos
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose A. Barasona
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Débora López
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Lucas Domínguez
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose M. Sánchez-Vizcaíno
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
21
|
Adiga V, Bindhu H, Ahmed A, Chetan Kumar N, Tripathi H, D’Souza G, Dias M, Shivalingaiah S, Rao S, K N S, Hawrylowicz C, Dwarkanath P, Vyakarnam A. Immune profiling reveals umbilical cord blood mononuclear cells from South India display an IL-8 dominant, CXCL-10 deficient polyfunctional monocyte response to pathogen-associated molecular patterns that is distinct from adult blood cells. Clin Exp Immunol 2024; 217:263-278. [PMID: 38695079 PMCID: PMC11310697 DOI: 10.1093/cei/uxae034] [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: 11/30/2023] [Revised: 02/28/2024] [Accepted: 05/14/2024] [Indexed: 08/10/2024] Open
Abstract
Neonate responses to pathogen-associated molecular patterns (PAMPS) differ from adults; such understanding is poor in Indian neonates, despite recognized significant infectious risk. Immune profiling analysis was undertaken of 10 secreted mediators contextualized with cellular source induced by six PAMPs in umbilical cord (CB; n = 21) and adult-blood (PBMC; n = 14) from a tertiary care hospital in South India. Differential cytokine expression analysis (minimum log2-fold difference; adj P-value < 0.05) identified bacterial PAMPs induced higher concentrations of IL-1β, IL-10, TNF-α in adults versus IL-8, GM-CSF, IFN-γ, and IL-2 in CB. CB responded to poly I:C and SARS-CoV-2 lysate with a dominant IL-8 response, whereas in PBMC, CXCL-10 dominated poly I:C, but not SARS-CoV-2, responses, highlighting potential IL-8 importance, in the absence of Type I Interferons, in antiviral CB immunity. Candida albicans was the only PAMP to uniformly induce higher secretion of effectors in CB. The predominant source of IL-8/IL-6/TNF-α/IL-1β in both CB and PBMC was polyfunctional monocytes and IFN-γ/IL-2/IL-17 from innate lymphocytes. Correlation matrix analyses revealed IL-8 to be the most differentially regulated, correlating positively in CB versus negatively in PBMC with IL-6, GM-CSF, IFN-γ, IL-2, consistent with more negatively regulated cytokine modules in adults, potentially linked to higher anti-inflammatory IL-10. Cord and adult blood from India respond robustly to PAMPs with unique effector combinations. These data provide a strong foundation to monitor, explore, mechanisms that regulate such immunity during the life course, an area of significant global health importance given infection-related infant mortality incidence.
Collapse
Affiliation(s)
- Vasista Adiga
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Hima Bindhu
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Asma Ahmed
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Nirutha Chetan Kumar
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Himanshu Tripathi
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - George D’Souza
- Department of Pulmonary Medicine, St. John’s Medical College, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | | | - Srishti Rao
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Shanti K N
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Catherine Hawrylowicz
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| | - Pratibha Dwarkanath
- Division of Nutrition, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Annapurna Vyakarnam
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| |
Collapse
|
22
|
Tsuge M, Ichihara E, Hasegawa K, Kudo K, Tanimoto Y, Nouso K, Oda N, Mitsumune S, Kimura G, Yamada H, Takata I, Mitsuhashi T, Taniguchi A, Tsukahara K, Aokage T, Hagiya H, Toyooka S, Tsukahara H, Maeda Y. Increased Oxidative Stress and Decreased Citrulline in Blood Associated with Severe Novel Coronavirus Pneumonia in Adult Patients. Int J Mol Sci 2024; 25:8370. [PMID: 39125944 PMCID: PMC11313210 DOI: 10.3390/ijms25158370] [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: 07/03/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
This study investigated the correlation between oxidative stress and blood amino acids associated with nitric oxide metabolism in adult patients with coronavirus disease (COVID-19) pneumonia. Clinical data and serum samples were prospectively collected from 100 adult patients hospitalized for COVID-19 between July 2020 and August 2021. Patients with COVID-19 were categorized into three groups for analysis based on lung infiltrates, oxygen inhalation upon admission, and the initiation of oxygen therapy after admission. Blood data, oxidative stress-related biomarkers, and serum amino acid levels upon admission were compared in these groups. Patients with lung infiltrations requiring oxygen therapy upon admission or starting oxygen post-admission exhibited higher serum levels of hydroperoxides and lower levels of citrulline compared to the control group. No remarkable differences were observed in nitrite/nitrate, asymmetric dimethylarginine, and arginine levels. Serum citrulline levels correlated significantly with serum lactate dehydrogenase and C-reactive protein levels. A significant negative correlation was found between serum levels of citrulline and hydroperoxides. Levels of hydroperoxides decreased, and citrulline levels increased during the recovery period compared to admission. Patients with COVID-19 with extensive pneumonia or poor oxygenation showed increased oxidative stress and reduced citrulline levels in the blood compared to those with fewer pulmonary complications. These findings suggest that combined oxidative stress and abnormal citrulline metabolism may play a role in the pathogenesis of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Mitsuru Tsuge
- Department of Pediatrics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Eiki Ichihara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama 700-8558, Japan; (E.I.); (A.T.)
| | - Kou Hasegawa
- Department of General Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.H.); (H.H.)
| | - Kenichiro Kudo
- Department of Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-1192, Japan; (K.K.); (S.M.)
| | - Yasushi Tanimoto
- Department of Allergy and Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-0304, Japan; (Y.T.); (G.K.)
| | - Kazuhiro Nouso
- Department of Gastroenterology, Okayama City Hospital, Okayama 700-0962, Japan;
| | - Naohiro Oda
- Department of Internal Medicine, Fukuyama City Hospital, Fukuyama 721-0971, Japan; (N.O.); (I.T.)
| | - Sho Mitsumune
- Department of Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-1192, Japan; (K.K.); (S.M.)
| | - Goro Kimura
- Department of Allergy and Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-0304, Japan; (Y.T.); (G.K.)
| | - Haruto Yamada
- Department of Infectious Disease, Okayama City Hospital, Okayama 700-0962, Japan;
| | - Ichiro Takata
- Department of Internal Medicine, Fukuyama City Hospital, Fukuyama 721-0971, Japan; (N.O.); (I.T.)
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Akihiko Taniguchi
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama 700-8558, Japan; (E.I.); (A.T.)
| | - Kohei Tsukahara
- Department of Emergency, Critical Care and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.T.); (T.A.)
| | - Toshiyuki Aokage
- Department of Emergency, Critical Care and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.T.); (T.A.)
| | - Hideharu Hagiya
- Department of General Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.H.); (H.H.)
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrine Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Hirokazu Tsukahara
- Department of Pediatrics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| |
Collapse
|
23
|
Yin Z, Zhang J, Qin J, Guo L, Guo Q, Kang W, Ma C, Chen L. Anti-inflammatory properties of polysaccharides from edible fungi on health-promotion: a review. Front Pharmacol 2024; 15:1447677. [PMID: 39130633 PMCID: PMC11310034 DOI: 10.3389/fphar.2024.1447677] [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: 06/12/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Edible fungus polysaccharides have garnered significant attention from scholars due to their safety and potential anti-inflammatory activity. However, comprehensive summaries of their anti-inflammatory properties are still rare. This paper provides a detailed overview of the anti-inflammatory effects and mechanisms of these polysaccharides, as well as their impact on inflammation-related diseases. Additionally, the relationship between their structure and anti-inflammatory activity is discussed. It is believed that this review will greatly enhance the understanding of the application of edible fungus polysaccharides in anti-inflammatory treatments, thereby significantly promoting the development and utilization of edible fungi.
Collapse
Affiliation(s)
- Zhenhua Yin
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Juanjuan Zhang
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Jingjing Qin
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Lin Guo
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Qingfeng Guo
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Wenyi Kang
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Lin Chen
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| |
Collapse
|
24
|
Negaresh M, Ghobadi H, Hoseininia S, Samadi Takaldani AH, Javanshir N, Iranijam E, Aslani MR. Evaluation of the Efficacy of Therapeutic and Prophylactic Anticoagulation in COVID-19 Patients With Venous Catheter and Its Correlation With Clinical Outcomes. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2024; 32. [DOI: 10.1097/ipc.0000000000001382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
IntroductionThe COVID-19 disease was first detected in December 2019, and since then, various treatments have been used to manage it. One such treatment method is therapeutic plasma exchange. This method involves implanting a venous catheter, which increases the risk of venous thromboembolism (VTE). Other risk factors for VTE include infections like COVID-19, inflammation, or malignancy.Materials and MethodsIn this retrospective study, patients with acute respiratory syndrome caused by COVID-19 who were hospitalized and had venous catheters inserted for therapeutic plasma exchange were enrolled. The prophylactic anticoagulant dose was started for all patients, and after the diagnosis of VTE, it was changed to the therapeutic dose. Patients' information, including demographic data, clinical information, and laboratory findings, was extracted from patients' records and recorded in a checklist designed for each patient.ResultsFrom a total of 168 patients, 26 were diagnosed with VTE (pulmonary embolism in 5 patients and deep vein thrombosis in 21 patients). The prevalence of VTE in COVID-19 patients with the venous catheter was 15.4%. The right femoral vein was the most used route for catheterization and had the highest occurrence of venous thromboses. The patients diagnosed with thrombosis showed a lower mortality rate, higher D-dimer and lactate dehydrogenase levels, and lower platelet counts.ConclusionsThis study showed a higher risk of VTE and subclinical thrombosis in COVID-19 patients with venous catheters. Continuous screening, higher doses of anticoagulants, and early removal of venous catheters are critical in preventing VTE and mortality.
Collapse
Affiliation(s)
- Mohammad Negaresh
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hassan Ghobadi
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saeed Hoseininia
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Hossein Samadi Takaldani
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Javanshir
- Faculty of Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Effat Iranijam
- Department of Internal Medicine (Hematology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
25
|
Bhuvaneshwar K, Madhavan S, Gusev Y. Integrative genomic analysis of the lung tissue microenvironment in SARS-CoV-2 and NL63 patients. Heliyon 2024; 10:e32772. [PMID: 39183848 PMCID: PMC11341340 DOI: 10.1016/j.heliyon.2024.e32772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/28/2024] [Accepted: 06/09/2024] [Indexed: 08/27/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has affected over 700 million people, and caused over 7 million deaths throughout the world as of April 2024, and continues to affect people through seasonal waves. While over 675 million people have recovered from this disease globally, the lingering effects of the disease are still under study. Long term effects of SARS-CoV-2 infection, known as 'long COVID,' include a wide range of symptoms including fatigue, chest pain, cellular damage, along with a strong innate immune response characterized by inflammatory cytokine production. Three years after the pandemic, data about long covid studies are finally emerging. More clinical studies and clinical trials are needed to understand and determine the factors that predispose individuals to these long-term side effects. In this methodology paper, our goal was to apply data driven approaches in order to explore the multidimensional landscape of infected lung tissue microenvironment to better understand complex interactions between viral infection, immune response and the lung microbiome of patients with (a) SARS-CoV-2 virus and (b) NL63 coronavirus. The samples were analyzed with several machine learning tools allowing simultaneous detection and quantification of viral RNA amount at genome and gene level; human gene expression and fractions of major types of immune cells, as well as metagenomic analysis of bacterial and viral abundance. To contrast and compare specific viral response to SARS-COV-2, we analyzed deep sequencing data from additional cohort of patients infected with NL63 strain of corona virus. Our correlation analysis of three types of RNA-seq based measurements in patients i.e. fraction of viral RNA (at genome and gene level), Human RNA (transcripts and gene level) and bacterial RNA (metagenomic analysis), showed significant correlation between viral load as well as level of specific viral gene expression with the fractions of immune cells present in lung lavage as well as with abundance of major fractions of lung microbiome in COVID-19 patients. Our methodology-based proof-of-concept study has provided novel insights into complex regulatory signaling interactions and correlative patterns between the viral infection, inhibition of innate and adaptive immune response as well as microbiome landscape of the lung tissue. These initial findings could provide better understanding of the diverse dynamics of immune response and the side effects of the SARS-CoV-2 infection and demonstrates the possibilities of the various types of analyses that could be performed from this type of data.
Collapse
Affiliation(s)
- Krithika Bhuvaneshwar
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Subha Madhavan
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Yuriy Gusev
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| |
Collapse
|
26
|
Prasanth MI, Wannigama DL, Reiersen AM, Thitilertdecha P, Prasansuklab A, Tencomnao T, Brimson S, Brimson JM. A systematic review and meta-analysis, investigating dose and time of fluvoxamine treatment efficacy for COVID-19 clinical deterioration, death, and Long-COVID complications. Sci Rep 2024; 14:13462. [PMID: 38862591 PMCID: PMC11166997 DOI: 10.1038/s41598-024-64260-9] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
There have been 774,075,242 cases of COVID-19 and 7,012,986 deaths worldwide as of January 2024. In the early stages of the pandemic, there was an urgent need to reduce the severity of the disease and prevent the need for hospitalization to avoid stress on healthcare systems worldwide. The repurposing of drugs to prevent clinical deterioration of COVID-19 patients was trialed in many studies using many different drugs. Fluvoxamine (an SSRI and sigma-1 receptor agonist) was initially identified to potentially provide beneficial effects in COVID-19-infected patients, preventing clinical deterioration and the need for hospitalization. Fourteen clinical studies have been carried out to date, with seven of those being randomized placebo-controlled studies. This systematic review and meta-analysis covers the literature from the outbreak of SARS-CoV-2 in late 2019 until January 2024. Search terms related to fluvoxamine, such as its trade names and chemical names, along with words related to COVID-19, such as SARS-CoV-2 and coronavirus, were used in literature databases including PubMed, Google Scholar, Scopus, and the ClinicalTrials.gov database from NIH, to identify the trials used in the subsequent analysis. Clinical deterioration and death data were extracted from these studies where available and used in the meta-analysis. A total of 7153 patients were studied across 14 studies (both open-label and double-blind placebo-controlled). 681 out of 3553 (19.17%) in the standard care group and 255 out of 3600 (7.08%) in the fluvoxamine-treated group experienced clinical deterioration. The estimated average log odds ratio was 1.087 (95% CI 0.200 to 1.973), which differed significantly from zero (z = 2.402, p = 0.016). The seven placebo-controlled studies resulted in a log odds ratio of 0.359 (95% CI 0.1111 to 0.5294), which differed significantly from zero (z = 3.103, p = 0.002). The results of this study identified fluvoxamine as effective in preventing clinical deterioration, and subgrouping analysis suggests that earlier treatment with a dose of 200 mg or above provides the best outcomes. We hope the outcomes of this study can help design future studies into respiratory viral infections and potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Thai Red Cross Society, Bangkok, Thailand
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Angela Michelle Reiersen
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Rama 1 Road, Pathumwan, Wang Mai, Bangkok, 10330, Thailand.
| |
Collapse
|
27
|
Ulloque-Badaracco JR, Al-Kassab-Córdova A, Alarcon-Braga EA, Hernandez-Bustamante EA, Huayta-Cortez MA, Cabrera-Guzmán JC, Robles-Valcarcel P, Benites-Zapata VA. Association of vitamin B12, folate, and homocysteine with COVID-19 severity and mortality: A systematic review and meta-analysis. SAGE Open Med 2024; 12:20503121241253957. [PMID: 38774742 PMCID: PMC11107318 DOI: 10.1177/20503121241253957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 04/24/2024] [Indexed: 05/24/2024] Open
Abstract
Objective We aimed to review the available evidence on the association between vitamin B12, folate, and homocysteine levels with worse outcomes among COVID-19 patients. Methods The search was carried out in ten databases simultaneously run on 10 May 2023, without language restrictions. We included cross-sectional, case-control, and cohort studies. The random-effects meta-analysis was performed using the Sidik-Jonkman method and corrected 95% confidence intervals using the truncated Knapp-Hartung standard errors. Standardized mean difference and 95% CI was used as the measure effect size. Results Thirteen articles were included in this review (n = 2134). Patients with COVID-19 who did not survive had the highest serum vitamin B12 values (SMD: 1.05; 95% CI: 0.31-1.78; p = 0.01, I2 = 91.22%). In contrast, low serum folate values were associated with patients with severe COVID-19 (SMD: -0.77; 95% CI: -1.35 to -0.19; p = 0.02, I2 = 59.09%). The remaining tested differences did not yield significant results. Conclusion Elevated serum levels of vitamin B12 were associated with higher mortality in patients with COVID-19. Severe cases of COVID-19 were associated with low serum folate levels. Future studies should incorporate a larger sample size.
Collapse
Affiliation(s)
| | - Ali Al-Kassab-Córdova
- Centro de Excelencia en Investigaciones Económicas y Sociales en Salud, Universidad San Ignacio de Loyola, Lima, Peru
| | | | - Enrique A Hernandez-Bustamante
- Sociedad Científica de Estudiantes de Medicina de la Universidad Nacional de Trujillo, Trujillo, Peru
- Grupo Peruano de Investigación Epidemiológica, Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima, Peru
| | | | - Juan C Cabrera-Guzmán
- Facultad de Ciencias de La Salud, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | | | - Vicente A Benites-Zapata
- Unidad de Investigación Para la Generación y Síntesis de Evidencias en Salud, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima, Peru
| |
Collapse
|
28
|
Taefehshokr N, Lac A, Vrieze AM, Dickson BH, Guo PN, Jung C, Blythe EN, Fink C, Aktar A, Dikeakos JD, Dekaban GA, Heit B. SARS-CoV-2 NSP5 antagonizes MHC II expression by subverting histone deacetylase 2. J Cell Sci 2024; 137:jcs262172. [PMID: 38682259 PMCID: PMC11166459 DOI: 10.1242/jcs.262172] [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: 04/01/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.
Collapse
Affiliation(s)
- Nima Taefehshokr
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Alex Lac
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Angela M. Vrieze
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Brandon H. Dickson
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Peter N. Guo
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Catherine Jung
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Eoin N. Blythe
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Corby Fink
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Amena Aktar
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Gregory A. Dekaban
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Bryan Heit
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| |
Collapse
|
29
|
Song J, Jeong S, Choi S, Oh YH, Park SJ, Chang JY, Cho Y, Byeon K, Choi JY, Lee S, Jung J, Park SM. Inverse association with COVID-19 vaccination status of the incidence of pneumonia after SARS-CoV-2 infection: A nationwide retrospective cohort study. J Infect Public Health 2024; 17:650-656. [PMID: 38430718 DOI: 10.1016/j.jiph.2024.02.005] [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: 03/12/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Although one of the characteristics of COVID-19 is accompanied by acute pneumonia immediately after infection, large-scale cohort studies focused on this issue are lacking. In addition, there is interest in how COVID-19 vaccinations reduce the incidence of acute pneumonia for people infected with different strains of SARS-CoV-2. Thus, we assess the short-term incidence of pneumonia after COVID-19 with the vaccination and SARS-CoV-2 variants. METHODS As data for 2136,751 COVID-19 patients between January 01, 2020 and February 28, 2022 was collected, they were observed for one month from the day of infection. Patients in retrospective cohort study were classified according to doses of the received vaccine and the epidemic phase when SARS-CoV-2 variants prevailed. Multivariable logistic regression analysis calculated adjusted odds ratios (aOR) and 95% confidence intervals (CIs) for the pneumonia risk. RESULTS In B.1.1.7-B.1.351, B.1.617.2, and B.1.617.2 variants, the aORs (95% CIs; p-value) for incidence of pneumonia were 0.93 (0.89-0.98; <0.001), 0.74 (0.70-0.78; <0.001), and 0.04 (0.038-0.043; <0.001), respectively, compared to the original strain. More than 80% of patients have received the second and more doses of the vaccine (average age=44.67 years). The aORs (95% CIs; p-value) for pneumonia were 0.61 (0.58-0.64; <0.001), 0.39 (0.38-0.40; <0.001), and 0.18 (0.166-0.184; <0.001) in patients who received the first (N = 68,216), second (N = 898,838), and ≥ third doses (N = 836,173), respectively, compared to unvaccinated patients. According to the received vaccine (second dose of mRNA or viral vector), those who received BNT162b2 and mRNA-1273 (N = 787,980) had lower risk of pneumonia, compared to that in those who received h ChAdOx1 nCov-19 and AD26. COV2-S (N = 89,024). CONCLUSIONS Our findings suggest that the second and ≥ third doses (61% and 82% of risk aversion effect increased, respectively) of the COVID-19 vaccine can prevent the COVID-19-related pneumonia, regardless of the variants.
Collapse
Affiliation(s)
- Jihun Song
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seulggie Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun Hwan Oh
- Department of Family Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong©, Republic of Korea
| | - Sun Jae Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Joo Young Chang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Yoosun Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, School of Medicine, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyeonghyang Byeon
- Associate Research Fellow, Big Data Department, National Health Insurance Service, Wonju, Republic of Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seju Lee
- Division of Infectious Diseases, Department of Internal Medicine, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jaehun Jung
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon 21556, Republic of Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Department of Family Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Liu R, Natekar JP, Kim KH, Pathak H, Bhatnagar N, Raha JR, Park BR, Guglani A, Shin CH, Kumar M, Kang SM. Multivalent and Sequential Heterologous Spike Protein Vaccinations Effectively Induce Protective Humoral Immunity against SARS-CoV-2 Variants. Vaccines (Basel) 2024; 12:362. [PMID: 38675744 PMCID: PMC11053539 DOI: 10.3390/vaccines12040362] [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: 03/06/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to cause challenging problems for the effective control of COVID-19. In this study, we tested the hypothesis of whether a strategy of multivalent and sequential heterologous spike protein vaccinations would induce a broader range and higher levels of neutralizing antibodies against SARS-CoV-2 variants and more effective protection than homologous spike protein vaccination in a mouse model. We determined spike-specific IgG, receptor-binding inhibition titers, and protective efficacy in the groups of mice that were vaccinated with multivalent recombinant spike proteins (Wuhan, Delta, Omicron), sequentially with heterologous spike protein variants, or with homologous spike proteins. Trivalent (Wuhan + Delta + Omicron) and sequential heterologous spike protein vaccinations were more effective in inducing serum inhibition activities of receptor binding to spike variants and virus neutralizing antibody titers than homologous spike protein vaccination. The higher efficacy of protection was observed in mice with trivalent and sequential heterologous spike protein vaccination after a challenge with a mouse-adapted SARS-CoV-2 MA10 strain compared to homologous spike protein vaccination. This study provides evidence that a strategy of multivalent and sequential heterologous variant spike vaccination might provide more effective protection against emerging SARS-CoV-2 variants than homologous spike vaccination and significantly alleviate severe inflammation due to COVID-19.
Collapse
Affiliation(s)
- Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Janhavi P. Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Heather Pathak
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Anchala Guglani
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| |
Collapse
|
31
|
Yu J, Li Y, Xiao L, Xie J, Guo Z, Ye Y, Lin Y, Cao Y, Wu X, Mao C, Li X, Pan M, Ye J, Zhou L, Huang J, Yang J, Wei Y, Zhang X, Zhang B, Kang R. Neglected Spleen Transcriptional Profile Reveals Inflammatory Disorder Conferred by Rabbit Hemorrhagic Disease Virus 2 Infection. Viruses 2024; 16:495. [PMID: 38675838 PMCID: PMC11054208 DOI: 10.3390/v16040495] [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: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Rabbit hemorrhagic disease (RHD) is an acute fatal disease caused by the rabbit hemorrhagic disease virus (RHDV). Since the first outbreaks of type 2 RHDV (RHDV2) in April 2020 in China, the persistence of this virus in the rabbit population has caused substantial economic losses in rabbit husbandry. Previous failures in preventing RHDV2 prompted us to further investigate the immune mechanisms underlying the virus's pathogenicity, particularly concerning the spleen, a vital component of the mononuclear phagocyte system (MPS). For this, a previous RHDV2 isolate, CHN/SC2020, was utilized to challenge naive adult rabbits. Then, the splenic transcriptome was determined by RNA-Seq. This study showed that the infected adult rabbits had 3148 differentially expressed genes (DEGs), which were associated with disease, signal transduction, cellular processes, and cytokine signaling categories. Of these, 100 upregulated DEGs were involved in inflammatory factors such as IL1α, IL-6, and IL-8. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that these DEGs were significantly enriched in the cytokine-cytokine receptor interaction signaling pathway, which may play a vital role in CHN/SC2020 infection. At the same time, proinflammatory cytokines and chemokines were significantly increased in the spleen at the late stages of infection. These findings suggested that RHDV2 (CHN/SC2020) might induce dysregulation of the cytokine network and compromise splenic immunity against viral infection, which expanded our understanding of RHDV2 pathogenicity.
Collapse
Affiliation(s)
- Jifeng Yu
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Yan Li
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (L.Z.); (J.H.); (B.Z.)
| | - Lu Xiao
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Jing Xie
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Zhiqiang Guo
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Yonggang Ye
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Yi Lin
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Ye Cao
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Xuejing Wu
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Congjian Mao
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Xingyu Li
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Meng Pan
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Jianqiang Ye
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Long Zhou
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (L.Z.); (J.H.); (B.Z.)
| | - Jian Huang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (L.Z.); (J.H.); (B.Z.)
| | - Junyan Yang
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Yong Wei
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Xianhui Zhang
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| | - Bin Zhang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (L.Z.); (J.H.); (B.Z.)
| | - Runmin Kang
- Sichuan Provincial Key Laboratory of Animal Breeding and Genetics, Sichuan Animal Science Academy, Chengdu 610066, China; (J.Y.); (L.X.); (J.X.); (Z.G.); (Y.Y.); (Y.L.); (Y.C.); (X.W.); (C.M.); (X.L.); (M.P.); (J.Y.); (J.Y.); (Y.W.); (X.Z.)
| |
Collapse
|
32
|
Healey AM, Fenner KN, O'Dell CT, Lawrence BP. Aryl hydrocarbon receptor activation alters immune cell populations in the lung and bone marrow during coronavirus infection. Am J Physiol Lung Cell Mol Physiol 2024; 326:L313-L329. [PMID: 38290163 PMCID: PMC11281796 DOI: 10.1152/ajplung.00236.2023] [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: 07/26/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Respiratory viral infections are one of the major causes of illness and death worldwide. Symptoms associated with respiratory infections can range from mild to severe, and there is limited understanding of why there is large variation in severity. Environmental exposures are a potential causative factor. The aryl hydrocarbon receptor (AHR) is an environment-sensing molecule expressed in all immune cells. Although there is considerable evidence that AHR signaling influences immune responses to other immune challenges, including respiratory pathogens, less is known about the impact of AHR signaling on immune responses during coronavirus (CoV) infection. In this study, we report that AHR activation significantly altered immune cells in the lungs and bone marrow of mice infected with a mouse CoV. AHR activation transiently reduced the frequency of multiple cells in the mononuclear phagocyte system, including monocytes, interstitial macrophages, and dendritic cells in the lung. In the bone marrow, AHR activation altered myelopoiesis, as evidenced by a reduction in granulocyte-monocyte progenitor cells and an increased frequency of myeloid-biased progenitor cells. Moreover, AHR activation significantly affected multiple stages of the megakaryocyte lineage. Overall, these findings indicate that AHR activation modulates multiple aspects of the immune response to a CoV infection. Given the significant burden of respiratory viruses on human health, understanding how environmental exposures shape immune responses to infection advances our knowledge of factors that contribute to variability in disease severity and provides insight into novel approaches to prevent or treat disease.NEW & NOTEWORTHY Our study reveals a multifaceted role for aryl hydrocarbon receptor (AHR) signaling in the immune response to coronavirus (CoV) infection. Sustained AHR activation during in vivo mouse CoV infection altered the frequency of mature immune cells in the lung and modulated emergency hematopoiesis, specifically myelopoiesis and megakaryopoiesis, in bone marrow. This provides new insight into immunoregulation by the AHR and extends our understanding of how environmental exposures can impact host responses to respiratory viral infections.
Collapse
Affiliation(s)
- Alicia M Healey
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Kristina N Fenner
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Colleen T O'Dell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| |
Collapse
|
33
|
Shyamsundar S, Pierson SK, Connolly CM, Teles M, Segev DL, Werbel WA, van Rhee F, Casper C, Brandstadter JD, Noy A, Fajgenbaum DC. Castleman disease patients report mild COVID-19 symptoms and mount a humoral response to SARS-CoV-2 vaccination. BLOOD NEOPLASIA 2024; 1:100002. [PMID: 39044861 PMCID: PMC11265787 DOI: 10.1016/j.bneo.2024.100002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The coronavirus disease of 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in increased morbidity and mortality in patients with impaired immunity, hematologic malignancies, and immunosuppressive regimens. COVID-19 can cause a cytokine storm with some patients benefiting from blockade of the pro-inflammatory cytokine, interleukin 6 (IL6). As Castleman disease (CD) is an atypical lymphoproliferative disorder that can involve a cytokine storm and often requires immunosuppressive therapies, including IL6 inhibition, we sought to evaluate outcomes following COVID-19 and SARS-CoV-2 vaccination in CD patients. We administered a survey in April 2021 to characterize experiences with COVID-19 and SARS-CoV-2 vaccination among 300 CD patients enrolled in ACCELERATE, a natural history registry of CD patients. Among 128 respondents, the prevalence of SARS-CoV-2 infection (16/95, 17%), severe disease (1/16, 6%), vaccination rates (112/128, 88%), and vaccine adverse effects after dose one (62/112, 55%) were comparable to the general U.S. population. While there were two cases of CD flares occurring shortly after SARS-CoV-2 infection (N=1) and vaccination (N=1), over 100 patients in this study that were infected and/or vaccinated did not experience CD flares. The median anti-spike titer six months after the second dose among CD patients was comparable to individuals with other immune-related diseases and healthy populations. Data from this small cohort suggest that, despite being on immunosuppressive therapies, CD patients do not appear to be at increased risk of poor COVID-19 outcomes and can mount a humoral response to SARS-CoV-2 vaccination. This study was registered on clinicaltrials.gov (#NCT02817997).
Collapse
Affiliation(s)
- Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila K. Pierson
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Caoilfhionn M. Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Mayan Teles
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Dorry L. Segev
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - William A. Werbel
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Corey Casper
- Access to Advanced Health Institute, Seattle, WA, 98102, USA
- Department of Global Health, University of Washington, Seattle, WA, 98105, USA
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joshua D. Brandstadter
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ariela Noy
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY, 10065, USA
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
34
|
Teixeira DG, Rodrigues-Neto JF, da Cunha DCS, Jeronimo SMB. Understanding SARS-CoV-2 spike glycoprotein clusters and their impact on immunity of the population from Rio Grande do Norte, Brazil. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 118:105556. [PMID: 38242186 DOI: 10.1016/j.meegid.2024.105556] [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: 11/15/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
SARS-CoV-2 genome underwent mutations since it started circulating within the human population. The aim of this study was to understand the fluctuation of the spike clusters concomitant to the population immunity either due to natural infection and/or vaccination in a state of Brazil that had both high rate of natural infection and vaccination coverage. A total of 1725 SARS-CoV-2 sequences from the state of Rio Grande do Norte, Brazil, were retrieved from GISAID and subjected to cluster analysis. Immunoinformatics were used to predict T- and B-cell epitopes, followed by simulation to estimate either pro- or anti-inflammatory responses and to correlate with circulating variants. From March 2020 to June 2022, the state of Rio Grande do Norte reported 579,931 COVID-19 cases with a 1.4% fatality rate across the three major waves: May-Sept 2020, Feb-Aug 2021, and Jan-Mar 2022. Cluster 0 variants (wild type strain, Zeta) were prevalent in the first wave and Delta (AY.*), which circulated in Brazil in the latter half of 2021, featuring fewer unique epitopes. Cluster 1 (Gamma (P.1 + P.1.*)) dominated the first half of 2021. Late 2021 had two new clusters, Cluster 2 (Omicron, (B.1.1.529 + BA.*)), and Cluster 3 (BA.*) with the most unique epitopes, in addition to Cluster 4 (Delta sub lineages) which emerged in the second half of 2021 with fewer unique epitopes. Cluster 1 epitopes showed a high pro-inflammatory propensity, while others exhibited a balanced cytokine induction. The clustering method effectively identified Spike groups that may contribute to immune evasion and clinical presentation, and explain in part the clinical outcome.
Collapse
Affiliation(s)
- Diego Gomes Teixeira
- Instituto de Medicina Tropical do Rio Grande do Norte, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - João Firmino Rodrigues-Neto
- Instituto de Medicina Tropical do Rio Grande do Norte, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil; Escola Multicampi de Ciências Médicas do Rio Grande do Norte, Universidade Federal do Rio Grande do Norte, Caicó, Rio Grande do Norte, Brazil
| | - Dayse Caroline Severiano da Cunha
- Instituto de Medicina Tropical do Rio Grande do Norte, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Selma Maria Bezerra Jeronimo
- Instituto de Medicina Tropical do Rio Grande do Norte, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil; Departmento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande Norte, Natal, Rio Grande do Norte, Brazil; Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
35
|
Ajami M, Sotoudeheian M, Houshiar-Rad A, Esmaili M, Naeini F, Mohammadi Nasrabadi F, Doaei S, Milani-Bonab A. Quercetin may reduce the risk of developing the symptoms of COVID-19. AVICENNA JOURNAL OF PHYTOMEDICINE 2024; 14:189-201. [PMID: 38966631 PMCID: PMC11221767 DOI: 10.22038/ajp.2023.22920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 07/06/2024]
Abstract
Objective Recent evidence reported that some dietary compounds like quercetin and apigenin as the most well-known flavonoids with anti-inflammatory effects may inhibit SARS-CoV-2 main protease. The hypothesis of the promising effects and possible mechanisms of action of quercetin against COVID-19 were assessed in this article. Materials and Methods Related papers on the inhibitory effects of quercetin against COVID-19 were collected using the following search strategy: "corona or coronavirus or COVID or COVID-19 or viral or virus" AND "nutrient or flavonoid or Quercetin". Results The findings indicated that quercetin can be considered an effective agent against COVID-19 because of its SARS-CoV-2 main protease and RNA-dependent RNA polymerase inhibitory effects. In addition, quercetin may attenuate angiotensin-converting enzyme-2 (ACE-2) receptors leading to a reduction of SARS-CoV-2 ability to enter host cells. Moreover, the antiviral, anti-inflammatory, and immunomodulatory activities of quercetin have been frequently reported. Conclusion Quercetin may be an effective agent for managing the complications of COVID-19. Further longitudinal human studies are warranted.
Collapse
Affiliation(s)
- Marjan Ajami
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Anahita Houshiar-Rad
- Department of Nutrition Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Esmaili
- Department of Nutrition Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran university of Medical Science, Tehran, Iran
| | - Fatemeh Mohammadi Nasrabadi
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saied Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Milani-Bonab
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Aldous C, Dancis BM, Dancis J, Oldfield PR. Wheel Replacing Pyramid: Better Paradigm Representing Totality of Evidence-Based Medicine. Ann Glob Health 2024; 90:17. [PMID: 38435471 PMCID: PMC10906340 DOI: 10.5334/aogh.4341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/20/2024] [Indexed: 03/05/2024] Open
Abstract
Background Evidence-based medicine (EBM), as originally conceived, used all types of peer-reviewed evidence to guide medical practice and decision-making. During the SARS-CoV-2 Coronavirus disease (COVID-19) pandemic, the standard usage of EBM, modeled by the Evidence-Based Medicine Pyramid, undermined EBM by incorrectly using pyramid levels to assign relative quality. The resulting pyramid-based thinking is biased against reports both in levels beneath randomized control trials (RCTs) and those omitted from the pyramid entirely. Thus, much of the evidence was ignored. Our desire for a more encompassing and effective medical decision-making process to apply to repurposed drugs led us to develop an alternative to the EBM Pyramid for EBM. Herein, we propose the totality of evidence (T-EBM) wheel. Objectives To create an easily understood graphic that models EBM by incorporating all peer-reviewed evidence that applies to both new and repurposed medicines, and to demonstrate its potential utility using ivermectin as a case study. Methods The graphics were produced using Microsoft Office Visio Professional 2003 except for part of the T-EBM wheel sunburst chart, which was produced using Microsoft 365 Excel. For the case study, PubMed® was used by searching for peer-reviewed reports containing "ivermectin" and either "covid" or "sars" in the title. Reports were filtered for those using ivermectin-based protocols in the treatment of COVID-19. The resulting 265 reports were evaluated for their study design types and treatment outcomes. The three-ringed graphical T-EBM wheel was composed of two inner rings showing all types of reports and an outer ring showing outcomes for each type. Findings-Conclusions The T-EBM wheel avoids the biases of the EBM Pyramid and includes all types of reports in the pyramid along with reports such as population and mechanistic studies. In both early and late stages of medical emergencies, pyramid-based thinking may overlook indications of efficacy in regions of the T-EBM wheel beyond RCTs. This is especially true when searching for ways to prevent and treat a novel disease with repurposed therapeutics before RCTs, safety assessments, and mechanisms of action of novel therapeutics are established. As such, T-EBM Wheels should replace the EBM Pyramids in medical decision-making and education. T-EBM Wheels can be expanded upon by implementing multiple outer rings, one for each different kind of outcome (efficacy, safety, etc.). A T-EBM Wheel can be created for any proprietary or generic medicine. The ivermectin (IVM) T-EBM Wheel displays the efficacy of IVM-based treatments of COVID-19 in a color-coded graphic, visualizing each type of evidence and the proportions of each of their outcomes (positive, inconclusive, negative).
Collapse
Affiliation(s)
- Colleen Aldous
- Nelson R. Mandela School of Clinical Medicine of the University of KwaZulu-Natal, Durban, South Africa
| | | | - Jerome Dancis
- Department of Mathematics, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
37
|
Sakaibara M, Yamamoto H, Murota H, Monma N, Sato S, Hirano-Iwata A. Enhanced responses to inflammatory cytokine interleukin-6 in micropatterned networks of cultured cortical neurons. Biochem Biophys Res Commun 2024; 695:149379. [PMID: 38159413 DOI: 10.1016/j.bbrc.2023.149379] [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: 11/28/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
Cortical neurons in dissociated cultures are an indispensable model system for pharmacological research that provides insights into chemical responses in well-defined environments. However, cortical neurons plated on homogeneous substrates develop an unstructured network that exhibits excessively synchronized activity, which occasionally masks the consequences induced by external substances. Here, we show that hyperactivity and excessive synchrony in cultured cortical networks can be effectively suppressed by growing neurons in microfluidic devices. These devices feature a hierarchically modular design that resembles the in vivo network. We focused on interleukin-6, a pro-inflammatory cytokine, and assessed its acute and chronic effects. Fluorescence calcium imaging of spontaneous neural activity for up to 20 days of culture showed detectable modulation of collective activity events and neural correlation in micropatterned neurons, which was not apparent in neurons cultured on homogeneous substrates. Our results indicate that engineered neuronal networks provide a unique platform for detecting and understanding the fundamental effects of biochemical compounds on neuronal networks.
Collapse
Affiliation(s)
- Mamoru Sakaibara
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Hideaki Yamamoto
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan.
| | - Hakuba Murota
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Nobuaki Monma
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Shigeo Sato
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Ayumi Hirano-Iwata
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan; Advanced Institute for Materials Research, Tohoku University, Sendai, Japan; Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
38
|
Harriott NC, Ryan AL. Proteomic profiling identifies biomarkers of COVID-19 severity. Heliyon 2024; 10:e23320. [PMID: 38163173 PMCID: PMC10755324 DOI: 10.1016/j.heliyon.2023.e23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
SARS-CoV-2 infection remains a major public health concern, particularly for the aged and those individuals with co-morbidities at risk for developing severe COVID-19. Understanding the pathogenesis and biomarkers associated with responses to SARS-CoV-2 infection remain critical components in developing effective therapeutic approaches, especially in cases of severe and long-COVID-19. In this study blood plasma protein expression was compared in subjects with mild, moderate, and severe COVID-19 disease. Evaluation of an inflammatory protein panel confirms upregulation of proteins including TNFβ, IL-6, IL-8, IL-12, already associated with severe cytokine storm and progression to severe COVID-19. Importantly, we identify several proteins not yet associated with COVID-19 disease, including mesothelin (MSLN), that are expressed at significantly higher levels in severe COVID-19 subjects. In addition, we find a subset of markers associated with T-cell and dendritic cell responses to viral infection that are significantly higher in mild cases and decrease in expression as severity of COVID-19 increases, suggesting that an immediate and effective activation of T-cells is critical in modulating disease progression. Together, our findings identify new targets for further investigation as therapeutic approaches for the treatment of SARS-CoV-2 infection and prevention of complications of severe COVID-19.
Collapse
Affiliation(s)
- Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| |
Collapse
|
39
|
Chau CW, Sugimura R. Organoids in COVID-19: can we break the glass ceiling? J Leukoc Biol 2024; 115:85-99. [PMID: 37616269 DOI: 10.1093/jleuko/qiad098] [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: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
COVID-19 emerged in September 2020 as a disease caused by the virus SARS-CoV-2. The disease presented as pneumonia at first but later was shown to cause multisystem infections and long-term complications. Many efforts have been put into discovering the exact pathogenesis of the disease. In this review, we aim to discuss an emerging tool in disease modeling, organoids, in the investigation of COVID-19. This review will introduce some methods and breakthroughs achieved by organoids and the limitations of this system.
Collapse
Affiliation(s)
- Chiu Wang Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
- Centre for Translational Stem Cell Biology, 17 Science Park W Ave, Science Park 999077, Hong Kong
| |
Collapse
|
40
|
Kounis NG, Gogos C, de Gregorio C, Hung MY, Kounis SN, Tsounis EP, Assimakopoulos SF, Pourmasumi S, Mplani V, Servos G, Dousdampanis P, Plotas P, Michalaki MA, Tsigkas G, Grammatikopoulos G, Velissaris D, Koniar I. "When," "Where," and "How" of SARS-CoV-2 Infection Affects the Human Cardiovascular System: A Narrative Review. Balkan Med J 2024; 41:7-22. [PMID: 38173173 PMCID: PMC10767774 DOI: 10.4274/balkanmedj.galenos.2023.2023-10-25] [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: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the novel severe acute respiratory coronavirus-2 (SARS-CoV-2). Several explanations for the development of cardiovascular complications during and after acute COVID-19 infection have been hypothesized. The COVID-19 pandemic, caused by SARS-CoV-2, has emerged as one of the deadliest pandemics in modern history. The myocardial injury in COVID-19 patients has been associated with coronary spasm, microthrombi formation, plaque rupture, hypoxic injury, or cytokine storm, which have the same pathophysiology as the three clinical variants of Kounis syndrome. The angiotensin-converting enzyme 2 (ACE2), reninaldosterone system (RAAS), and kinin-kallikrein system are the main proposed mechanisms contributing to cardiovascular complications with the COVID-19 infection. ACE receptors can be found in the heart, blood vessels, endothelium, lungs, intestines, testes, neurons, and other human body parts. SARS-CoV-2 directly invades the endothelial cells with ACE2 receptors and constitutes the main pathway through which the virus enters the endothelial cells. This causes angiotensin II accumulation downregulation of the ACE2 receptors, resulting in prothrombotic effects, such as hemostatic imbalance via activation of the coagulation cascade, impaired fibrinolysis, thrombin generation, vasoconstriction, endothelial and platelet activation, and pro-inflammatory cytokine release. The KKS system typically causes vasodilation and regulates tissue repair, inflammation, cell proliferation, and platelet aggregation, but SARS-CoV-2 infection impairs such counterbalancing effects. This cascade results in cardiac arrhythmias, cardiac arrest, cardiomyopathy, cytokine storm, heart failure, ischemic myocardial injuries, microvascular disease, Kounis syndrome, prolonged COVID, myocardial fibrosis, myocarditis, new-onset hypertension, pericarditis, postural orthostatic tachycardia syndrome, pulmonary hypertension, stroke, Takotsubo syndrome, venous thromboembolism, and thrombocytopenia. In this narrative review, we describe and elucidate when, where, and how COVID-19 affects the human cardiovascular system in various parts of the human body that are vulnerable in every patient category, including children and athletes.
Collapse
Affiliation(s)
- Nicholas G. Kounis
- Department of Cardiology, University of Patras Medical School, Rio, Greece
| | - Christos Gogos
- Clinic of Cardiology, COVID-19 Unit, Papageorgiou General Hospital, Pavlos Melas, Greece
| | - Cesare de Gregorio
- Department of Clinical and Experimental Medicine, University of Messina Medical School, Messina, Italy
| | - Ming-Yow Hung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | | | - Efthymios P. Tsounis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Rio, Greece
| | - Stelios F. Assimakopoulos
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, Rio, Greece
| | - Soheila Pourmasumi
- Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Virginia Mplani
- Intensive Care Unit, Patras University Hospital, Rio, Greece
| | - George Servos
- Pediatric Cardiology Unit, “P. & A. Kyriakou” Children’s Hospital, Athina, Greece
| | | | - Panagiotis Plotas
- Department of Cardiology, University of Patras Medical School, Rio, Greece
| | - Marina A. Michalaki
- Department of Internal Medicine, Division of Endocrinology, University of Patras, School of Health Sciences, Rio, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, University of Patras Medical School, Rio, Greece
| | | | - Dimitrios Velissaris
- Department of Internal Medicine, University of Patras Medical School, Rio, Greece
| | - Ioanna Koniar
- Electrophysiology and Device Department, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
41
|
Razzaq A, Disoma C, Zhou Y, Tao S, Chen Z, Liu S, Zheng R, Zhang Y, Liao Y, Chen X, Liu S, Dong Z, Xu L, Deng X, Li S, Xia Z. Targeting epidermal growth factor receptor signalling pathway: A promising therapeutic option for COVID-19. Rev Med Virol 2024; 34:e2500. [PMID: 38126937 DOI: 10.1002/rmv.2500] [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: 08/14/2023] [Revised: 11/20/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuously producing new variants, necessitating effective therapeutics. Patients are not only confronted by the immediate symptoms of infection but also by the long-term health issues linked to long COVID-19. Activation of epidermal growth factor receptor (EGFR) signalling during SARS-CoV-2 infection promotes virus propagation, mucus hyperproduction, and pulmonary fibrosis, and suppresses the host's antiviral response. Over the long term, EGFR activation in COVID-19, particularly in COVID-19-induced pulmonary fibrosis, may be linked to the development of lung cancer. In this review, we have summarised the significance of EGFR signalling in the context of SARS-CoV-2 infection. We also discussed the targeting of EGFR signalling as a promising strategy for COVID-19 treatment and highlighted erlotinib as a superior option among EGFR inhibitors. Erlotinib effectively blocks EGFR and AAK1, thereby preventing SARS-CoV-2 replication, reducing mucus hyperproduction, TNF-α expression, and enhancing the host's antiviral response. Nevertheless, to evaluate the antiviral efficacy of erlotinib, relevant clinical trials involving an appropriate patient population should be designed.
Collapse
Affiliation(s)
- Aroona Razzaq
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Cyrollah Disoma
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Department of Biology, College of Natural Sciences and Mathematics, Mindanao State University, Marawi City, Philippines
| | - Yuzheng Zhou
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Siyi Tao
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Zongpeng Chen
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Sixu Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Rong Zheng
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yongxing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yujie Liao
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xuan Chen
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Sijie Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Zijun Dong
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Liangtao Xu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xu Deng
- Xiangya School of Pharmaceutical Science, Central South University, Changsha, China
| | - Shanni Li
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Centre for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
42
|
Wolszczak-Biedrzycka B, Dorf J, Wojewódzka-Żelezniakowicz M, Żendzian-Piotrowska M, Dymicka-Piekarska VJ, Matowicka-Karna J, Maciejczyk M. Unveiling COVID-19 Secrets: Harnessing Cytokines as Powerful Biomarkers for Diagnosis and Predicting Severity. J Inflamm Res 2023; 16:6055-6070. [PMID: 38107380 PMCID: PMC10723593 DOI: 10.2147/jir.s439217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction In coronavirus disease (COVID-19), inflammation takes center stage, with a cascade of cytokines released, contributing to both inflammation and lung damage. The objective of this study is to identify biomarkers for diagnosing and predicting the severity of COVID-19. Materials and Methods Cytokine levels were determined in the serum from venous blood samples collected from 100 patients with COVID-19 and 50 healthy controls. COVID-19 patients classified based on the Modified Early Warning (MEWS) score. Cytokine concentrations were determined with a multiplex ELISA kit (Bio-Plex Pro™ Human Cytokine Screening Panel). Results The concentrations of all analyzed cytokines were elevated in the serum of COVID-19 patients relative to the control group, but no significant differences were observed in interleukin-9 (IL-9) and IL-12 p70 levels. In addition, the concentrations of IL-1α, IL-1β, IL-1ra, IL-2Rα, IL-6, IL-12 p40, IL-18, and tumor necrosis factor alpha (TNFα) were significantly higher in symptomatic patients with accompanying pneumonia without respiratory failure (stage 2) than in asymptomatic/mildly symptomatic patients (stage 1). Conclusion The study revealed that IL-1ra, IL-2Rα, IL-6, IL-8, IL-12 p40, IL-16, and IL-18 levels serve as potential diagnostic biomarkers in COVID-19 patients. Furthermore, elevated IL-1α levels proved to be valuable in assessing the severity of COVID-19.
Collapse
Affiliation(s)
- Blanka Wolszczak-Biedrzycka
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, 10-900, Poland
| | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, 15-089, Poland
| | | | | | | | - Joanna Matowicka-Karna
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, 15-089, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Bialystok, 15-089, Poland
| |
Collapse
|
43
|
Liu L, Zhou C, Jiang H, Wei H, Zhou Y, Zhou C, Ji X. Epidemiology, pathogenesis, and management of Coronavirus disease 2019-associated stroke. Front Med 2023; 17:1047-1067. [PMID: 38165535 DOI: 10.1007/s11684-023-1041-7] [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: 05/05/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) epidemic has triggered a huge impact on healthcare, socioeconomics, and other aspects of the world over the past three years. An increasing number of studies have identified a complex relationship between COVID-19 and stroke, although active measures are being implemented to prevent disease transmission. Severe COVID-19 may be associated with an increased risk of stroke and increase the rates of disability and mortality, posing a serious challenge to acute stroke diagnosis, treatment, and care. This review aims to provide an update on the influence of COVID-19 itself or vaccines on stroke, including arterial stroke (ischemic stroke and hemorrhagic stroke) and venous stroke (cerebral venous thrombosis). Additionally, the neurovascular mechanisms involved in SARS-CoV-2 infection and the clinical characteristics of stroke in the COVID-19 setting are presented. Evidence on vaccinations, potential therapeutic approaches, and effective strategies for stroke management has been highlighted.
Collapse
Affiliation(s)
- Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Chenxia Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
| |
Collapse
|
44
|
Raouf GA, Mohammad FK, Merza MA. Polypharmacy and the In Silico Prediction of Potential Body Proteins Targeted by These Drugs Among Hospitalized COVID-19 Patients With Cytokine Storm. Cureus 2023; 15:e48834. [PMID: 38106718 PMCID: PMC10722521 DOI: 10.7759/cureus.48834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Background and objective Polypharmacy is prevalent in coronavirus disease 2019 (COVID-19) patients with severe disease. However, information on polypharmacy among COVID-19 patients who also suffer from cytokine storm is scarce. In light of this, the purpose of the present study was to assess the incidence of polypharmacy and in silico prediction of potential body proteins targeted by these drugs among hospitalized COVID-19 patients who were identified to have the additional burden of cytokine storm in the city of Duhok, Kurdistan Region, Iraq. Methods This was a cross-sectional observational study conducted from June 2021 to April 2022; the phenomena of major polypharmacy (six to nine medications) and excessive polypharmacy (≥10 medications) were documented among 33 (15 males and 18 females) COVID-19 patients with cytokine storm during their hospital stay (8-45 days) in Duhok, Kurdistan Region, Iraq. The SwissTargetPrediction program was utilized in silico to predict and identify human body proteins that could be potentially targeted by selected medications involved in polypharmacy. Results All patients had tested positive for COVID-19 via PCR testing, and they showed different signs and symptoms of the disease. None of the patients recovered and all of them deceased. All 33 patients received many therapeutic agents that ranged in number from eight to 20/patient during their hospital stay. The mean number of medications was 15 ± 3. We identified 2/33 (6%) patients with major polypharmacy (eight and nine) and 31/33 (94%) with excessive polypharmacy (15.5 ± 2.7). The total number of medications identified in polypharmacy was 37, excluding vitamins, minerals, and intravenous solutions. The frequency of medications administered was as follows: antibiotics (67, 13.7%), mucolytic agents (56, 11.5%), corticosteroids (54, 11%), anticoagulants (48, 9.8%), antiviral agents (41, 8.4%), antihypertensive agents (32, 6.5%), analgesics (28, 5.7%), antifungal drugs (27, 5.5%), antidiabetics (26, 5.3%), and other medications (2-19, 0.41-3.9%). Using the SwissTargetPrediction program, various drugs including antiviral agents involved in polypharmacy were found to target, in silico, body proteins at a prediction percentage that ranged from 6.7% to 40%. Conclusions Major and extensive polypharmacy conditions were identified in hospitalized COVID-19 patients suffering from cytokine storm. The severity of COVID-19 with cytokine storm, comorbidities, and hospitalization were key factors associated with polypharmacy in the patients. The SwissTargetPrediction web server is useful for predicting in silico potential human body protein targets that could possibly be sources of additional information on the adverse/toxic effects of polypharmacy medications administered concurrently. Further research in current medication protocols prescribed for advanced COVID-19 illness with cytokine storm is warranted to gain deeper insights into the topic.
Collapse
Affiliation(s)
- Ghazwan A Raouf
- Department of Pharmacology, College of Pharmacy, University of Duhok, Duhok, IRQ
| | - Fouad K Mohammad
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Mosul, Mosul, IRQ
- College of Nursing, The American University of Kurdistan, Duhok, IRQ
| | - Muayad A Merza
- Department of Internal Medicine, University of Duhok, Duhok, IRQ
| |
Collapse
|
45
|
Hamad RS, Al-Kuraishy HM, Alexiou A, Papadakis M, Ahmed EA, Saad HM, Batiha GES. SARS-CoV-2 infection and dysregulation of nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. Cell Stress Chaperones 2023; 28:657-673. [PMID: 37796433 PMCID: PMC10746631 DOI: 10.1007/s12192-023-01379-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/19/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a recent pandemic caused by a novel severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) leading to pulmonary and extra-pulmonary manifestations due to the development of oxidative stress (OS) and hyperinflammation. The underlying cause for OS and hyperinflammation in COVID-19 may be related to the inhibition of nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of antioxidative responses and cellular homeostasis. The Nrf2 pathway inhibits the expression of pro-inflammatory cytokines and the development of cytokine storm and OS in COVID-19. Nrf2 activators can attenuate endothelial dysfunction (ED), renin-angiotensin system (RAS) dysregulation, immune thrombosis, and coagulopathy. Hence, this review aimed to reveal the potential role of the Nrf2 pathway and its activators in the management of COVID-19. As well, we tried to revise the mechanistic role of the Nrf2 pathway in COVID-19.
Collapse
Affiliation(s)
- Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, 31982, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| |
Collapse
|
46
|
Liu S, Zhao Y, Feng X, Xu H. SARS-CoV-2 infection threatening intestinal health: A review of potential mechanisms and treatment strategies. Crit Rev Food Sci Nutr 2023; 63:12578-12596. [PMID: 35894645 DOI: 10.1080/10408398.2022.2103090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic has brought great problems to mankind, including economic recession and poor health. COVID-19 patients are frequently reported with gastrointestinal symptoms such as diarrhea and vomiting in clinical diagnosis. Maintaining intestinal health is the key guarantee to maintain the normal function of multiple organs, otherwise it will be a disaster. Therefore, the purpose of this review was deeply understanded the potential mechanism of SARS-CoV-2 infection threatening intestinal health and put forward reasonable treatment strategies. Combined with the existing researches, we summarized the mechanism of SARS-CoV-2 infection threatening intestinal health, including intestinal microbiome disruption, intestinal barrier dysfunction, intestinal oxidative stress and intestinal cytokine storm. These adverse intestinal events may affect other organs through the circulatory system or aggravate the course of the disease. Typically, intestinal disadvantage may promote the progression of SARS-CoV-2 through the gut-lung axis and increase the disease degree of COVID-19 patients. In view of the lack of specific drugs to inhibit SARS-CoV-2 replication, the current review described new strategies of probiotics, prebiotics, postbiotics and nutrients to combat SARS-CoV-2 infection and maintain intestinal health. To provide new insights for the prevention and treatment of gastrointestinal symptoms and pneumonia in patients with COVID-19.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
47
|
Singh M, Pushpakumar S, Zheng Y, Smolenkova I, Akinterinwa OE, Luulay B, Tyagi SC. Novel mechanism of the COVID-19 associated coagulopathy (CAC) and vascular thromboembolism. NPJ VIRUSES 2023; 1:3. [PMID: 38077924 PMCID: PMC10710223 DOI: 10.1038/s44298-023-00003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 01/31/2024]
Abstract
Previous studies from our laboratory revealed that SARS-CoV-2 spike protein (SP) administration to a genetically engineered model expressing the human angiotensin-converting enzyme 2; ACE2 receptor (i.e., hACE2 humanized mouse) mimicked the coronavirus disease-19 (COVID-19) pathology. In humans the cause of high morbidity, and mortality is due to 'cytokine-storm' led thromboembolism; however, the exact mechanisms of COVID-19 associated coagulopathy (CAC) have yet to be discovered. Current knowledge suggests that CAC is distinct from the standard coagulopathy, in that the intrinsic and extrinsic thrombin-dependent coagulation factors, and the pathway(s) that are common to coagulopathy, are not recruited by SARS-CoV-2. Findings from patients revealed that there is little change in their partial thromboplastin, or the prothrombin time coupled with a significant decline in platelets. Further, there appears to be an endothelial dysfunction during COVID-19 suggesting an interaction of the endothelia with immune cells including neutrophils. There are also reports that inflammatory NGAL is elevated during COVID-19. Furthermore, the levels of NPT are also increased indicating an increase in inflammatory M1 macrophage iNOS which sequesters BH4; an essential enzyme co-factor that acts as a potent antioxidant thus causing damage to endothelia. SARS-CoV-2 entry into the host cells is facilitated by a co-operative action between TMPRSS2 and the main ACE2 receptor. Interestingly, after infection ADAMTS13; a von Willebrand factor; VWF cleaving enzyme is found to be decreased. Based on these facts, we hypothesize that vascular thromboembolism is associated with serine and metalloproteinase, and in that context, we opine that inhibition of iNOS might help mitigate COVID-19 harmful effects. To test this hypothesis, we administered SP to the hACE2 mice that were subsequently treated with amino guanidine (AG; a potent inhibitor of glycoxidation, lipoxidation and oxidative vicious cycles). Our results revealed increase in TMPRSS2, and NGAL by SP but treatment with AG mitigated their levels. Similarly, levels of MMP-2, and -9 were increased; however, AG treatment normalized these levels. Our findings suggest that occurrence of CAC is influenced by TMPRSS2, ADAMTS13, NGAL and MMP- 2, and -9 factors, and an intervention with iNOS blocker helped mitigate the CAC condition in experimental settings.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- These authors contributed equally: Mahavir Singh, Sathnur Pushpakumar
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- These authors contributed equally: Mahavir Singh, Sathnur Pushpakumar
| | - Yuting Zheng
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Irina Smolenkova
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Oluwaseun E. Akinterinwa
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bana Luulay
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suresh C. Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
48
|
Lücke J, Heinrich F, Malsy J, Meins N, Schnell J, Böttcher M, Nawrocki M, Zhang T, Bertram F, Sabihi M, Kempski J, Blankenburg T, Duprée A, Reeh M, Wolter S, Mann O, Izbicki JR, Lohse AW, Gagliani N, Lütgehetmann M, Bunders MJ, Altfeld M, Sauter G, Giannou AD, Krasemann S, Ondruschka B, Huber S. Intestinal IL-1β Plays a Role in Protecting against SARS-CoV-2 Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1052-1061. [PMID: 37556130 PMCID: PMC10476162 DOI: 10.4049/jimmunol.2200844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/11/2023] [Indexed: 08/10/2023]
Abstract
The intestine is constantly balancing the maintenance of a homeostatic microbiome and the protection of the host against pathogens such as viruses. Many cytokines mediate protective inflammatory responses in the intestine, among them IL-1β. IL-1β is a proinflammatory cytokine typically activated upon specific danger signals sensed by the inflammasome. SARS-CoV-2 is capable of infecting multiple organs, including the intestinal tract. Severe cases of COVID-19 were shown to be associated with a dysregulated immune response, and blocking of proinflammatory pathways was demonstrated to improve patient survival. Indeed, anakinra, an Ab against the receptor of IL-1β, has recently been approved to treat patients with severe COVID-19. However, the role of IL-1β during intestinal SARS-CoV-2 infection has not yet been investigated. Here, we analyzed postmortem intestinal and blood samples from patients who died of COVID-19. We demonstrated that high levels of intestinal IL-1β were associated with longer survival time and lower intestinal SARS-CoV-2 RNA loads. Concurrently, type I IFN expression positively correlated with IL-1β levels in the intestine. Using human intestinal organoids, we showed that autocrine IL-1β sustains RNA expression of IFN type I by the intestinal epithelial layer. These results outline a previously unrecognized key role of intestinal IL-1β during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Heinrich
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Malsy
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
- German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Germany
| | - Nicholas Meins
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Josa Schnell
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marius Böttcher
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Bertram
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Kempski
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tom Blankenburg
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Duprée
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W. Lohse
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madeleine J. Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasios D. Giannou
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Krasemann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Gasparello J, Marzaro G, Papi C, Gentili V, Rizzo R, Zurlo M, Scapoli C, Finotti A, Gambari R. Effects of Sulforaphane on SARS‑CoV‑2 infection and NF‑κB dependent expression of genes involved in the COVID‑19 'cytokine storm'. Int J Mol Med 2023; 52:76. [PMID: 37477130 PMCID: PMC10555481 DOI: 10.3892/ijmm.2023.5279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/09/2023] [Indexed: 07/22/2023] Open
Abstract
Since its spread at the beginning of 2020, the coronavirus disease 2019 (COVID‑19) pandemic represents one of the major health problems. Despite the approval, testing, and worldwide distribution of anti‑severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) vaccines, the development of specific antiviral agents targeting the SARS‑CoV‑2 life cycle with high efficiency, and/or interfering with the associated 'cytokine storm', is highly required. A recent study, conducted by the authors' group indicated that sulforaphane (SFN) inhibits the expression of IL‑6 and IL‑8 genes induced by the treatment of IB3‑1 bronchial cells with a recombinant spike protein of SARS‑CoV‑2. In the present study, the ability of SFN to inhibit SARS‑CoV‑2 replication and the expression of pro‑inflammatory genes encoding proteins of the COVID‑19 'cytokine storm' was evaluated. SARS‑CoV‑2 replication was assessed in bronchial epithelial Calu‑3 cells. Moreover, SARS‑CoV‑2 replication and expression of pro‑inflammatory genes was evaluated by reverse transcription quantitative droplet digital PCR. The effects on the expression levels of NF‑κB were assessed by western blotting. Molecular dynamics simulations of NF‑kB/SFN interactions were conducted with Gromacs 2021.1 software under the Martini 2 CG force field. Computational studies indicated that i) SFN was stably bound with the NF‑κB monomer; ii) a ternary NF‑kB/SFN/DNA complex was formed; iii) the SFN interacted with both the protein and the nucleic acid molecules modifying the binding mode of the latter, and impairing the full interaction between the NF‑κB protein and the DNA molecule. This finally stabilized the inactive complex. Molecular studies demonstrated that SFN i) inhibits the SARS‑CoV‑2 replication in infected Calu‑3 cells, decreasing the production of the N‑protein coding RNA sequences, ii) decreased NF‑κB content in SARS‑CoV‑2 infected cells and inhibited the expression of NF‑kB‑dependent IL‑1β and IL‑8 gene expression. The data obtained in the present study demonstrated inhibitory effects of SFN on the SARS‑CoV‑2 life cycle and on the expression levels of the pro‑inflammatory genes, sustaining the possible use of SFN in the management of patients with COVID‑19.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, I-35131 Padova
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Valentina Gentili
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Roberta Rizzo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Chiara Scapoli
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| |
Collapse
|
50
|
Song Y, You Q, Chen X. Transition Metal-Based Therapies for Inflammatory Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2212102. [PMID: 36863722 DOI: 10.1002/adma.202212102] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/15/2023] [Indexed: 08/04/2023]
Abstract
Inflammatory disease (ID) is a general term that covers all diseases in which chronic inflammation performs as the major manifestation of pathogenesis. Traditional therapies based on the anti-inflammatory and immunosuppressive drugs are palliative with the short-term remission. The emergence of nanodrugs has been reported to solve the potential causes and prevent recurrences, thus holding great potential for the treatment of IDs. Among various nanomaterial systems, transition metal-based smart nanosystems (TMSNs) with unique electronic structures possess therapeutic advantages owing to their large surface area to volume ratio, high photothermal conversion efficiency, X-ray absorption capacity, and multiple catalytic enzyme activities. In this review, the rationale, design principle, and therapeutic mechanisms of TMSNs for treatments of various IDs are summarized. Specifically, TMSNs can not only be designed to scavenge danger signals, such as reactive oxygen and nitrogen species and cell-free DNA, but also can be engineered to block the mechanism of initiating inflammatory responses. In addition, TMSNs can be further applied as nanocarriers to deliver anti-inflammatory drugs. Finally, the opportunities and challenges of TMSNs are discussed, and the future directions of TMSN-based ID treatment for clinical applications are emphasized.
Collapse
Affiliation(s)
- Yilin Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Qing You
- Departments of Diagnostic, Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program NUS center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic, Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program NUS center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|