1
|
Wu H, Liu Z, Li Y. Intestinal microbiota and respiratory system diseases: Relationships with three common respiratory virus infections. Microb Pathog 2025; 203:107500. [PMID: 40139334 DOI: 10.1016/j.micpath.2025.107500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
In recent years, the role of the intestinal microbiota in regulating host health and immune balance has attracted widespread attention. This study provides an in-depth analysis of the close relationship between the intestinal microbiota and respiratory system diseases, with a focus on three common respiratory virus infections, including respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and influenza virus. The research indicates that during RSV infection, there is a significant decrease in intestinal microbial diversity, suggesting the impact of the virus on the intestinal ecosystem. In SARS-CoV-2 infection, there are evident alterations in the intestinal microbiota, which are positively correlated with the severity of the disease. Similarly, influenza virus infection is associated with dysbiosis of the intestinal microbiota, and studies have shown that the application of specific probiotics exhibits beneficial effects against influenza virus infection. Further research indicates that the intestinal microbiota exerts a wide and profound impact on the occurrence and development of respiratory system diseases through various mechanisms, including modulation of the immune system and production of short-chain fatty acids (SCFAs). This article comprehensively analyzes these research advances, providing new perspectives and potential strategies for the prevention and treatment of future respiratory system diseases. This study not only deepens our understanding of the relationship between the intestinal microbiota and respiratory system diseases but also offers valuable insights for further exploring the role of host-microbiota interactions in the development of diseases.
Collapse
Affiliation(s)
- Haonan Wu
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China
| | - Ziyu Liu
- The First Hospital of Jilin University, Changchun, China.
| | - Yanan Li
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Raha JR, Kim KH, Le CTT, Bhatnagar N, Pal SS, Liu R, Grovenstein P, Yeasmin M, Racheal F, Shin CH, Wang BZ, Kang SM. Intranasal vaccination with multi-neuraminidase and M2e virus-like particle vaccine results in greater mucosal immunity and protection against influenza than intramuscular injection. Vaccine 2025; 57:127206. [PMID: 40339180 DOI: 10.1016/j.vaccine.2025.127206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Intramuscular injection of seasonal influenza vaccines provides strain-specific neutralizing antibodies, but not against variants, and no effective mucosal immunity. Here, we report that multi-subtype neuraminidase (NA) and M2 ectodomain repeat (5xM2e) virus-like particle vaccine (NA-M2e) conferred higher efficacy of broad cross-protection after two doses of intranasal delivery than intramuscular injection. The intranasally vaccinated mice displayed high levels of IgA antibodies, IFN-γ+ CD4 and CD8 T cells, germinal center B cells, plasma cells, and early innate immune cells locally in the lungs. In contrast, intramuscular vaccination systemically induced innate and adaptive immune responses in the spleen. Our findings demonstrate that the intranasal delivery of NA-M2e vaccine induces enhanced mucosal immunity and comparable serum IgG antibodies, offering improved efficacy of cross-protection against diverse influenza virus strains compared to the intramuscular injection in a mouse model.
Collapse
Affiliation(s)
- Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Mahmuda Yeasmin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Farayola Racheal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
3
|
Jin Y, Fu Y, Liu Q, Li S, Zeng Y, Fu L, Zhang Y. RH-RPA: A Rapid and Highly Sensitive Assay for Nucleic Acid Detection Based on RNase HII Combined with Recombinase Polymerase Amplification. Anal Chem 2025; 97:9220-9227. [PMID: 40273346 DOI: 10.1021/acs.analchem.4c06578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Currently, RPA-exo and RPA-nfo are the primary methods for RPA/RT-RPA probe assays, both of which have been widely applied to the detection of various targets. However, RPA-nfo exhibits lower sensitivity compared with the exo probe method, while RPA-exo lacks the capability for equipment-free visualization inherent to RPA-nfo. Both of the approaches mentioned above limit the broader application of RPA/RT-RPA probe assays. To address those limitations, we have developed a novel recombinase polymerase amplification (RPA) combined with an E. coli RNase HII assay (RH-RPA). This approach supports both fluorescence signal detection and lateral-flow strip readouts. Due to the high efficiency and specificity of E. coli RNase HII in recognizing and cleaving targets, this method serves as a rapid and accurate molecular diagnostic platform. Under the fluorescence detection mode, RH-RPA achieves a limit of detection as low as 10 copies per reaction for both DNA and RNA within 20 min. Additionally, the lateral-flow strip mode enables the detection of as few as 5 copies per reaction of nucleic acids within 20 min. In clinical sample analysis, the RT RH-RPA demonstrated 100% accuracy in detecting the influenza A virus, underscoring its reliability in practical diagnostics. These findings highlight the stable specificity, rapid performance, high sensitivity, and cost-effectiveness of the RH-RPA methods, showcasing their potential as promising tools for point-of-care nucleic acid detection.
Collapse
Affiliation(s)
- Yuting Jin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangnan Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| | - Qingyang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| | - Suhang Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| | - Yingzhou Zeng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| | - Lijuan Fu
- Department of Infection, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Quality Control Center of Infectious Diseases, Xiamen, Fujian 361000, China
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
4
|
Vasilakopoulos AP, Kainth MK. A Review of Pediatric Influenza. Pediatr Ann 2025; 54:e174-e178. [PMID: 40305632 DOI: 10.3928/19382359-20250307-06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Influenza remains a significant public health concern, particularly among children. This comprehensive review focuses on pathophysiology, epidemiology, clinical presentation, treatment, and vaccines for pediatric influenza. Key genetic and immune-related factors, as well as excessive neutrophil activation, contribute to severe disease and outcomes. Epidemiologically, influenza affects 8% to 10% of children in the United States annually, with higher hospitalization rates among children who are Black, Hispanic, Native American and Alaska Native, and Asian and Pacific Islander. Clinically, children exhibit more severe symptoms and higher rates of gastrointestinal manifestations compared with adults. Treatment primarily involves neuraminidase inhibitors, like oseltamivir; although, therapies, such as baloxavir marboxil, are also effective. Vaccination remains the cornerstone of prevention, yet declining rates of uptake and increasing vaccine hesitancy pose challenges for population immunity. Universal influenza vaccine research offers hope for broader protection. This review aims to inform evidence-based practices in managing pediatric influenza and highlights areas for future research. [Pediatr Ann. 2025;54(5):e174-e178.].
Collapse
|
5
|
Boyd DF, Jordan SV, Balachandran S. ZBP1-driven cell death in severe influenza. Trends Microbiol 2025; 33:521-532. [PMID: 39809680 DOI: 10.1016/j.tim.2024.12.008] [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: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Influenza A virus (IAV) infections can cause life-threatening illness in humans. The severity of disease is directly linked to virus replication in the alveoli of the lower respiratory tract. In particular, the lytic death of infected alveolar epithelial cells (AECs) is a major driver of influenza severity. Recent studies have begun to define the molecular mechanisms by which IAV triggers lytic cell death. Z-form nucleic-acid-binding protein 1 (ZBP1) senses replicating IAV and drives programmed cell death (PCD) in infected cells, including apoptosis and necroptosis in AECs and pyroptosis in myeloid cells. Necroptosis and pyroptosis, both lytic forms of death, contribute to pathogenesis during severe infections. Pharmacological blockade of necroptosis shows strong therapeutic potential in mouse models of lethal influenza. We suggest that targeting ZBP1-initiated necroinflammatory cell lysis, either alone or in combination antiviral drugs, will provide clinical benefit in severe influenza.
Collapse
Affiliation(s)
- David F Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | - Summer Vaughn Jordan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
6
|
Nishioka K, Nakagawa M, Tanino Y, Nakaya T. Neisseria perflava isolated from a clinical sample reduces influenza virus replication in respiratory cells. J Oral Biosci 2025; 67:100665. [PMID: 40280275 DOI: 10.1016/j.job.2025.100665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVES Various bacteria are present in the oral cavity and constitute the oral microbiota. Although the oral microbiota has been analyzed using next-generation sequencing, few studies have investigated whether specific commensal bacteria directly affect immune responses to infections. Here, we focused on Neisseria species present in the oral cavity and investigated their effects on respiratory cells infected with several viruses. METHODS Six Neisseria species were isolated from human saliva. The epithelial cell lines were stimulated with bacterial culture supernatants before viral infection. Changes in the viral susceptibility were assessed. RESULTS Culture supernatants of two Neisseria species were found to affect cells susceptible to influenza viral infection and suppress influenza viral replication. The mechanism underlying the suppression of N. perflava was further investigated. This activity was observed in the 10-30 kDa protein range fractionated by ultrafiltration. Although viral replication was suppressed by stimulation with bacterial proteins, the infection efficiency of the virus and cytokine production were unaffected. Replication of SARS-CoV-2 and human rhinovirus were also suppressed. CONCLUSION Viral infection was performed after supernatant stimulation, suggesting that exposure to oral bacteria directly affects viral infection in the surrounding cells. This effect has been observed for several viruses. Viral genome replication in cells may be suppressed by enhanced expression of viral replication suppression genes. Further analyses are required to elucidate the detailed underlying mechanisms.
Collapse
Affiliation(s)
- Keisuke Nishioka
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| | - Maki Nakagawa
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoko Tanino
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; Department of Clinical Investigation, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takaaki Nakaya
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
7
|
Li D, van der Veen TA, de Groot LES, de Jager MH, Lan A, Baarsma HA, Lutter R, van der Graaf K, Gosens R, Schmidt M, Melgert BN. Pre-Existing Allergic Inflammation Alters Both Innate and Adaptive Immune Responses in Mice Co-Infected with Influenza Virus. Int J Mol Sci 2025; 26:3483. [PMID: 40331962 PMCID: PMC12027099 DOI: 10.3390/ijms26083483] [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: 01/18/2025] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 05/08/2025] Open
Abstract
Asthma, a chronic airway disease, is marked by allergic inflammation, hyperresponsiveness, and tissue remodeling. Influenza infections in asthma patients can cause severe exacerbations, though the underlying mechanisms remain unclear. This study investigated how pre-existing allergic inflammation affects immune responses to influenza infection in mice exposed to house dust mite (HDM). Mice were repeatedly exposed to HDM, followed by infection with the influenza A virus, and were sacrificed three days post-infection. Plasma was analyzed for HDM-specific immunoglobulins, while lung tissue was used for immune cell flow cytometry and RNA sequencing analysis. HDM exposure induced allergic inflammation, evidenced by more HDM-specific IgE, IgG1, IgG2, eosinophils, neutrophils, Th1, and Th17 cells compared to controls. Upon influenza infection, the effects of HDM and influenza co-infection interacted, showing fewer Th1 cells and regulatory T cells and more Th2 cells compared to mice exposed to the influenza virus alone. Interestingly, RNA-seq analysis revealed less upregulation of Th1-related genes and antiviral pathways in co-exposed mice, suggesting impaired Th1 immunity and antiviral responses. Pre-existing allergic inflammation significantly altered immune responses in mice co-infected with influenza, revealing underdeveloped antiviral responses as early as three days post-infection. These findings may explain the increased susceptibility of patients with asthma to severe viral diseases.
Collapse
Affiliation(s)
- Dan Li
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - T. Anienke van der Veen
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Linsey E. S. de Groot
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands (R.L.)
| | - Marina H. de Jager
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
| | - Andy Lan
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
| | - Hoeke A. Baarsma
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - René Lutter
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands (R.L.)
| | | | - Reinoud Gosens
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Martina Schmidt
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Barbro N. Melgert
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
8
|
Zhao M, Deng D, Liu H, Guo R, Wu J, Hao Y, Yang M. Berberine Suppresses Influenza A Virus-Triggered Pyroptosis in Macrophages via Intervening in the mtROS-MAVS-NLRP3 Inflammasome Pathway. Viruses 2025; 17:539. [PMID: 40284982 PMCID: PMC12030943 DOI: 10.3390/v17040539] [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: 02/21/2025] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Infection with influenza A virus (IAV) may trigger excessive inflammatory responses, leading to severe viral pneumonia and accelerating disease progression. Therefore, controlling these excessive inflammatory responses is crucial for the prevention and treatment of pneumonia caused by IAV. Berberine (BBR), an isoquinoline alkaloid extracted from traditional Chinese medicine, possesses extensive pharmacological activities. However, its immunoregulatory effects and molecular mechanisms in the context of IAV infection require further investigation. This study explored the impact of BBR on macrophage pyroptosis and inflammatory responses induced by IAV infection. Our findings revealed that BBR effectively inhibits the release of IL-1β and TNF-α induced by IAV infection and suppresses gasdermin D (GSDMD)-mediated pyroptosis in a dose-dependent manner. Further research indicates that BBR alleviates macrophage pyroptosis and inflammatory responses in IAV-infected cells by reducing the release of mitochondrial reactive oxygen species (mtROS), inhibiting mitochondrial antiviral signaling protein (MAVS) expression and blocking the activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. Experiments using siRNA to knockdown MAVS further confirmed the pivotal role of MAVS in BBR's inhibition of IAV-induced macrophage pyroptosis. This study provides a scientific basis for the application of BBR as an anti-inflammatory drug in the treatment of inflammatory diseases caused by IAV infection and directs future research endeavors.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Hao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.Z.); (D.D.); (H.L.); (R.G.); (J.W.)
| | - Mingrui Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.Z.); (D.D.); (H.L.); (R.G.); (J.W.)
| |
Collapse
|
9
|
Li Y, Li J, Li T, Liu C, Du J, Li Y, Chen Y, Zhang Y, Wang X, Wang X, Jia X, Deng H. Plasma nontargeted metabolomics study of H1N1 and H3N2 influenza in children. Front Cell Infect Microbiol 2025; 15:1537726. [PMID: 40256448 PMCID: PMC12006178 DOI: 10.3389/fcimb.2025.1537726] [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: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 04/22/2025] Open
Abstract
Background This study used a nontargeted metabolomic approach to investigate small molecular metabolites in the peripheral blood of pediatric patients with influenza. By comparing these metabolites with those in healthy children, potential biomarkers for the early detection and diagnosis of influenza were explored. Methods Plasma samples were collected from 47 children with H1N1 influenza, 40 with H3N2 influenza, and 40 healthy controls at Xi'an Children's Hospital, Xi'an Jiaotong University Second Affiliated Hospital, and Xi'an Central Hospital between May and September 2023. Nontargeted metabolomic detection and analysis were performed. Results In the H1N1 group, 14 glycerophospholipid metabolites were significantly altered compared to controls, with 11 (78.5%) markedly downregulated. These downregulated metabolites showed negative correlations with inflammatory markers, including white blood cell (WBC) count, neutrophils, C-reactive protein (CRP), and Procalcitonin (PCT), whereas the upregulated metabolite PC(P-18:1(9Z)/16:0) showed positive correlations with validation markers. In the H3N2 group, 12 glycerophospholipid metabolites were significantly altered, with 9 being downregulated. The downregulated LysoPC(20:0/0:0) showed a positive correlation with alanine aminotransferase (ALT) but a negative correlation with WBC count, while the upregulated metabolite LysoPA(18:1(9Z)0:0) correlated positively with ALT, aspartate aminotransferase (AST), and lactate dehydrogenase (LDH). Conclusions Distinct metabolomic profiles were identified in pediatric H1N1 and H3N2 influenza cases compared to healthy controls. Specific glycerophospholipid metabolites were closely associated with inflammatory and liver function markers, highlighting their potential as biomarkers for disease monitoring and early diagnosis.
Collapse
Affiliation(s)
- Yaping Li
- Department of Infectious Diseases, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Jiaxin Li
- Department of Infectious Diseases, Xi’an Children’s Hospital, Xi’an, China
- School of Public Health, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Ting Li
- Department of Infectious Diseases, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Chenrui Liu
- Department of Infectious Diseases, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Jiayi Du
- Epidemiology of Microbial Disease, Yale University School of Public Health, New Haven, CT, United States
| | - Yuxin Li
- Department of Infectious Diseases, Xi’an Children’s Hospital, Xi’an, China
| | - Yuan Chen
- Department of Neurology, Xi’an Children’s Hospital, Xi’an, China
| | - Yufeng Zhang
- Department of Infectious Diseases, Xi’an Children’s Hospital, Xi’an, China
| | - Xiaoyan Wang
- Department of Infectious Diseases, Xi’an Children’s Hospital, Xi’an, China
| | - Xinyu Wang
- Department of Infectious Diseases, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Xiaoli Jia
- Department of Infectious Diseases, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Huiling Deng
- Department of Infectious Diseases, Xi’an Children’s Hospital, Xi’an, China
- Department of Pediatrics, Xi’an Central Hospital, Xi’an, China
| |
Collapse
|
10
|
Branche A, Mulligan MJ, Maniar A, Puente O, Oladipupo I, Crowther G, Zareba AM, Yi Z, Scully I, Gomme E, Koury K, Kitchin N, Allen PS, Anderson AS, Gurtman A, Lindert K. A Phase 1/2 Randomized Study to Evaluate the Safety, Tolerability, and Immunogenicity of Nucleoside-Modified Messenger RNA Influenza Vaccines in Healthy Adults. Vaccines (Basel) 2025; 13:383. [PMID: 40333267 PMCID: PMC12031420 DOI: 10.3390/vaccines13040383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Background/Objectives: Circulating influenza strains antigenically differing from vaccine antigens increase disease burden by decreasing vaccine efficacy. Nucleoside-modified mRNA (modRNA) influenza vaccines may facilitate rapid production allowing later antigen selection and improved antigenic similarity compared to circulating strains. We studied different influenza modRNA vaccine (IRV) formulations and dose levels. Methods: This phase 1/2 randomized study evaluated IRV safety/tolerability and immunogenicity in healthy 18- through 85-year-olds. Based on safety and immunogenicity for different IRV doses, schedules, and valencies versus the quadrivalent influenza vaccine (QIV; Fluzone High-Dose Quadrivalent, Sanofi Pasteur) in phase 1 (65-85-year-olds), quadrivalent IRV (qIRV) was further evaluated in 65- through 85-year-olds and 18- through 64-year-olds in phase 2, leading to phase 3 dose selection. Results: Phase 1 (65-85-year-olds) safety/tolerability and immunogenicity findings supported qIRV 30-µg and 60-µg phase 2 assessment (18-85-year-olds, N = 610). qIRV was well tolerated. Injection site pain was the most frequently reported local reaction. Reactogenicity event incidences ≤ 7 days postvaccination for qIRV were generally higher versus QIV, observed more frequently in 18- through 64-year-olds than 65- through 85-year-olds, and showed dose-related trends (60 μg > 30 μg). qIRV and QIV adverse event profiles in 65- through 85-year-olds were similar. There were higher postvaccination hemagglutination inhibition assay geometric mean titers and fold rises and seroconversion rates observed with qIRV versus QIV for A strains, with no consistent pattern for B strains. Cell-mediated immune responses to qIRV by Day 7 showed overall higher T-cell responses against all strains versus QIV. Antibody and cell-mediated immune responses showed comparable trends across qIRV doses in 18- through 85-year-olds; a dose-related pattern was observed in 65- through 85-year-olds (60 μg > 30 μg). Conclusions: Phase 3 investigations of qIRV 60 µg in older adults and qIRV 30 µg in younger adults are warranted (ClinicalTrials.gov Identifier: NCT05052697).
Collapse
Affiliation(s)
- Angela Branche
- Department of Medicine, Division of Infectious Diseases, University of Rochester, Rochester, NY 14642, USA
| | - Mark J. Mulligan
- New York University (NYU) Vaccine Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Alok Maniar
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Orlando Puente
- Miami Dade Medical Research Institute, Miami, FL 33176, USA
| | - Islamiat Oladipupo
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Graham Crowther
- Vaccine Research and Development, Pfizer Ltd., Hurley SL6 6RJ, UK
| | | | - Zhuobiao Yi
- Vaccine Research and Development, Pfizer Inc., Collegeville, PA 19426, USA
| | - Ingrid Scully
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Emily Gomme
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Kenneth Koury
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Nicholas Kitchin
- Vaccine Research and Development, Pfizer Ltd., Hurley SL6 6RJ, UK
| | | | | | - Alejandra Gurtman
- Vaccine Research and Development, Pfizer Inc., Pearl River, NY 10965, USA
| | - Kelly Lindert
- Vaccine Research and Development, Pfizer Inc., Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Zheng Y, Zhang Y, Chen Y, Deng X, Liu B, Xu Q, Qian C, Zhang Z, Wang K, Zeng Y, Liang Z, Sang L, Nong L, Liu X, Xu Y, Li Y, Huang Y. Indoleamine 2,3-dioxygenase 1 drives epithelial cells ferroptosis in influenza-induced acute lung injury. Redox Biol 2025; 81:103572. [PMID: 40023977 PMCID: PMC11915170 DOI: 10.1016/j.redox.2025.103572] [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] [Received: 02/12/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening complication of influenza A virus (IAV) infection, characterized by high morbidity and mortality. Recent studies have implicated ferroptosis, a distinct form of regulated cell death characterized by iron-dependent lipid peroxidation, in the pathogenesis of IAV-induced ALI. However, the underlying mechanisms and key regulators of IAV-induced ferroptosis remain largely unknown. In this study, we found that IAV infection induces predominant ferroptosis in alveolar and bronchial epithelial cells, contributing to tissue damage and the development of acute lung injury. Treatment with the ferroptosis inhibitor ferrostatin-1 improved survival, mitigated weight loss, and alleviated lung injury in IAV-infected mice. Mechanistically, IAV-induced ferroptosis was associated with excess lipid peroxidation, nitrative stress, and disrupted iron metabolism. Targeted lipidomic analysis revealed that phospholipid peroxidation is a crucial mechanism in IAV-induced ferroptosis. Importantly, we identified indoleamine 2,3-dioxygenase 1 (IDO1) as a key regulator of IAV-induced ferroptosis. IDO1 knockdown inhibited IAV-induced cell death, and reduced intracellular reactive oxygen species, peroxynitrite, and inducible nitric oxide synthase expression. Furthermore, pharmacological inhibition of IDO1 with 1-methyl-tryptophan improved ALI phenotype in IAV-infected mice. These findings highlight the critical role of ferroptosis in IAV-induced ALI pathogenesis and identify IDO1 as a potential therapeutic target for the treatment of this life-threatening condition.
Collapse
Affiliation(s)
- Yongxin Zheng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yu Zhang
- Department of Critical Care Medicine, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiumei Deng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qiang Xu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Chuyun Qian
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, China
| | - Zhihui Zhang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ke Wang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yuan Zeng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zhenting Liang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ling Sang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Lingbo Nong
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| | - Yongbo Huang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
12
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [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: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
13
|
Arumugam H, Wong KH, Low ZY, Lal S, Choo WS. Plant extracts as a source of antiviral agents against influenza A virus. J Appl Microbiol 2025; 136:lxaf056. [PMID: 40058769 DOI: 10.1093/jambio/lxaf056] [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: 06/13/2024] [Revised: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
The influenza virus, especially influenza A (IAV), has remained a constant global health threat due to its high morbidity rate and ability to undergo antigenic shifts and drifts, causing pandemics and epidemics. Due to the rapid evolution of IAV, novel therapeutics are urgently required to combat these viruses effectively, as they develop resistance against current therapeutics. Natural products have been the subject of debate for alternative IAV therapy, where the abundance of bioactive compounds offers numerous potentials for novel anti-IAV drug discovery. Therefore, this review discusses the antiviral effects of natural plant extracts against IAV. Examples are Silybum marianum, Scutellaria baicalensis, Angelica dahurica, Peganum harmala, Sambucus nigra, Echinacea purpurea, Panax ginseng, and Camellia sinensis. Most studies found that Si. marianum inhibits viral ribonucleic acid (RNA) synthesis. In contrast, Sc. baicalensis, A. dahurica, Sa. nigra, C. sinensis, and E. purpurea were effective in preventing the entry or binding of IAV into host cells. On the other hand, Sc. baicalensis and Pa. ginseng exert their anti-IAV effect via immunomodulation. Peganum harmala, on the contrary, exhibits a direct virucidal effect against IAV. These studies have shown promising results from using natural products against IAV, which may aid in formulating combinatorial compounds as anti-IAV therapy.
Collapse
Affiliation(s)
- Hanushree Arumugam
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor DE, Malaysia
| | - Ka Heng Wong
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor DE, Malaysia
| | - Zheng Yao Low
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor DE, Malaysia
| | - Sunil Lal
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor DE, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor DE, Malaysia
| |
Collapse
|
14
|
Tenforde MW, Noah KP, O'Halloran AC, Kirley PD, Hoover C, Alden NB, Armistead I, Meek J, Yousey-Hindes K, Openo KP, Witt LS, Monroe ML, Ryan PA, Falkowski A, Reeg L, Lynfield R, McMahon M, Hancock EB, Hoffman MR, McGuire S, Spina NL, Felsen CB, Gaitan MA, Lung K, Shiltz E, Thomas A, Schaffner W, Talbot HK, Crossland MT, Price A, Masalovich S, Adams K, Holstein R, Sundaresan D, Uyeki TM, Reed C, Bozio CH, Garg S. Timing of Influenza Antiviral Therapy and Risk of Death in Adults Hospitalized With Influenza-Associated Pneumonia, Influenza Hospitalization Surveillance Network (FluSurv-NET), 2012-2019. Clin Infect Dis 2025; 80:461-468. [PMID: 39172994 PMCID: PMC11847407 DOI: 10.1093/cid/ciae427] [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: 04/26/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Pneumonia is common in adults hospitalized with laboratory-confirmed influenza, but the association between timeliness of influenza antiviral treatment and severe clinical outcomes in patients with influenza-associated pneumonia is not well characterized. METHODS We included adults aged ≥18 years hospitalized with laboratory-confirmed influenza and a discharge diagnosis of pneumonia over 7 influenza seasons (2012-2019) sampled from a multistate population-based surveillance network. We evaluated 3 treatment groups based on timing of influenza antiviral initiation relative to admission date (day 0, day 1, days 2-5). Baseline characteristics and clinical outcomes were compared across groups using unweighted counts and weighted percentages accounting for the complex survey design. Logistic regression models were generated to evaluate the association between delayed treatment and 30-day all-cause mortality. RESULTS A total of 26 233 adults were sampled in the analysis. Median age was 71 years and most (92.2%) had ≥1 non-immunocompromising condition. Overall, 60.9% started antiviral treatment on day 0, 29.5% on day 1, and 9.7% on days 2-5 (median, 2 days). Baseline characteristics were similar across groups. Thirty-day mortality occurred in 7.5%, 8.5%, and 10.2% of patients who started treatment on day 0, day 1, and days 2-5, respectively. Compared to those treated on day 0, adjusted odds ratio for death was 1.14 (95% confidence interval [CI], 1.01-1.27) in those starting treatment on day 1 and 1.40 (95% CI, 1.17-1.66) in those starting on days 2-5. CONCLUSIONS Delayed initiation of antiviral treatment in patients hospitalized with influenza-associated pneumonia was associated with higher risk of death, highlighting the importance of timely initiation of antiviral treatment at admission.
Collapse
Affiliation(s)
- Mark W Tenforde
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kameela P Noah
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Alissa C O'Halloran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Pam Daily Kirley
- California Emerging Infections Program, Oakland, California, USA
| | - Cora Hoover
- California Department of Public Health, Richmond, Virginia, USA
| | - Nisha B Alden
- Colorado Department of Public Health and Environment, Denver, Colorado, USA
| | - Isaac Armistead
- Colorado Department of Public Health and Environment, Denver, Colorado, USA
| | - James Meek
- Connecticut Emerging Infections Program, Yale School of Public Health, New Haven, Connecticut, USA
| | - Kimberly Yousey-Hindes
- Connecticut Emerging Infections Program, Yale School of Public Health, New Haven, Connecticut, USA
| | - Kyle P Openo
- Georgia Department of Public Health, Georgia Emerging Infections Program, Atlanta, Georgia, USA
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Research, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Lucy S Witt
- Georgia Department of Public Health, Georgia Emerging Infections Program, Atlanta, Georgia, USA
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maya L Monroe
- Maryland Department of Health, Emerging Infections Program, Baltimore, Maryland, USA
| | - Patricia A Ryan
- Maryland Department of Health, Emerging Infections Program, Baltimore, Maryland, USA
| | - Anna Falkowski
- Michigan Department of Health and Human Services, Lansing, Michigan, USA
| | - Libby Reeg
- Michigan Department of Health and Human Services, Lansing, Michigan, USA
| | - Ruth Lynfield
- Health Protection Bureau, Minnesota Department of Health, St Paul, Minnesota, USA
| | - Melissa McMahon
- Health Protection Bureau, Minnesota Department of Health, St Paul, Minnesota, USA
| | - Emily B Hancock
- New Mexico Department of Health, New Mexico Emerging Infections Program, Santa Fe, New Mexico, USA
| | - Marisa R Hoffman
- New Mexico Department of Health, New Mexico Emerging Infections Program, Santa Fe, New Mexico, USA
| | | | - Nancy L Spina
- New York State Department of Health, Albany, New York, USA
| | - Christina B Felsen
- University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Maria A Gaitan
- University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Krista Lung
- Ohio Department of Health, Columbus, Ohio, USA
| | - Eli Shiltz
- Ohio Department of Health, Columbus, Ohio, USA
| | - Ann Thomas
- Public Health Division, Oregon Health Authority, Salem, Oregon, USA
| | - William Schaffner
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - H Keipp Talbot
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Andrea Price
- Salt Lake County Health Department, Salt Lake City, Utah, USA
| | - Svetlana Masalovich
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine Adams
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Rachel Holstein
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Devi Sundaresan
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Timothy M Uyeki
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Carrie Reed
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Catherine H Bozio
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Shikha Garg
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Shirey KA, Joseph J, Coughlan L, Nijhuis H, Varley AW, Blanco JCG, Vogel SN. An adenoviral vector encoding an inflammation-inducible antagonist, HMGB1 Box A, as a novel therapeutic approach to inflammatory diseases. mBio 2025; 16:e0338724. [PMID: 39699172 PMCID: PMC11796352 DOI: 10.1128/mbio.03387-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Influenza, as well as other respiratory viruses, can trigger local and systemic inflammation resulting in an overall "cytokine storm" that produces serious outcomes such as acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). We hypothesized that gene therapy platforms could be useful in these cases if the production of an anti-inflammatory protein reflects the intensity and duration of the inflammatory condition. The recombinant protein would be produced and released only in the presence of the inciting stimulus, avoiding immunosuppression or other unwanted side effects that may occur when treating infectious diseases with anti-inflammatory drugs. To test this hypothesis, we developed AdV.C3-Tat/HIV-Box A, an inflammation-inducible cassette that remains innocuous in the absence of inflammation but releases HMGB1 Box A, an antagonist of high mobility group box 1 (HMGB1), in response to inflammatory stimuli such as lipopolysaccharide (LPS) or influenza virus infection. We report here that this novel inflammation-inducible HMGB1 Box A construct in a non-replicative adenovirus (AdV) vector mitigates lung and systemic inflammation therapeutically in response to influenza infection. We anticipate that this strategy will apply to the treatment of multiple diseases in which HMGB1-mediated signaling is a central driver of inflammation.IMPORTANCEMany inflammatory diseases are mediated by the action of a host-derived protein, HMGB1, on Toll-like receptor 4 (TLR4) to elicit an inflammatory response. We have engineered a non-replicative AdV vector that produces HMGB1 Box A, an antagonist of HMGB1-induced inflammation, under the control of an endogenous complement component C3 (C3) promoter sequence, that is inducible by LPS and influenza in vitro and ex vivo in macrophages (Mϕ) and protects mice and cotton rats therapeutically against infection with mouse-adapted and human non-adapted influenza strains, respectively, in vivo. We anticipate that this novel strategy will apply to the treatment of multiple infectious and non-infectious diseases in which HMGB1-mediated TLR4 signaling is a central driver of inflammation.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - John Joseph
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | | | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Kim J, Kim J, Heo S, Yeom CH, Duong BT, Sung HW, Yeo SJ, Park H, Poo H, Yang J. A low pathogenic avian influenza A/Mallard/South Korea/KNU2019-34/2019 (H1N1) virus has the potential to increase the mammalian pathogenicity. Virol Sin 2025; 40:24-34. [PMID: 39736322 PMCID: PMC11963063 DOI: 10.1016/j.virs.2024.12.005] [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: 07/11/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025] Open
Abstract
Influenza, a highly contagious respiratory infectious disease caused by an influenza virus, is a threat to public health worldwide. Avian influenza viruses (AIVs) have the potential to cause the next pandemic by crossing the species barrier through mutation of viral genome. Here, we investigated the pathogenicity of AIVs obtained from South Korea and Mongolia during 2018-2019 by measuring viral titers in the lungs and extrapulmonary organs of mouse models. In addition, we assessed the pathogenicity of AIVs in ferret models. Moreover, we compared the ability of viruses to replicate in mammalian cells, as well as the receptor-binding preferences of AIV isolates. Genetic analyses were finally performed to identify the genetic relationships and amino acid substitutions between viral proteins during mammalian adaptation. Of the 24 AIV isolates tested, A/Mallard/South Korea/KNU2019-34/2019 (KNU19-34; H1N1) caused severe bodyweight loss and high mortality in mice. The virus replicated in the lungs, kidneys, and heart. Importantly, KNU19-34-infected ferrets showed high viral loads in both nasal washes and lungs. KNU19-34 replicated rapidly in A549 and bound preferentially to human like α2,6-linked sialic acids rather than to avian-like α2,3-linked sialic acids, similar to the pandemic A/California/04/2009 (H1N1) strain. Gene segments of KNU19-34 were distributed in Egypt and Asia lineages from 2015 to 2018, and the virus had several amino acid substitutions compared to H1N1 AIV isolates that were non-pathogenic in mice. Collectively, the data suggest that KNU19-34 has zoonotic potential and the possibility of new mutations responsible for mammalian adaptation.
Collapse
Affiliation(s)
- Jaemoo Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Jungho Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Suhyeon Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Chang-Hun Yeom
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Bao Tuan Duong
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54651, Republic of Korea
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 200-701 24341, Republic of Korea
| | - Seon-Ju Yeo
- Department of Tropical Medicine and Parasitology, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 08826, Republic of Korea; Institute of Endemic Diseases, Medical Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54651, Republic of Korea
| | - Haryoung Poo
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea.
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.
| |
Collapse
|
17
|
Chen AS, Farmer MM, Han L, Runels T, Bade B, Crothers K, Bastian LA, Bazan IS, Bean-Mayberry BA, Brandt CA, Akgün KM. Factors Associated With Influenza Vaccination in a National Veteran Cohort. AJPM FOCUS 2025; 4:100290. [PMID: 39611140 PMCID: PMC11602634 DOI: 10.1016/j.focus.2024.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Introduction Only 53% of American adults receive influenza vaccination, and disparities in vaccination exist among particular racial and ethnic groups. This study determines how race, ethnicity, sex, and rurality are associated with influenza vaccination adherence in a national Veteran Health Affairs Administration cohort. Methods The authors examined differences in documented influenza vaccinations for the 2019-2020 influenza season among Veteran Health Affairs Administration patients in a retrospective cohort study using Veteran Health Affairs Administration administrative electronic health record data. The author used logistic regression to model receipt of influenza vaccination in association with race, ethnicity, sex, and rurality while controlling for clinical diagnoses, demographics, and ambulatory care utilization. The authors also stratified the models by sex and rurality. Results Among 5,943,918 veterans, 48.6% received influenza vaccination. Unadjusted comparisons showed that those who were vaccinated were more likely to be White, to be of male sex, and to be older. Similar proportions of unvaccinated and unvaccinated veterans were from rural settings. In adjusted models, Black race was most strongly associated with decreased vaccination (AOR=0.69; 95% CI=0.69, 0.70), and American Indian/Alaskan Native race also had reduced odds of vaccination (AOR=0.94; 95% CI=0.92, 0.95) compared with White race. Female veterans had increased odds of vaccination (AOR=1.20; 95% CI=1.19, 1.20) compared with men. Rurality (AOR=0.97; 95% CI=0.96, 0.97) was associated with a small decreased odds of vaccination compared with urban. In stratified models, Black veterans were less likely to receive influenza vaccination regardless of sex and rurality than White veterans. American Indian/Alaska Native female veterans had equal odds of vaccination as White female veterans, whereas American Indian/Alaska Native male veterans had reduced odds of vaccination compared with White male veterans. Conclusions During the 2019-2020 influenza season, Black and American Indian/Alaskan Native veterans had lower odds of vaccination. Despite the Veteran Health Affairs Administration's universal approach to healthcare, racial disparities still exist in preventive care.
Collapse
Affiliation(s)
- Alissa S. Chen
- Section of General Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Melissa M. Farmer
- Center for Study of Healthcare Innovation, Implementation, and Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Ling Han
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Tessa Runels
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Brett Bade
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Kristina Crothers
- VA Puget Sound, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington
| | - Lori A. Bastian
- Section of General Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Isabel S. Bazan
- Pulmonary, Critical Care, & Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Bevanne A. Bean-Mayberry
- Center for Study of Healthcare Innovation, Implementation, and Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Cynthia A. Brandt
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, Connecticut
- Department of Emergency Medicine, Yale University School of Medicine, New Haven, Connecticut
- Section of Biomedical Informatics and Data Science, Yale University School of Medicine, New Haven, Connecticut
| | - Kathleen M. Akgün
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, Connecticut
- Pulmonary, Critical Care, & Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
18
|
Yaeger MJ, Leuenberger L, Shaikh SR, Gowdy KM. Omega-3 Fatty Acids and Chronic Lung Diseases: A Narrative Review of Impacts from Womb to Tomb. J Nutr 2025; 155:453-464. [PMID: 39424068 PMCID: PMC12002217 DOI: 10.1016/j.tjnut.2024.10.028] [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: 03/19/2024] [Revised: 08/16/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
The lungs are a mucosal organ constantly exposed to potentially harmful compounds and pathogens. Beyond their role in gas exchange, they must perform a well-orchestrated protective response against foreign invaders. The lungs identify these foreign compounds, respond to them by eliciting an inflammatory response, and restore tissue homeostasis after inflammation to ensure the lungs continue to function. In addition, lung function can be affected by genetics, environmental exposures, and age, leading to pulmonary diseases that infringe on quality of life. Recent studies indicate that diet can influence pulmonary health including the incidence and/or severity of lung diseases. Specifically, long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs) have gained attention because of their potential to reduce inflammation and promote resolution of inflammation. Docosahexaenoic acid and eicosapentaenoic acid are 2 potentially beneficial n-3 PUFAs primarily acquired through dietary intake. Here we review current literature examining the role of n-3 PUFAs and the biological mechanisms by which these fatty acids alter the incidence and pathologies of chronic lung diseases including asthma, chronic obstructive pulmonary disease, and interstitial lung disease. We also highlight the role of n-3 PUFAs in vulnerable populations such as pre/postnatal children, those with obesity, and the elderly. Lastly, we review the impact of n-3 PUFA intake and supplementation to evaluate if increasing consumption can mitigate mechanisms driving chronic lung diseases.
Collapse
Affiliation(s)
- Michael J Yaeger
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States.
| | - Laura Leuenberger
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
19
|
Wu X, Xu L, Xu G, Xu Y, Liu H, Hu Y, Ye X, Huang Q, Tang C, Duan N, Chen X, Yang XD, Zhang W, Zheng Y. Fei-yan-qing-hua decoction exerts an anti-inflammatory role during influenza by inhibiting the infiltration of macrophages and neutrophils through NF-κB and p38 MAPK pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118846. [PMID: 39306208 DOI: 10.1016/j.jep.2024.118846] [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: 08/11/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fei-Yan-Qing-Hua decoction (FYQHD) is an empirical formula that has shown clinical success in treating community-acquired pneumonia (CAP) for two decades. Influenza viral infection is a significant trigger for severe pneumonia, yet the role of FYQHD in treating influenza remains unclear. AIM OF THE STUDY This study aimed to assess the potential efficacy of FYQHD in treating influenza viral infection and to elucidate its underlying mechanisms. MATERIALS AND METHODS The protective effects of FYQHD against influenza were evaluated through survival assessments and pathological analyses. Transcriptomic analysis was performed to identify the genes and pathways influenced by FYQHD in influenza. The anti-inflammatory effects and molecular mechanisms of FYQHD were studied in macrophages stimulated by Toll-like receptor (TLR) 7 ligation in vitro. The key constituents of FYQHD absorbed in mouse sera were identified using untargeted metabolomics, and the anti-inflammatory activity of some of these compounds in macrophages was evaluated using ELISA. RESULTS Our findings demonstrate that FYQHD enhances survival and reduces lung damage in PR8-infected mice, primarily through its anti-inflammatory properties. Lung indexes and organ damage were significantly lower in the PR8 + OSV + FYQHD group compared to the PR8 + OSV group, indicating a potential complementary therapeutic effect of FYQHD and OSV in treating influenza. FYQHD effectively reduced chemokine expression, thereby decreasing the chemotaxis and infiltration of inflammatory monocytes/macrophages and neutrophils in the lungs. The anti-inflammatory effects of FYQHD in macrophages were achieved through the inhibition of NF-κB activation and p38 phosphorylation. The key constituents of FYQHD absorbed in mouse sera were identified, with some, such as wogonin, luteolin, kaempferol, and isorhamnetin, showing anti-inflammatory effects in primary macrophages. CONCLUSION FYQHD demonstrates protective efficacy against influenza and shows promise as an adjuvant therapeutic agent, particularly when used in combination with antiviral drugs like OSV. The potent anti-inflammatory components within FYQHD provide a basis for further exploration in drug research and development aimed at combating influenza.
Collapse
Affiliation(s)
- Xiao Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lirong Xu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanwu Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hui Liu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - You Hu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaolan Ye
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qilin Huang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chenchen Tang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Naifan Duan
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuan Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Dong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wei Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
20
|
Wang Y, Weng L, Wu X, Du B. The role of programmed cell death in organ dysfunction induced by opportunistic pathogens. Crit Care 2025; 29:43. [PMID: 39856779 PMCID: PMC11761187 DOI: 10.1186/s13054-025-05278-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Sepsis is a life-threatening condition resulting from pathogen infection and characterized by organ dysfunction. Programmed cell death (PCD) during sepsis has been associated with the development of multiple organ dysfunction syndrome (MODS), impacting various physiological systems including respiratory, cardiovascular, renal, neurological, hematological, hepatic, and intestinal systems. It is well-established that pathogen infections lead to immune dysregulation, which subsequently contributes to MODS in sepsis. However, recent evidence suggests that sepsis-related opportunistic pathogens can directly induce organ failure by promoting PCD in parenchymal cells of each affected organ. This study provides an overview of PCD in damaged organ and the induction of PCD in host parenchymal cells by opportunistic pathogens, proposing innovative strategies for preventing organ failure in sepsis.
Collapse
Affiliation(s)
- Yangyanqiu Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Weng
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xunyao Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Clinical and Science Investigation Institute, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Bin Du
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Clinical and Science Investigation Institute, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
21
|
Larson PS, Steiner AL, O'Neill MS, Baptist AP, Gronlund CJ. Chronic and infectious respiratory mortality and short-term exposures to four types of pollen taxa in older adults in Michigan, 2006-2017. BMC Public Health 2025; 25:173. [PMID: 39815234 PMCID: PMC11737261 DOI: 10.1186/s12889-025-21386-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
INTRODUCTION Levels of plant-based aeroallergens are rising as growing seasons lengthen and intensify with anthropogenic climate change. Increased exposure to pollens could increase risk for mortality from respiratory causes, particularly among older adults. We determined short-term, lag associations of four species classes of pollen (ragweed, deciduous trees, grass pollen and evergreen trees) with respiratory mortality (all cause, chronic and infectious related) in Michigan, USA. METHODS We obtained records for all Michigan deaths from 2006-2017 from the Michigan Department of Health and Human Services (MDHHS). Deaths from infectious and chronic respiratory-related causes were selected using International Classification of Diseases (ICD-10) codes. Pollen data were obtained from a prognostic model of daily pollen concentrations at 25 km resolution. Case-crossover models with distributed lag non-linear crossbases for pollen were used to estimate associations between lags of daily pollen concentrations with mortality and to explore effect modification by sex and racial groups. RESULTS 127,163 deaths were included in the study. Cumulative daily high concentrations (90th percentile) of deciduous broadleaf, grass and ragweed were associated with all-cause respiratory mortality at early lags with e.g., a 1.81 times higher risk of all respiratory deaths at cumulative 7 day lag exposure to deciduous broadleaf pollen at the 90th percentile (95% confidence interval: 1.04, 3.15). Exposure to high concentrations of grass and ragweed pollens was associated with increased risk for death from chronic respiratory causes. No association was found for any pollen species with death from infectious respiratory causes though there was a positive but non-significant association of exposure to deciduous broadleaf and ragweed pollens. We found no evidence to suggest effect modification by race or sex. CONCLUSIONS Modelled exposures to high concentrations of pollen taxa were associated with increased all-cause and chronic respiratory mortality among older adults. Results suggest that pollen exposure may become more important to respiratory mortality as the temperatures increase and pollen seasons lengthen.
Collapse
Affiliation(s)
- Peter S Larson
- Social Environment and Health Program, Institute for Social Research, University of Michigan, 426 Thompson St., Ann Arbor, MI, 48104, USA.
- Department of Epidemiology, School of Public Health, University of Michigan, 123 Observatory, Ann Arbor, MI, 48104, USA.
| | - Allison L Steiner
- Climate and Space Sciences and Engineering, University of Michigan, 2455 Hayward St., Ann Arbor, MI, 48109, USA
| | - Marie S O'Neill
- Department of Epidemiology, School of Public Health, University of Michigan, 123 Observatory, Ann Arbor, MI, 48104, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 123 Observatory, Ann Arbor, MI, 48104, USA
| | - Alan P Baptist
- Division of Allergy and Clinical Immunology, Henry Ford Health, 1 Ford Place, Detroit, MI, 48202, USA
- Health Behavior and Health Education, University of Michigan School of Public Health, University of Michigan, 123 Observatory, Ann Arbor, MI, 48104, USA
| | - Carina J Gronlund
- Social Environment and Health Program, Institute for Social Research, University of Michigan, 426 Thompson St., Ann Arbor, MI, 48104, USA
- Department of Epidemiology, School of Public Health, University of Michigan, 123 Observatory, Ann Arbor, MI, 48104, USA
| |
Collapse
|
22
|
Chakraborty S, Cheng BYL, Edwards DL, Gonzalez JC, Chiu DKC, Zheng H, Scallan C, Guo X, Tan GS, Coffey GP, Conley PB, Hume PS, Janssen WJ, Byers DE, Mudd PA, Taubenberger J, Memoli M, Davis MM, Chua KF, Diamond MS, Andreakos E, Khatri P, Wang TT. Sialylated IgG induces the transcription factor REST in alveolar macrophages to protect against lung inflammation and severe influenza disease. Immunity 2025; 58:182-196.e10. [PMID: 39541970 PMCID: PMC11735284 DOI: 10.1016/j.immuni.2024.10.002] [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: 06/15/2024] [Revised: 08/15/2024] [Accepted: 10/04/2024] [Indexed: 11/17/2024]
Abstract
While most respiratory viral infections resolve with little harm to the host, severe symptoms arise when infection triggers an aberrant inflammatory response that damages lung tissue. Host regulators of virally induced lung inflammation have not been well defined. Here, we show that enrichment for sialylated, but not asialylated immunoglobulin G (IgG), predicted mild influenza disease in humans and was broadly protective against heterologous influenza viruses in a murine challenge model. Mechanistic studies show that sialylated IgG mediated this protection by inducing the transcription factor repressor element-1 silencing transcription factor (REST), which repressed nuclear factor κB (NF-κB)-driven responses, preventing severe lung inflammation and protecting lung function during influenza infection. Therapeutic administration of a recombinant, sialylated Fc molecule in clinical development similarly activated REST and protected against severe influenza disease, demonstrating that this pathway could be clinically harnessed. Overall, induction of REST through sialylated IgG signaling is a strategy to limit inflammatory disease sequelae in infections caused by antigenically distinct influenza strains.
Collapse
Affiliation(s)
- Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bowie Yik-Ling Cheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Desmond L Edwards
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Gonzalez
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Kung-Chun Chiu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Courtney Scallan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinrong Guo
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gene S Tan
- J. Craig Venter Institute, La Jolla, San Diego, CA 92037, USA; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA 92037, USA
| | - Greg P Coffey
- Nuvig Therapeutics Inc., Redwood City, CA 94061, USA
| | | | - Patrick S Hume
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - William J Janssen
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology and Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jeffery Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Matthew Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; HHMI, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin F Chua
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305, USA; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Center for Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
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
|
24
|
Srivastava S, Jayaswal N, Kumar S, Rao GSNK, Budha RR, Mohanty A, Mehta R, Apostolopoulos V, Sah S, Bonilla-Aldana DK, Ulloque-Badaracco R, Rodriguez-Morales AJ. Targeting H3N2 influenza: advancements in treatment and vaccine strategies. Expert Rev Anti Infect Ther 2025; 23:5-18. [PMID: 39688174 DOI: 10.1080/14787210.2024.2443920] [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: 07/17/2024] [Revised: 10/20/2024] [Accepted: 12/15/2024] [Indexed: 12/18/2024]
Abstract
INTRODUCTION The emergence of the H3N2 influenza virus in 1968 marked a significant event as it crossed the species barrier. This shift led to a pandemic, resulting in the deaths of one million people globally and highlighting the virus's severe impact on older individuals due to antigenic drift. AREA COVERED This review comprehensively examines the virological characteristics, evolutionary trends, and global epidemiology of the Influenza A (H3N2) virus. It delves into vaccination strategies, antiviral interventions, and emerging diagnostic approaches. The impact of antigenic variation on vaccine design and effectiveness, seasonal outbreak patterns, and pandemic potential are explored. Additionally, the interplay between viral factors and host immune responses is assessed. Researchers are actively investigating innovative strategies to enhance vaccine efficacy against H3N2 mutations, such as precise antigenic material administration, controlled release patterns, understanding immune system mechanisms, and glycan engineering. EXPERT OPINION The ongoing mutational dynamics of the H3N2 virus necessitate regular vaccine updates, as advocated by the WHO. Research in the Western Pacific region underscores the need for heightened awareness and effective control strategies. Evaluating antiviral therapies and addressing drug resistance requires multidisciplinary approaches involving researchers, healthcare professionals, and policymakers. This comprehensive understanding of H3N2 is vital for improving public health interventions and preparing for future influenza challenges.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
| | - Nandani Jayaswal
- Faculty of Pharmaceutical Sciences, Mahayogi Gorakhnath University Gorakhpur, Gorakhpur, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, India
| | - Roja Rani Budha
- Department of Pharmacology, H.K. College of Pharmacy, Jogeshwari, Mumbai, India
| | - Aroop Mohanty
- Department of Clinical Microbiology, All India Institute of Medical Sciences, Gorakhpur, India
| | - Rachana Mehta
- Dr Lal PathLabs Nepal, Kathmandu, Nepal
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, India
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora VIC 3083, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, VIC, Australia
| | - Sanjit Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed-to-be-University), Pimpri, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Medicine, Korea Universtiy, Seoul, South Korea
| | | | | | - Alfonso J Rodriguez-Morales
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Pereira, Colombia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
25
|
Iglesias-Ussel MD, O’Grady N, Anderson J, Mitsis PG, Burke TW, Henao R, Scavetta J, Camilleri C, Naderi S, Carittini A, Perelman M, Myers RA, Ginsburg GS, Ko ER, McClain MT, van Westrienen J, Tsalik EL, Tillekeratne LG, Woods CW. A Rapid Host Response Blood Test for Bacterial/Viral Infection Discrimination Using a Portable Molecular Diagnostic Platform. Open Forum Infect Dis 2025; 12:ofae729. [PMID: 39758742 PMCID: PMC11697109 DOI: 10.1093/ofid/ofae729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/10/2024] [Indexed: 01/07/2025] Open
Abstract
Background Difficulty discriminating bacterial versus viral etiologies of infection drives unwarranted antibacterial prescriptions and, therefore, antibacterial resistance. Methods Utilizing a rapid portable test that measures peripheral blood host gene expression to discriminate bacterial and viral etiologies of infection (the HR-B/V assay on Biomeme's polymerase chain reaction-based Franklin platform), we tested 3 cohorts of subjects with suspected infection: the HR-B/V training cohort, the HR-B/V technical correlation cohort, and a coronavirus disease 2019 cohort. Results The Biomeme HR-B/V test showed very good performance at discriminating bacterial and viral infections, with a bacterial model accuracy of 84.5% (95% confidence interval [CI], 80.8%-87.5%), positive percent agreement (PPA) of 88.5% (95% CI, 81.3%-93.2%), negative percent agreement (NPA) of 83.1% (95% CI, 78.7%-86.7%), positive predictive value of 64.1% (95% CI, 56.3%-71.2%), and negative predictive value of 95.5% (95% CI, 92.4%-97.3%). The test showed excellent agreement with a previously developed BioFire HR-B/V test, with 100% (95% CI, 85.7%-100.0%) PPA and 94.9% (95% CI, 86.1%-98.3%) NPA for bacterial infection, and 100% (95% CI, 93.9%-100.0%) PPA and 100% (95% CI, 85.7%-100.0%) NPA for viral infection. Among subjects with acute severe acute respiratory syndrome coronavirus 2 infection of ≤7 days, accuracy was 93.3% (95% CI, 78.7%-98.2%) for 30 outpatients and 75.9% (95% CI, 57.9%-87.8%) for 29 inpatients. Conclusions The Biomeme HR-B/V test is a rapid, portable test with high performance at identifying patients unlikely to have bacterial infection, offering a promising antibiotic stewardship strategy that could be deployed as a portable, laboratory-based test.
Collapse
Affiliation(s)
| | - Nicholas O’Grady
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jack Anderson
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Thomas W Burke
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ricardo Henao
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biostatistics and Informatics, Duke University, Durham, North Carolina, USA
| | | | | | | | | | | | - Rachel A Myers
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Geoffrey S Ginsburg
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- All of Us Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Emily R Ko
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Hospital Medicine, Division of General Internal Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Micah T McClain
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | | | - Ephraim L Tsalik
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Danaher Corporation, Washington, District of Columbia, USA
| | - L Gayani Tillekeratne
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| | - Christopher W Woods
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Biomeme, Inc, Philadelphia, Pennsylvania, USA
- Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
26
|
Rosero CI, Gravenstein S, Saade EA. Influenza and Aging: Clinical Manifestations, Complications, and Treatment Approaches in Older Adults. Drugs Aging 2025; 42:39-55. [PMID: 39775605 DOI: 10.1007/s40266-024-01169-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Influenza, a highly contagious respiratory viral illness, poses significant global health risks, particularly affecting older and those with chronic health conditions. Influenza viruses, primarily types A and B, are responsible for seasonal human infections and exhibit a propensity for antigenic drift and shift, contributing to seasonal epidemics and pandemics. The severity of influenza varies, but severe cases often lead to pneumonia, acute respiratory distress syndrome, and multiorgan failure. Older adults, especially those over 65 years of age, face increased risks of immune senescence, chronic comorbidities, and decreased vaccine efficacy. Globally, influenza affects millions of people annually, with significant morbidity and mortality among older. Epidemiological patterns vary with climate, and risk factors include age, immunocompromised status, and preexisting chronic conditions. In older adults, influenza frequently results in hospitalization and death, which is exacerbated by immunosenescence and biological organ changes associated with aging. Clinical manifestations range from mild symptoms to severe complications such as viral pneumonia and multiorgan failure. Diagnosis often relies on antigen or molecular tests, with radiological examination aiding in severe cases. Treatment primarily involves antiviral agents, such as oseltamivir and peramivir, with the greatest benefit observed when initiated early. Management of severe cases may require hospitalization and supportive care, including addressing complications, such as secondary bacterial infections and cardiovascular events. This article highlights the need for improved vaccination strategies and novel treatments, including monoclonal antibodies and adoptive T cell therapies, to better manage severe influenza infections in vulnerable populations such as older.
Collapse
Affiliation(s)
| | - Stefan Gravenstein
- Brown University School of Public Health, Providence, RI, 02903, USA
- Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
- Providence Veterans Affairs Medical Center, Providence, RI, 02908, USA
| | - Elie A Saade
- University Hospitals of Cleveland, 11100 Euclid Ave, Mailstop 5083, Cleveland, OH, 44106, USA.
- Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
27
|
Xu JQ, Zhang WY, Fu JJ, Fang XZ, Gao CG, Li C, Yao L, Li QL, Yang XB, Ren LH, Shu HQ, Peng K, Wu Y, Zhang DY, Qiu Y, Zhou X, Yao YM, Shang Y. Viral sepsis: diagnosis, clinical features, pathogenesis, and clinical considerations. Mil Med Res 2024; 11:78. [PMID: 39676169 PMCID: PMC11648306 DOI: 10.1186/s40779-024-00581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Sepsis, characterized as life-threatening organ dysfunction resulting from dysregulated host responses to infection, remains a significant challenge in clinical practice. Despite advancements in understanding host-bacterial interactions, molecular responses, and therapeutic approaches, the mortality rate associated with sepsis has consistently ranged between 10 and 16%. This elevated mortality highlights critical gaps in our comprehension of sepsis etiology. Traditionally linked to bacterial and fungal pathogens, recent outbreaks of acute viral infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), among other regional epidemics, have underscored the role of viral pathogenesis in sepsis, particularly when critically ill patients exhibit classic symptoms indicative of sepsis. However, many cases of viral-induced sepsis are frequently underdiagnosed because standard evaluations typically exclude viral panels. Moreover, these viruses not only activate conventional pattern recognition receptors (PRRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) but also initiate primary antiviral pathways such as cyclic guanosine monophosphate adenosine monophosphate (GMP-AMP) synthase (cGAS)-stimulator of interferon genes (STING) signaling and interferon response mechanisms. Such activations lead to cellular stress, metabolic disturbances, and extensive cell damage that exacerbate tissue injury while leading to a spectrum of clinical manifestations. This complexity poses substantial challenges for the clinical management of affected cases. In this review, we elucidate the definition and diagnosis criteria for viral sepsis while synthesizing current knowledge regarding its etiology, epidemiology, and pathophysiology, molecular mechanisms involved therein as well as their impact on immune-mediated organ damage. Additionally, we discuss clinical considerations related to both existing therapies and advanced treatment interventions, aiming to enhance the comprehensive understanding surrounding viral sepsis.
Collapse
Affiliation(s)
- Ji-Qian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wan-Ying Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Ji Fu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Gang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chang Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lu Yao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi-Lan Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Bo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Le-Hao Ren
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua-Qing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Ying Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, 430072, China
| | - Ding-Yu Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yang Qiu
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Xi Zhou
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and the Fourth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
28
|
Lin X, Lin J, Ji L, Zhang J, Zhang Y, Hong J, Li G, Lin X. Protective effect of Haoqin Qingdan decoction on pulmonary and intestinal injury in mice with influenza viral pneumonia. Front Pharmacol 2024; 15:1449322. [PMID: 39712501 PMCID: PMC11658977 DOI: 10.3389/fphar.2024.1449322] [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: 06/14/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Background Haoqin Qingdan decoction (HQQD), composed of eleven herbs, is a traditional Chinese formula widely recognized for its efficacy in treating pulmonary inflammation induced by viral infections. Despite its extensive use, the potential pulmonary and intestinal protective effects of HQQD on influenza viral pneumonia (IVP) and the underlying molecular mechanisms remain unclear. Materials and Methods Ultra-high-performance liquid chromatography coupled with mass spectrometry (UHPLC-MS) was employed to identify the major chemical constituents of the prescription. Subsequently, network analysis was conducted to predict the potential therapeutic targets of HQQD in IVP. The mechanisms by which HQQD mitigates lung and intestinal damage were further elucidated by assessing NP protein expression, inflammatory factors, TLR7/MyD88/NF-κB signaling pathway mRNAs and proteins, and through intestinal flora analysis. Results The protective effects of HQQD on pulmonary and intestinal injuries induced by IVP were thoroughly investigated using comprehensive network analysis, signaling pathway validation, and gut microflora analysis. UHPLC-MS analysis identified the primary chemical constituents. Validation experiments demonstrated a significant reduction in NP protein expression in the lungs. HQQD notably alleviated immune damage in the lungs and intestines of mice by inhibiting NP protein expression and the release of inflammatory factors such as interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ); downregulating the expression levels of TLR7, MyD88, and phospho-NF-κB p65 (p-p65); lowering serum LPS levels; and reducing the relative abundance of Proteobacteria. Conclusion HQQD exerts therapeutic effects against influenza viral pneumonia through antiviral and anti-inflammatory mechanisms and by remodeling the intestinal flora. This study provides initial insights into the "gut-lung" axis mechanism of HQQD in combating respiratory influenza virus infection.
Collapse
Affiliation(s)
- Xi Lin
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian Lin
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lichun Ji
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaona Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yezi Zhang
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junbin Hong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Li
- Chinese Medicine Guangdong Laboratory, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingdong Lin
- The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
29
|
Li H, Zong Y, Li J, Zhou Z, Chang Y, Shi W, Guo J. Research trends and hotspots on global influenza and inflammatory response based on bibliometrics. Virol J 2024; 21:313. [PMID: 39623458 PMCID: PMC11613568 DOI: 10.1186/s12985-024-02588-4] [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: 06/12/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
The influenza virus is considered as a kind of significant zoonotic infectious disease identified to date, with severe infections in humans characterized by excessive inflammation and tissue damage, usually resulting in serious complications. Although the molecular mechanisms underlying inflammation after influenza infection have been extensively studied, bibliometric analysis on the research hotspots and developing trends in this field has not been published heretofore. Articles related to influenza and inflammatory response were retrieved from the Web of Science Core Collection (WoSCC) database (1992-2024) and analyzed using various visualization tools. Finally, this study collected a total of 2,176 relevant articles, involving 13,184 researchers, 2,647 institutions, 78 countries/regions, and published in 723 journals. Most articles were published in the United States (928 articles), China (450 articles) and the United Kingdom (158 articles). Ross Vlahos was the most productive author. Furthermore, some journals, such as PLoS One and Frontiers in Immunology, made much contribution to the topic. The future research trends include airway stem cells and neuroendocrine cells as new directions for the treatment of influenza complications, as well as measures related to prevention, treatment, and research and development based on the COVID-19 pandemic. Through bibliometric analysis and summary of inflammatory response of influenza-related articles, this study ultimately summarizes new directions for preventing and treating influenza.
Collapse
Affiliation(s)
- Hui Li
- Center for Xin'an Medicine and Modemization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yanping Zong
- Center for Xin'an Medicine and Modemization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jiajie Li
- Key Laboratory of Xin'an Medical Education, Anhui University of Traditional Chinese Medicine, Hefei, 230012, China
| | - Zheng Zhou
- Key Laboratory of Xin'an Medical Education, Anhui University of Traditional Chinese Medicine, Hefei, 230012, China
| | - Yonglong Chang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Weibing Shi
- Center for Xin'an Medicine and Modemization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China.
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China.
| | - Jinchen Guo
- Center for Xin'an Medicine and Modemization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
30
|
Kodaira M, Hasan MS, Grossman Y, Guerrero C, Guo L, Liu A, Therrien J, Marelli A. Risk of cardiovascular events after influenza infection-related hospitalizations in adults with congenital heart disease: A nationwide population based study. Am Heart J 2024; 278:93-105. [PMID: 39241939 DOI: 10.1016/j.ahj.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Cardiovascular complications due to viral infection pose a significant risk in vulnerable patients such as those with congenital heart disease (CHD). Limited data exists regarding the incidence of influenza and its impact on cardiovascular outcomes among this specific patient population. METHODS A retrospective cohort study was designed using the Canadian Congenital Heart Disease (CanCHD) database-a pan-Canadian database of CHD patients with up to 35 years of follow-up. CHD patients aged 40 to 65 years with influenza virus-associated hospitalizations between 2010 and 2017 were identified and 1:1 matched with CHD patients with limb fracture hospitalizations on age and calendar time. Our primary endpoint was cardiovascular complications: heart failure, acute myocardial infarction, atrial arrhythmia, ventricular arrhythmia, heart block, myocarditis, and pericarditis. RESULTS Of the 303 patients identified with incident influenza virus-associated hospitalizations, 255 were matched to 255 patients with limb fracture hospitalizations. Patients with influenza virus-related hospitalizations showed significantly higher cumulative probability of cardiovascular complications at 1 year (0.16 vs. 0.03) and 5 years (0.33 vs. 0.15) compared to patients hospitalized with bone fracture. Time-dependent hazard function modeling demonstrated a significantly higher risk of cardiovascular complications within 9 months postdischarge for influenza-related hospitalizations. This association was confirmed by Cox regression model (average hazard ratio throughout follow-up: 2.48; 95% CI: 1.59-3.84). CONCLUSIONS This pan-Canadian cohort study of adults with CHD demonstrated an association between influenza virus-related hospitalization and risk of cardiovascular complications during the 9 months post discharge. This data is essential in planning surveillance strategies to mitigate adverse outcomes and provides insights into interpreting complication rates of other emerging pathogens, such as COVID-19.
Collapse
Affiliation(s)
- Masaki Kodaira
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada; Beth Raby Adult Congenital Heart Disease Clinic, Jewish General Hospital, Montréal, Québec, Canada
| | - Mohammad Sazzad Hasan
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada
| | - Yoni Grossman
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada
| | - Carlos Guerrero
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada; Beth Raby Adult Congenital Heart Disease Clinic, Jewish General Hospital, Montréal, Québec, Canada
| | - Liming Guo
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada
| | - Aihua Liu
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada
| | - Judith Therrien
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada; Beth Raby Adult Congenital Heart Disease Clinic, Jewish General Hospital, Montréal, Québec, Canada
| | - Ariane Marelli
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
31
|
Yu J, Lu Z, Chen B, He X, Zhao W, Cao H, Li Y, Peng G, Ou J, Ma Q, Yu L, Liu J. Liang-Ge-San protects against viral infection-induced acute lung injury through inhibiting α7nAChR-mediated mitophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156231. [PMID: 39566410 DOI: 10.1016/j.phymed.2024.156231] [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: 09/03/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is the main cause of death in clinical respiratory virus infection. Liang-Ge-San (LGS), a famous traditional Chinese formula, has been proved to be effective in treating ALI caused by lipopolysaccharide. However, the effects of LGS on ALI induced by viral infections remain uncertain. PURPOSE To investigate the effect and mechanism of action of LGS on viral infection-induced ALI. METHODS The inhibitory effects of LGS on virus-induced inflammation in vitro were evaluated by qRT-PCR and ELISA. The protein expression of α7nAChR was examined by Western blotting. α7nAChR was inhibited by the transfection of siRNA or methyllycaconitine citrate (MLA, an α7nAChR inhibitor) to investigate the role of α7nAChR in the anti-inflammatory effect of LGS. Adoptive culture and co-culture systems of macrophages RAW264.7 and alveolar epithelial cells MLE-12 were established to mimic their interaction. Western blotting, immunofluorescence, flow cytometry and transmission electron microscopy were used to examine the effects of LGS on mitophagy inhibition. In vivo, ALI mouse models induced by SARS-CoV-2, H1N1 or Poly(I:C) infection were established to explore the therapeutic effect and mechanism of LGS. RESULTS LGS reduced the release of IL-6, TNF-α and IL-1β and increased the expression of α7nAChR in virus-infected RAW264.7 cells. The blockage of α7nAChR counteracted the anti-inflammatory effect of LGS. Moreover, LGS significantly inhibited autophagy in MLE-12 cells induced by Poly(I:C) in adoptive culture and co-culture systems of RAW264.7 and MLE-12 cells, which could be attenuated after the inhibition of α7nAChR in RAW264.7 cells by decreasing the secretion of IL-6, TNF-α and IL-1β. Further study showed that LGS suppressed TNF-α-induced mitochondrial damage and mitophagy by inhibiting the generation of ROS in MLE-12 cells. In vivo, LGS significantly prolonged the survival time, alleviated pathological injury and acute inflammation of ALI mice induced by SARS-CoV-2, H1N1 or Poly(I:C) infection which were associated with the inhibition of α7nAChR-mediated mitophagy. CONCLUSION This study first demonstrates that LGS inhibits virus infection-induced inflammation in RAW246.7 cells by increasing the expression of α7nAChR, thereby inhibiting mitophagy induction in MLE-12 cells to alleviate ALI. This work indicates that LGS may serve as a candidate drug for treating ALI/ARDS caused by viral infection.
Collapse
Affiliation(s)
- Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Bing Chen
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Xuemei He
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Huihui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Yuhua Li
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Gefei Peng
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Jinying Ou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510182, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China.
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, PR China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, PR China.
| |
Collapse
|
32
|
Salatzki J, Ochs A, Weberling LD, Heins J, Zahlten M, Whayne JG, Stehning C, Giannitsis E, Denkinger CM, Merle U, Buss SJ, Steen H, André F, Frey N. Absence of cardiac impairment in patients after severe acute respiratory syndrome coronavirus type 2 infection: A long-term follow-up study. J Cardiovasc Magn Reson 2024; 26:101124. [PMID: 39549839 DOI: 10.1016/j.jocmr.2024.101124] [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: 05/11/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Concerns exist that long-term cardiac alterations occur after severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection, particularly in patients who were hospitalized in the acute phase or who remain symptomatic. This study investigates potential long-term functional and morphological alterations after SARS-CoV-2 infection. METHODS The authors of this study investigated patients after SARS-CoV-2 infection by using a mobile 1.5T clinical magnetic resonance scanner for cardiac alterations. Cardiac function and dimensions were assessed using a highly efficient cardiac magnetic resonance protocol, which included cine sequences, global longitudinal and circumferential strain assessed by fast-Strain-ENCoded imaging, and T1 and T2 mapping. We assessed symptoms through a questionnaire. Patients were compared with a control group matched for age, gender, body mass index, and body surface area. RESULTS Median follow-up time was 395 (192-408) days. The final population included 183 participants (age 48.4 ± 14.3 years, 48.1% male (88/183)). During the acute phase of SARS-CoV-2 infection, 27 patients were hospital-admitted. Forty-two patients reported persistent symptoms (shortness of breath, chest pain, palpitations, or leg edema), and 63 reported impaired exercise tolerance. Left ventricular (LV) functional and morphological parameters were within the normal range. T1- and T2-relaxation times were also within the normal range, indicating that the presence of myocardial edema or fibrosis was unlikely. Persistently symptomatic patients showed a slightly reduced indexed LV stroke volume. Functional parameters remained normal in patients who were hospitalized for SARS-CoV-2, persistently symptomatic, or with ongoing impaired exercise tolerance. CONCLUSION Irrespective of ongoing symptoms or severity of prior illness, patients who have recovered from SARS-CoV-2 infection demonstrate normal functional and morphological cardiac parameters. Long-term cardiac changes due to SARS-CoV-2 infection appear to be rare.
Collapse
Affiliation(s)
- Janek Salatzki
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| | - Andreas Ochs
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| | - Lukas D Weberling
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| | - Jannick Heins
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
| | - Marc Zahlten
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
| | - James G Whayne
- Myocardial Solutions Inc., Morrisville, North Carolina, USA.
| | | | - Evangelos Giannitsis
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine, University Hospital Heidelberg, Heidelberg, Germany; German Center of Infection Research, partner site Heidelberg, Heidelberg, Germany.
| | - Uta Merle
- Department of Gastroenterology, Infectious Diseases and Intoxication, University Hospital Heidelberg, Heidelberg, Germany.
| | | | - Henning Steen
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; medneo, Hamburg, Germany.
| | - Florian André
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
33
|
Gao T, Liu J, Huang N, Zhou Y, Li C, Chen Y, Hong Z, Deng X, Liang X. Sangju Cold Granule exerts anti-viral and anti-inflammatory activities against influenza A virus in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118521. [PMID: 38969152 DOI: 10.1016/j.jep.2024.118521] [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: 04/18/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sangju Cold Granule (SJCG) is a classical traditional Chinese medicine (TCM) prescription described in "Item Differentiation of Warm Febrile Diseases". Historically, SJCG was employed to treat respiratory illnesses. Despite its popular usage, the alleviating effect of SJCG on influenza A virus infection and its mechanisms have not been fully elucidated. AIM OF THE STUDY Influenza is a severe respiratory disease that threatens human health. This study aims to assess the therapeutic potential of SJCG and the possible molecular mechanism underlying its activity against influenza A virus in vitro and in vivo. MATERIALS AND METHODS Ultrahigh-performance liquid chromatography (UPLC)-Q-Exactive was used to identify the components of SJCG. The 50% cytotoxic concentration of SJCG in MDCK and A549 cells were determined using the CCK-8 assay. The activity of SJCG against influenza A virus H1N1 was evaluated in vitro using plaque reduction and progeny virus titer reduction assays. RT-qPCR was performed to obtain the expression levels of inflammatory mediators and the transcriptional regulation of RIG-I and MDA5 in H1N1-infected A549 cells. Then, the mechanism of SJCG effect on viral replication and inflammation was further explored by measuring the expressions of proteins of the RIG-I/NF-kB/IFN(I/III) signaling pathway by Western blot. The impact of SJCG was explored in vivo in an intranasally H1N1-infected BALB/c mouse pneumonia model treated with varying doses of SJCG. The protective role of SJCG in this model was evaluated by survival, body weight monitoring, lung viral titers, lung index, lung histological changes, lung inflammatory mediators, and peripheral blood leukocyte count. RESULTS The main SJCG chemical constituents were flavonoids, carbohydrates and glycosides, amino acids, peptides, and derivatives, organic acids and derivatives, alkaloids, fatty acyls, and terpenes. The CC50 of SJCG were 24.43 mg/mL on MDCK cells and 20.54 mg/mL on A549 cells, respectively. In vitro, SJCG significantly inhibited H1N1 replication and reduced the production of TNF-α, IFN-β, IL-6, IL-8, IL-13, IP-10, RANTES, TRAIL, and SOCS1 in infected A549 cells. Intracellularly, SJCG reduced the expression of RIG-I, MDA5, P-NF-κB P65 (P-P65), P-IκBα, P-STAT1, P-STAT2, and IRF9. In vivo, SJCG enhanced the survival rate and decreased body weight loss in H1N1-infected mice. Mice with H1N1-induced pneumonia treated with SJCG showed a lower lung viral load and lung index than untreated mice. SJCG effectively alleviated lung damage and reduced the levels of TNF-α, IFN-β, IL-6, IP-10, RANTES, and SOCS1 in lung tissue. Moreover, SJCG significantly ameliorated H1N1-induced leukocyte changes in peripheral blood. CONCLUSIONS SJCG significantly reduced influenza A virus and virus-mediated inflammation through inhibiting the RIG-I/NF-kB/IFN(I/III) signaling pathway. Thus, SJCG could provide an effective TCM for influenza treatment.
Collapse
Affiliation(s)
- Taotao Gao
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jinbing Liu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, China; Department of Ultrasound Medicine, Liwan Central Hospital of Guangzhou, 35 Liwan Road, Guangzhou, 510000, Guangdong, China
| | - Nan Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yingxuan Zhou
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Conglin Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yintong Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zifan Hong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaoyan Deng
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoli Liang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
34
|
Kyaw MH, Chen SB, Wu S, Foo CY, Welch V, Boikos C, Jagun O. Systematic Review on Influenza Burden in Emerging Markets in 2018-2023-An Evidence Update to Guide Influenza Vaccination Recommendations. Vaccines (Basel) 2024; 12:1251. [PMID: 39591154 PMCID: PMC11599016 DOI: 10.3390/vaccines12111251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Influenza is a contagious respiratory illness responsible for seasonal epidemics and with potential to cause pandemics. The decline in influenza-related studies published since 2018 resulted in data gaps, particularly in emerging markets. Methods: This systematic review searched for studies in six databases and gray literature sources to define the clinical burden of influenza and influenza-like illness (ILIs) and their associated sequelae among humans across emerging markets. Eligible studies were published in English, Spanish, or Chinese between January 2018 and September 2023 and conducted in Asia, the Middle East, Africa, and Latin America. Results: In total, 256 articles were included, mostly on lab-confirmed influenza infections (n = 218). Incidences of lab-confirmed influenza cases in Asia (range 540-1279 cases/100,000 persons) and Sub-Saharan Africa (range 34,100-47,800 cases/100,000 persons) were higher compared to Latin America (range 0.7-112 cases/100,000 persons) and the Middle East and North Africa (range 0.1-10 cases/100,000 persons). Proportions of lab-confirmed influenza cases and influenza-associated outcomes (i.e., hospitalization, ICU admission and death) varied widely across regions. Temporal variation in influenza trend was observed before and during the COVID-19 pandemic. Conclusions: In conclusion, influenza causes significant disease burden in emerging markets. Robust large real-world studies using a similar methodology are needed to have more accurate estimates and compare studies within age groups and regions. Continuous monitoring of influenza epidemiology is important to inform vaccine programs in emerging markets with heavy influenza disease burden.
Collapse
Affiliation(s)
- Moe H. Kyaw
- Pfizer Inc., Collegeville, PA 19426, USA; (V.W.); (C.B.)
| | - Sophie Bozhi Chen
- Real World Solutions, IQVIA, Singapore 079906, Singapore; (S.B.C.); (S.W.); (C.Y.F.)
| | - Shishi Wu
- Real World Solutions, IQVIA, Singapore 079906, Singapore; (S.B.C.); (S.W.); (C.Y.F.)
| | - Chee Yoong Foo
- Real World Solutions, IQVIA, Singapore 079906, Singapore; (S.B.C.); (S.W.); (C.Y.F.)
| | - Verna Welch
- Pfizer Inc., Collegeville, PA 19426, USA; (V.W.); (C.B.)
| | | | | |
Collapse
|
35
|
Nakhaie M, Rukerd MRZ, Shahpar A, Pardeshenas M, Khoshnazar SM, Khazaeli M, Bashash D, Nezhad NZ, Charostad J. A Closer Look at the Avian Influenza Virus H7N9: A Calm before the Storm? J Med Virol 2024; 96:e70090. [PMID: 39601174 DOI: 10.1002/jmv.70090] [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: 05/27/2024] [Revised: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The avian influenza A (H7N9) virus, which circulates in wild birds and poultry, has been a major concern for public health since it was first discovered in China in 2013 due to its demonstrated ability to infect humans, causing severe respiratory illness with high mortality rates. According to the World Health Organization (WHO), a total of 1568 human infections with 616 fatal cases caused by novel H7N9 viruses have been reported in China from early 2013 to January 2024. This manuscript provides a comprehensive review of the virology, evolutionary patterns, and pandemic potential of H7N9. The H7N9 virus exhibits a complex reassortment history, receiving genes from H9N2 and other avian influenza viruses. The presence of certain molecular markers, such as mutations in the hemagglutinin and polymerase basic protein 2, enhances the virus's adaptability to human hosts. The virus activates innate immune responses through pattern recognition receptors, leading to cytokine production and inflammation. Clinical manifestations range from mild to severe, with complications including pneumonia, acute respiratory distress syndrome, and multiorgan failure. Diagnosis relies on molecular assays such as reverse transcription-polymerase chain reaction. The increasing frequency of human infections, along with the virus's ability to bind to human receptors and cause severe disease, highlights its pandemic potential. Continued surveillance, vaccine development, and public health measures are crucial to limit the risk posed by H7N9. Understanding the virus's ecology, transmission dynamics, and pathogenesis is essential for developing effective prevention and control strategies.
Collapse
Affiliation(s)
- Mohsen Nakhaie
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Rezaei Zadeh Rukerd
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Pardeshenas
- Department of Microbiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mana Khazaeli
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Zeinali Nezhad
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Javad Charostad
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
36
|
Rago F, Melo EM, Miller LM, Duray AM, Batista Felix F, Vago JP, de Faria Gonçalves AP, Angelo ALPM, Cassali GD, de Gaetano M, Brennan E, Owen B, Guiry P, Godson C, Alcorn JF, Teixeira MM. Treatment with lipoxin A 4 improves influenza A infection outcome, induces macrophage reprogramming, anti-inflammatory and pro-resolutive responses. Inflamm Res 2024; 73:1903-1918. [PMID: 39214890 DOI: 10.1007/s00011-024-01939-9] [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: 05/28/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Influenza A is a virus from the Orthomixoviridae family responsible for high lethality rates and morbidity, despite clinically proven vaccination strategies and some anti-viral therapies. The eicosanoid Lipoxin A4 (LXA4) promotes the resolution of inflammation by decreasing cell recruitment and pro-inflammatory cytokines release, but also for inducing activation of apoptosis, efferocytosis, and macrophage reprogramming. OBJECTIVE Here, we evaluated whether a synthetic lipoxin mimetic, designated AT-01-KG, would improve the course of influenza A infection in a murine model. METHOD Mice were infected with influenza A/H1N1 and treated with AT-01-KG (1.7 μg/kg/day, i.p.) at day 3 post-infection. RESULTS AT-01-KG attenuated mortality, reducing leukocyte infiltration and lung damage at day 5 and day 7 post-infection. AT-01-KG is a Formyl Peptide Receptor 2 (designated FPR2/3 in mice) agonist, and the protective responses were not observed in fpr2/3 -/- animals. In mice treated with LXA4 (50 μg/kg/day, i.p., days 3-6 post-infection), at day 7, macrophage reprogramming was observed, as seen by a decrease in classically activated macrophages and an increase in alternatively activated macrophages in the lungs. Furthermore, the number of apoptotic cells and cells undergoing efferocytosis was increased in the lavage of treated mice. Treatment also modulated the adaptive immune response, increasing the number of T helper 2 cells (Th2) and regulatory T (Tregs) cells in the lungs of the treated mice. CONCLUSION Therefore, treatment with a lipoxin A4 analog was beneficial in a model of influenza A infection in mice. The drug decreased inflammation and promoted resolution and beneficial immune responses, suggesting it may be useful in patients with severe influenza.
Collapse
Affiliation(s)
- Flavia Rago
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| | - Eliza Mathias Melo
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil
| | - Leigh M Miller
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Alexis M Duray
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Franciel Batista Felix
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana Priscila Vago
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula de Faria Gonçalves
- Immunology of Viral Diseases, René Rachou Research Center, Oswaldo Cruz Foundation (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | | | - Geovanni D Cassali
- Comparative Pathology Laboratory, Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monica de Gaetano
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Benjamin Owen
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Patrick Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
| |
Collapse
|
37
|
Zheng YB, Lu S, Chu TB, Pang GF, Yang LY, Zhang Q. Investigate the potential impact of Hemagglutinin from the H1N1 strain on severe pneumonia. Gene 2024; 926:148559. [PMID: 38740352 DOI: 10.1016/j.gene.2024.148559] [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: 01/20/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
The most prevalent glycoprotein on the influenza virus envelope is called hemagglutinin (HA), yet little is known about its involvement in the pathophysiology and etiology of severe influenza pneumonia. Here, after stimulating human bronchial epithelial cells (16-HBE) and mice with HA of H1N1 for 12 h, we investigated the proliferation, migration, inflammatory cytokines expression, and apoptosis in 16-HBE and the pathological damage in mouse lung tissue. The expression of inflammatory cytokines plasminogen activator inhibitor 1(PAI-1), urokinase-type (uPA) and tissue-type (tPA) plasminogen activators, and apoptosis were all enhanced by HA, which also prevented the proliferation and migration of bronchial epithelial cells. HA enhanced up-regulated PAI-1, uPA, and tPA protein expression within mouse lung tissue and caused lung injury. In conclusion, HA alone, but not the whole H1N1 virus, induces lung tissue injury by inhibiting cell proliferation and migration, while promoting the expression of inflammatory cytokines and apoptosis.
Collapse
Affiliation(s)
- Yu-Bi Zheng
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China.
| | - Song Lu
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Tian-Bao Chu
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Gui-Feng Pang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Lin-Ying Yang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Qing Zhang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China.
| |
Collapse
|
38
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
39
|
Ho TL, Ahn SY, Ko EJ. Adjuvant potential of Peyssonnelia caulifera extract on the efficacy of an influenza vaccine in a murine model. Sci Rep 2024; 14:25353. [PMID: 39455811 PMCID: PMC11512024 DOI: 10.1038/s41598-024-76736-9] [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: 03/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Natural adjuvants have recently garnered interest in the field of vaccinology as their immunostimulatory effects. In this study, we aimed to investigate the potential use of Peyssonnelia caulifera (PC), a marine alga, as a natural adjuvant for an inactivated split A/Puerto Rico/8/1934 H1N1 influenza vaccine (sPR8) in a murine model. We administered PC-adjuvanted vaccines to a murine model via intramuscular prime and boost vaccinations, and subsequently analyzed the induced immunological responses, particularly the production of antigen-specific IgG1 and IgG2a antibodies, memory T and B cell responses, and the protective efficacy against a lethal viral infection. PC extract significantly bolstered the vaccine efficacy, demonstrating balanced Th1/Th2 responses, increased memory T and B cell activities, and improved protection against viral infection. Notably, within 3 days post-vaccination, the PC adjuvant stimulated activation markers on dendritic cells (DCs) and macrophages at the inguinal lymph nodes (ILN), emphasizing its immunostimulatory capabilities. Furthermore, the safety profile of PC was confirmed, showing minimal local inflammation and no significant adverse effects post-vaccination. These findings contribute to our understanding of the immunomodulatory properties of natural adjuvants and suggest the promising roles of natural adjuvants in the development of more effective vaccines for infectious diseases.
Collapse
Affiliation(s)
- Thi Len Ho
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - So Yeon Ahn
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Eun-Ju Ko
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea.
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea.
- Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
40
|
Huang CG, Hsieh MJ, Wu YC, Huang PW, Lin YJ, Tsao KC, Shih SR, Lee LA. Influence of Donor-Specific Characteristics on Cytokine Responses in H3N2 Influenza A Virus Infection: New Insights from an Ex Vivo Model. Int J Mol Sci 2024; 25:10941. [PMID: 39456722 PMCID: PMC11507259 DOI: 10.3390/ijms252010941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Influenza A virus (IAV) is known for causing seasonal epidemics ranging from flu to more severe outcomes like pneumonia, cytokine storms, and acute respiratory distress syndrome. The innate immune response and inflammasome activation play pivotal roles in sensing, preventing, and clearing the infection, as well as in the potential exacerbation of disease progression. This study examines the complex relationships between donor-specific characteristics and cytokine responses during H3N2 IAV infection using an ex vivo model. At 24 h post infection in 31 human lung explant tissue samples, key cytokines such as interleukin (IL)-6, IL-10, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ) were upregulated. Interestingly, a history of lung cancer did not impact the acute immune response. However, cigarette smoking and programmed death-ligand 1 (PD-L1) expression on macrophages significantly increased IL-2 levels. Conversely, age inversely affected IL-4 levels, and diabetes mellitus negatively influenced IL-6 levels. Additionally, both diabetes mellitus and programmed cell death protein 1 (PD-1) expression on CD3+/CD4+ T cells negatively impacted TNF-α levels, while body mass index was inversely associated with IFN-γ production. Toll-like receptor 2 (TLR2) expression emerged as crucial in mediating acute innate and adaptive immune responses. These findings highlight the intricate interplay between individual physiological traits and immune responses during influenza infection, underscoring the importance of tailored and personalized approaches in IAV treatment and prevention.
Collapse
Affiliation(s)
- Chung-Guei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ming-Ju Hsieh
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (M.-J.H.); (Y.-C.W.)
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Cheng Wu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (M.-J.H.); (Y.-C.W.)
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- School of Medicine, College of Life Science and Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Po-Wei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
| | - Ya-Jhu Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
| | - Kuo-Chien Tsao
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shin-Ru Shih
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Li-Ang Lee
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- School of Medicine, College of Life Science and Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan 33305, Taiwan
| |
Collapse
|
41
|
Jugulete G, Olariu MC, Stanescu R, Luminos ML, Pacurar D, Pavelescu C, Merișescu MM. The Clinical Effectiveness and Tolerability of Oseltamivir in Unvaccinated Pediatric Influenza Patients during Two Influenza Seasons after the COVID-19 Pandemic: The Impact of Comorbidities on Hospitalization for Influenza in Children. Viruses 2024; 16:1576. [PMID: 39459910 PMCID: PMC11512198 DOI: 10.3390/v16101576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Antiviral therapy such as oseltamivir has been recommended for hospitalized children with suspected and confirmed influenza for almost 20 years. The therapy is officially authorized for newborns two weeks of age or older, however, questions about its safety and effectiveness still surround it. Our goals were to assess the epidemiological features of two consecutive seasonal influenza cases in children following the COVID-19 pandemic; to observe the clinical effectiveness and tolerability of oseltamivir in hospitalized children who were not vaccinated against influenza and had different influenza subtypes, including A(H1N1), A(H3N2), and B; and to identify specific comorbidities associated with influenza in children. We performed an observational study on 1300 children, enrolled between 1 October 2022 and 30 May 2023 and between 1 October 2023 and 4 May 2024, to the IX Pediatric Infectious Diseases Clinical Section of the National Institute of Infectious Diseases "Prof. Dr. Matei Balș". During the 2022-2023 influenza season, 791 pediatric patients tested positive for influenza and received oseltamivir. Of these, 89% (704/791) had influenza A, with 86.4% having subtype A(H1N1) and 13.6% of cases having A(H3N2), and for influenza B, 11% (87/791) of the pediatric patients. Of the total group, 59% were male, and the median age was 2.4 years (1.02-9.28). For the 2023-2024 influenza season, 509 pediatric patients tested positive for influenza, with 56.9% being of the male gender and who were treated with oseltamivir. Of these patients, 81.6% had influenza A and 18.4% had influenza B. Treatment with neuraminidase inhibitors, specifically oseltamivir, 2 mg/kg/dose administered twice daily for 5 days, was well tolerated by the children, and we recorded no deaths. The duration of hospitalization for patients with a fever after the oseltamivir administration was significantly longer for patients with A(H1N1) infection than A(H3N2), during both seasons. We identified more complications in the 2022-2023 season and a decreasing number of influenza B for the 2023-2024 season. Among children with comorbidities, the most common were asthma, gastrointestinal diseases, and metabolic and endocrine diseases. In terms of effectiveness, oseltamivir significantly reduced the intensity of influenza symptoms, thus reducing the number of days of hospitalization (p = 0.001) as well as post-infection complications (p = 0.005) in both groups. In this study, we evaluated the clinical effectiveness of oseltamivir therapy for all influenza types/subtypes in children, and the length of hospitalization. We identified comorbidities associated with the prolonged duration of hospitalization. Influenza vaccination should be the main tool in the prevention of influenza and its complications in children, especially those with comorbidities.
Collapse
Affiliation(s)
- Gheorghiță Jugulete
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
- “Matei Balş” National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, 2nd District, 021105 Bucharest, Romania
| | - Mihaela Cristina Olariu
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
- “Matei Balş” National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, 2nd District, 021105 Bucharest, Romania
| | - Raluca Stanescu
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
| | - Monica Luminita Luminos
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
- “Matei Balş” National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, 2nd District, 021105 Bucharest, Romania
| | - Daniela Pacurar
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
- Department of Pediatrics, “Grigore Alexandrescu” Emergency Clinical Hospital for Children, No. 30-32, Iancu de Hunedoara Blvd., 011743 Bucharest, Romania
| | - Carmen Pavelescu
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
| | - Mădălina-Maria Merișescu
- Faculty of Medicine, University of Medicine and Pharmacy, “Carol Davila”, No. 37, Dionisie Lupu Street, 2nd District, 020021 Bucharest, Romania; (G.J.); (M.C.O.); (M.L.L.); (D.P.); (C.P.); (M.-M.M.)
- “Matei Balş” National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, 2nd District, 021105 Bucharest, Romania
| |
Collapse
|
42
|
Serna Villa V, Ren X. Lung Progenitor and Stem Cell Transplantation as a Potential Regenerative Therapy for Lung Diseases. Transplantation 2024; 108:e282-e291. [PMID: 38416452 DOI: 10.1097/tp.0000000000004959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Chronic lung diseases are debilitating illnesses ranking among the top causes of death globally. Currently, clinically available therapeutic options capable of curing chronic lung diseases are limited to lung transplantation, which is hindered by donor organ shortage. This highlights the urgent need for alternative strategies to repair damaged lung tissues. Stem cell transplantation has emerged as a promising avenue for regenerative treatment of the lung, which involves delivery of healthy lung epithelial progenitor cells that subsequently engraft in the injured tissue and further differentiate to reconstitute the functional respiratory epithelium. These transplanted progenitor cells possess the remarkable ability to self-renew, thereby offering the potential for sustained long-term treatment effects. Notably, the transplantation of basal cells, the airway stem cells, holds the promise for rehabilitating airway injuries resulting from environmental factors or genetic conditions such as cystic fibrosis. Similarly, for diseases affecting the alveoli, alveolar type II cells have garnered interest as a viable alveolar stem cell source for restoring the lung parenchyma from genetic or environmentally induced dysfunctions. Expanding upon these advancements, the use of induced pluripotent stem cells to derive lung progenitor cells for transplantation offers advantages such as scalability and patient specificity. In this review, we comprehensively explore the progress made in lung stem cell transplantation, providing insights into the current state of the field and its future prospects.
Collapse
Affiliation(s)
- Vanessa Serna Villa
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | | |
Collapse
|
43
|
Chakraborty S, Chauhan A. Fighting the flu: a brief review on anti-influenza agents. Biotechnol Genet Eng Rev 2024; 40:858-909. [PMID: 36946567 DOI: 10.1080/02648725.2023.2191081] [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: 10/26/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The influenza virus causes one of the most prevalent and lethal infectious viral diseases of the respiratory system; the disease progression varies from acute self-limiting mild fever to disease chronicity and death. Although both the preventive and treatment measures have been vital in protecting humans against seasonal epidemics or sporadic pandemics, there are several challenges to curb the influenza virus such as limited or poor cross-protection against circulating virus strains, moderate protection in immune-compromised patients, and rapid emergence of resistance. Currently, there are four US-FDA-approved anti-influenza drugs to treat flu infection, viz. Rapivab, Relenza, Tamiflu, and Xofluza. These drugs are classified based on their mode of action against the viral replication cycle with the first three being Neuraminidase inhibitors, and the fourth one targeting the viral polymerase. The emergence of the drug-resistant strains of influenza, however, underscores the need for continuous innovation towards development and discovery of new anti-influenza agents with enhanced antiviral effects, greater safety, and improved tolerability. Here in this review, we highlighted commercially available antiviral agents besides those that are at different stages of development including under clinical trials, with a brief account of their antiviral mechanisms.
Collapse
Affiliation(s)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala, India
| |
Collapse
|
44
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
45
|
Hall S, Faridi S, Trivedi P, Castonguay M, Kelly M, Zhou J, Lehmann C. Cannabidiol Reduces Systemic Immune Activation in Experimental Acute Lung Injury. Cannabis Cannabinoid Res 2024; 9:1301-1311. [PMID: 37815809 DOI: 10.1089/can.2023.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Background: The underlying pathomechanism of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is the immune response to inflammation or infection within the pulmonary microcirculation. Systemic spread of pathogens, activated immune cells, and inflammatory mediators contributes significantly to mortality in patients with ARDS. Objective: The endogenous cannabinoid system is a major modulator of the immune response during inflammation and infection. Phytocannabinoids, such as cannabidiol (CBD), have shown promising anti-inflammatory effects in several pathologies. The overall objective of this study was to evaluate the effects of CBD on local and systemic inflammation in endotoxin-induced ALI in mice. Materials and Methods: ALI was induced by pulmonary endotoxin challenge. Four groups of male C57BL/6 mice were randomized in this study: control, ALI, ALI with CBD treatment, and control with CBD treatment. Analyses of local and systemic cytokine levels, lung histology, and leukocyte activation as visualized by intravital microscopy of the intestinal and pulmonary microcirculation were performed 6 h following intranasal endotoxin administration. Results: Pulmonary endotoxin challenge induced significant inflammation evidenced by local and systemic cytokine and chemokine release, lung histopathology, and leukocyte adhesion. Intraperitoneal CBD treatment resulted in a significant decrease in systemic inflammation as shown by reduced leukocyte adhesion in the intestinal microcirculation and reduced plasma cytokine and chemokine levels. Pulmonary chemokine levels were decreased, while pulmonary cytokine levels were unchanged. Surprisingly, the ALI score was slightly increased by CBD treatment in a manner driven by enhanced neutrophil infiltration of the alveoli. Conclusion: In this model of experimental ALI, CBD administration was associated with reduced systemic inflammation and heterogeneous effects on pulmonary inflammation. Future studies should explore the mechanisms involved as they relate to neutrophil infiltration and proinflammatory mediator production within the lungs.
Collapse
Affiliation(s)
- Stefan Hall
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sufyan Faridi
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Purvi Trivedi
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mathieu Castonguay
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Melanie Kelly
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Ophthalmology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Juan Zhou
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Christian Lehmann
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Anesthesia, Pain Management, and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
46
|
Abu Aid O, Rohana H, Azrad M, Peretz A. Evaluation of vaccine perceptions in Israel's Elderly: A Comparative study of COVID-19 and influenza vaccination attitudes. Vaccine X 2024; 20:100569. [PMID: 39886533 PMCID: PMC11780388 DOI: 10.1016/j.jvacx.2024.100569] [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: 02/25/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 02/01/2025] Open
Abstract
Purpose This study aimed to evaluate the attitudes of Israeli elderly population towards COVID-19 and influenza vaccines, and to assess factors contributing to these attitudes. Methods Four-hundred and one participants exhibiting symptoms consistent with COVID-19 or influenza were enrolled and filled out a questionnaire. A second questionnaire was filled out for hospitalized patients at discharge. Nasopharyngeal samples were collected and detected for COVID-19 and influenza presence by reverse transcription PCR. Participants were divided into 3 groups according to their attitude towards vaccine- Pro-vaccine, Anti-vaccine and Dependent group, which represented participants whose stance depended on disease infection rate. Results Out of 401 participants, 11.2% (45/401) tested positive for COVID-19, 10.5% (42/401) were positive for Influenza A and one (0.2%) patient had Influenza B. The participants expressed varied beliefs about COVID-19 vaccine: 14.7% (59/401) agreed that it causes disease, 25.4% (102/401) doubted vaccine effectiveness and 22.9% (92/401) questioned vaccine safety. A higher percentage of individuals in Pro-Vaccine group (66.3%, 179/270) as compared to Anti-Vaccine (45.3%, 24/53) and to Dependent (60.3%, 47/78) groups had a COVID-19 history. Hospitalization history was significantly more common in Pro-Vaccine (11.1%, 30/270) and Dependent groups (16.7%, 13/78) than in Anti-Vaccine group (1.9%, 1/53).Influenza vaccine effectiveness was doubted by 19.7% (79/401), 18% (72/401) participants questioned safety, and 18.7% (75/401) agreed that the vaccine causes disease. The majority of both Dependent (54.2%, 13/24) and Pro-Vaccine (56.2%, 167/297) groups believed they received sufficient information about the vaccine, while only 25% (20/80) of the Anti-Vaccine group has similar impressions. Conclusions This analysis reveals a notable disinclination towards vaccination among some of the elderly, reflecting their deep and ingrained hesitancy. These findings emphasize the need for customized approaches to improve vaccine acceptance in this vulnerable group. Such strategies should consider the various motivations and influences shaping elderly perspectives, from individual health experiences to wider social and cultural factors.
Collapse
Affiliation(s)
- Odai Abu Aid
- The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hanan Rohana
- Clinical Microbiology Laboratory, Tzafon Medical Center, Poriya, Israel, affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Maya Azrad
- Clinical Microbiology Laboratory, Tzafon Medical Center, Poriya, Israel, affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Avi Peretz
- Clinical Microbiology Laboratory, Tzafon Medical Center, Poriya, Israel, affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| |
Collapse
|
47
|
Ranjan AD, Bhowmick S, Gupta A, Mallick AI, Banerjee A. Biologically Active Micropatterns of Biomolecules and Living Matter Using Microbubble Lithography. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401127. [PMID: 38884187 DOI: 10.1002/smll.202401127] [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: 02/12/2024] [Revised: 05/17/2024] [Indexed: 06/18/2024]
Abstract
In situ patterning of biomolecules and living organisms while retaining their biological activity is extremely challenging, primarily because such patterning typically involves thermal stresses that could be substantially higher than the physiological thermal or stress tolerance level. Top-down patterning approaches are especially prone to these issues, while bottom-up approaches suffer from a lack of control in developing defined structures and the time required for patterning. A microbubble generated and manipulated by optical tweezers (microbubble lithography) is used to self-assemble and pattern living organisms in continuous microscopic structures in real-time, where the material thus patterned remains biologically active due to their ability to withstand elevated temperatures for short exposures. Successful patterns of microorganisms (Escherichia coli, Lactococcus. lactis and the Type A influenza virus) are demonstrated, as well as reporter proteins such as green fluorescent protein (GFP) on functionalized substrates with high signal-to-noise ratio and selectivity. Together, the data presented herein may open up fascinating possibilities in rapid in situ parallelized diagnostics of multiple pathogens and bioelectronics.
Collapse
Affiliation(s)
- Anand Dev Ranjan
- Department of Physical Sciences, IISER Kolkata, Mohanpur, West Bengal, 741246, India
| | - Sucharita Bhowmick
- Department of Biological Sciences, IISER Kolkata, Mohanpur, West Bengal, 741246, India
| | - Arnab Gupta
- Department of Biological Sciences, IISER Kolkata, Mohanpur, West Bengal, 741246, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, IISER Kolkata, Mohanpur, West Bengal, 741246, India
| | - Ayan Banerjee
- Department of Physical Sciences, IISER Kolkata, Mohanpur, West Bengal, 741246, India
| |
Collapse
|
48
|
Engin MMN, Özdemir Ö. Role of vitamin D in COVID-19 and other viral infections. World J Virol 2024; 13:95349. [PMID: 39323448 PMCID: PMC11401007 DOI: 10.5501/wjv.v13.i3.95349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
Vitamin D is a steroid hormone that is naturally produced in the body or obtained through dietary sources, primarily under the influence of UVB radiation. This essential nutrient has a vital role in numerous physiological processes, encompassing immune function, cell growth, differentiation, insulin regulation, and cardiovascular well-being, along with its pivotal role in sustaining the delicate equilibrium of calcium and phosphate concentrations in the body. Moreover, vitamin D reinforces mucosal defense and bolsters the immune system through immunomodulation, making it a critical component of overall health. Numerous studies have unveiled the profound connection between vitamin D and the predisposition to respiratory tract infections, including well-known viruses such as influenza and the novel severe acute respiratory syndrome coronavirus 2. Vitamin D deficiency has been consistently linked to increased severity of coronavirus disease 2019 (COVID-19) and a heightened risk of mortality among afflicted individuals. Retrospective observational studies have further substantiated these findings, indicating that levels of vitamin D are linked with both the occurrence and severity of COVID-19 cases. Vitamin D has its influence on viral infections through a multitude of mechanisms, such as promoting the release of antimicrobial peptides and fine-tuning the responses of the immune system. Additionally, vitamin D is intertwined with the intricate network of the renin-angiotensin system, suggesting a potential impact on the development of complications related to COVID-19. While further clinical trials and extensive research are warranted, the existing body of evidence strongly hints at the possible use of vitamin D as a valuable tool in the prophylaxis and management of COVID-19 and other viral infectious diseases.
Collapse
Affiliation(s)
| | - Öner Özdemir
- Division of Allergy and Immunology, Department of Pediatrics, Sakarya Research and Training Hospital, Sakarya University, Faculty of Medicine, Sakarya 54100, Türkiye
| |
Collapse
|
49
|
Galindo-Fraga A, del Carmen Guerra-de-Blas P, Ortiz-Hernández AA, Rubenstein K, Ortega-Villa AM, Ramírez-Venegas A, Valdez-Vázquez R, Moreno-Espinosa S, Llamosas-Gallardo B, Pérez-Patrigeon S, Noyola DE, Magaña-Aquino M, Vilardell-Dávila A, Guerrero ML, Powers JH, Beigel J, Ruiz-Palacios GM. Prospective cohort study of patient demographics, viral agents, seasonality, and outcomes of influenza-like illness in Mexico in the late H1N1-pandemic and post-pandemic years (2010-2014). IJID REGIONS 2024; 12:100394. [PMID: 39045384 PMCID: PMC11265585 DOI: 10.1016/j.ijregi.2024.100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024]
Abstract
Objectives Influenza-like illness (ILI) caused by respiratory viruses results in various respiratory clinical manifestations. The ILI002 prospective observational cohort study aimed to describe viral agents, seasonality, and outcomes of patients with ILI during four seasons in the influenza H1N1-pandemic and post-pandemic years (2010-2014). Methods Patients from six Mexican hospitals were enrolled from April 2010 to March 2014. Clinical data and nasopharyngeal swabs were obtained and tested for viral respiratory pathogens by real-time reverse-transcription polymerase chain reaction. Results Of the 5662 enrolled participants, 64.9% were adults and 35.1% were children. Among the 5629 participants with single-pathogen detection, rhinovirus (20.2%), influenza virus (11.2%), respiratory syncytial virus (RSV) (7.2%), and coronavirus (6.8%) were the most frequent pathogens. Co-infection occurred in 14.5% of cases; 49.3% of participants required hospitalization, particularly in RSV cases (42.9% adults, 89.6% children). The mortality rate was 2.8% higher among older adult participants and those with comorbidities. Influenza H1N1 had the highest mortality rate, yet almost half of the deceased had no pathogen. Rhinovirus persisted year-round, while influenza, coronavirus, and RSV peaked during cooler months. Conclusions Analyses showed that some viruses causing ILI may lead to severe disease and hospitalization irrespective of comorbidities. These findings may help in decision-making about public health policies on prevention measures, vaccination, treatment, and administration of health care.
Collapse
Affiliation(s)
- Arturo Galindo-Fraga
- Subdirección de Epidemiología Hospitalaria, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Ana A. Ortiz-Hernández
- División de Desarrollo y Enlace Interinstitucional, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Kevin Rubenstein
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Alejandra Ramírez-Venegas
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Rafael Valdez-Vázquez
- Departamento de Infectología, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
| | | | - Beatriz Llamosas-Gallardo
- División de Desarrollo y Enlace Interinstitucional, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Santiago Pérez-Patrigeon
- Division of Infectious Diseases, Queen's University, Kingston, Ontario, Canada
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel E. Noyola
- Departamento de Microbiología y Centro de Investigación en Ciencias de la Salud y Biomedicina, Facultad de Medicina, Universidad Autónoma de San Luís Potosí, San Luis Potosí, Mexico
| | | | - Ana Vilardell-Dávila
- The Mexican Emerging Infectious Diseases Clinical Research Network (LaRed), Mexico City, Mexico
| | - M. Lourdes Guerrero
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - John H. Powers
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John Beigel
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Guillermo M. Ruiz-Palacios
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Mexican Emerging Infectious Disease Clinical Research Network
- Subdirección de Epidemiología Hospitalaria, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- The Mexican Emerging Infectious Diseases Clinical Research Network (LaRed), Mexico City, Mexico
- División de Desarrollo y Enlace Interinstitucional, Instituto Nacional de Pediatría, Mexico City, Mexico
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Departamento de Infectología, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
- Departamento de Infectología, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Division of Infectious Diseases, Queen's University, Kingston, Ontario, Canada
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Microbiología y Centro de Investigación en Ciencias de la Salud y Biomedicina, Facultad de Medicina, Universidad Autónoma de San Luís Potosí, San Luis Potosí, Mexico
- Hospital Regional Dr. Ignacio Morones Prieto, San Luis Potosí, Mexico
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| |
Collapse
|
50
|
Haddara A, Houry Z, Zahreddine N, Atallah M, Boutros CF, Tannous J, Sadaka C, Wehbe S, Kadi T, Ibrahim A, Ahmadieh R, Kardas T, Soudani N, Kalamouni HA, Zgheib Y, Yaman NE, Khafaja S, Lteif M, Shaker R, Casals AA, Youssef Y, Youssef N, Zein ZE, Chamseddine S, Chmaisse A, Haj M, Fayad D, Kiblawi S, Isaac I, Anan H, Radwan N, Wakim RH, Zaraket H, Kanj SS, Dbaibo GS. Characteristics of medically attended influenza infection across age groups before the COVID-19 pandemic in Lebanon. J Infect Public Health 2024; 17:102521. [PMID: 39173555 DOI: 10.1016/j.jiph.2024.102521] [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: 04/06/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Influenza represents a significant global health burden for individuals and society. This study assessed the burden of medically attended influenza at a tertiary medical center in Lebanon to describe the demographics, risk factors, and outcomes prior to the COVID-19 pandemic. METHODS This was a retrospective review of patients who tested positive for the influenza virus during three seasons between July 1, 2016 to June 30, 2019, at the American University of Beirut Medical Center. RESULTS A total of 2049 patients who tested positive for influenza were analyzed. Influenza A accounted for 79.6 % of cases, and influenza B for 19.7 %, with influenza activity starting in October/November and peaking in December/January. Older age above 65 years (AOR=3.584), obesity (AOR=2.183), and chronic conditions such as chronic lung diseases (AOR=1.832), and bacterial co-infection (AOR= 2.834) were found to be independent risk factors for developing complications. Viral co-infection increased the likelihood of death tenfold. Vaccinated patients had a shorter mean hospital stay duration and a lower intensive care unit admission rate. CONCLUSION The burden of medically attended influenza at our tertiary medical center in Lebanon prior to the COVID-19 pandemic was high. Vaccination decreased the likelihood of complications leading to intensive care unit admission in patients at risk.
Collapse
Affiliation(s)
- Amani Haddara
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Zeina Houry
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Nada Zahreddine
- Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Mark Atallah
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Celina F Boutros
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Joseph Tannous
- Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Infection Control Department, Emirates health services, Dubai Silicon Oasis, Dubai, United Arab Emirates
| | - Christian Sadaka
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Sarah Wehbe
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Tamara Kadi
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Ahmad Ibrahim
- Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Rihab Ahmadieh
- Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Tala Kardas
- Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Infection Control Department, Danat Al Emarat Hospital for Women & Children, Rabdan, Abu Dhabi, United Arab Emirates
| | - Nadia Soudani
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Habib Al Kalamouni
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Youssef Zgheib
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Noha El Yaman
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Sarah Khafaja
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Mireille Lteif
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Rouba Shaker
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Aia Assaf Casals
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Yolla Youssef
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Nour Youssef
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Zeinab El Zein
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Sarah Chamseddine
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Ahmad Chmaisse
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Magda Haj
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Danielle Fayad
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Shereen Kiblawi
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Imad Isaac
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon
| | - Hind Anan
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Najwa Radwan
- Faculty of Medicine, American University of Beirut, Hamra, Beirut, Lebanon
| | - Rima Hanna Wakim
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon
| | - Hassan Zaraket
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.
| | - Souha S Kanj
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Infection Control and Prevention Program, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Infectious Diseases Division, Department of Internal Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon.
| | - Ghassan S Dbaibo
- Center for Infectious Diseases Research, American University of Beirut, Hamra, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Hamra, Beirut, Lebanon.
| |
Collapse
|