1
|
Çalışkan DM, Kumar S, Hinse S, Schughart K, Wiewrodt R, Fischer S, Krueger V, Wiebe K, Barth P, Mellmann A, Ludwig S, Brunotte L. Molecular characterisation of influenza B virus from the 2017/18 season in primary models of the human lung reveals improved adaptation to the lower respiratory tract. Emerg Microbes Infect 2024; 13:2402868. [PMID: 39248230 PMCID: PMC11421153 DOI: 10.1080/22221751.2024.2402868] [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: 05/24/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
The 2017/18 influenza season was characterized by unusual high numbers of severe infections and hospitalizations. Instead of influenza A viruses, this season was dominated by infections with influenza B viruses of the Yamagata lineage. While this IBV/Yam dominance was associated with a vaccine mismatch, a contribution of virus intrinsic features to the clinical severity of the infections was speculated. Here, we performed a molecular and phenotypic characterization of three IBV isolates from patients with severe flu symptoms in 2018 and compared it to an IBV/Yam isolate from 2016 using experimental models of increasing complexity, including human lung explants, lung organoids, and alveolar macrophages. Viral genome sequencing revealed the presence of clade but also isolate specific mutations in all viral genes, except NP, M1, and NEP. Comparative replication kinetics in different cell lines provided further evidence for improved replication fitness, tolerance towards higher temperatures, and the development of immune evasion mechanisms by the 2018 IBV isolates. Most importantly, immunohistochemistry of infected human lung explants revealed an impressively altered cell tropism, extending from AT2 to AT1 cells and macrophages. Finally, transcriptomics of infected human lung explants demonstrated significantly reduced amounts of type I and type III IFNs by the 2018 IBV isolate, supporting the existence of additional immune evasion mechanisms. Our results show that the severeness of the 2017/18 Flu season was not only the result of a vaccine mismatch but was also facilitated by improved adaptation of the circulating IBV strains to the environment of the human lower respiratory tract.
Collapse
Affiliation(s)
- Duygu Merve Çalışkan
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Sriram Kumar
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Saskia Hinse
- Institute of Virology, University of Münster, Münster, Germany
| | - Klaus Schughart
- Institute of Virology, University of Münster, Münster, Germany
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rainer Wiewrodt
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Stefan Fischer
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Vera Krueger
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Karsten Wiebe
- Department of Thoracic Surgery, University Hospital Münster, Muenster, Germany
| | - Peter Barth
- Gerhard-Domagk-Institute of Pathology, University of Münster, Muenster, Germany
| | | | - Stephan Ludwig
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Linda Brunotte
- Institute of Virology, University of Münster, Münster, Germany
| |
Collapse
|
2
|
Almeida PRJ, Periard AM, Tana FL, Avila RE, Milhorato LB, Alcantara KMM, Resende CB, Serufo AV, Santos FR, Teixeira DC, Queiroz-Junior CM, Fonseca TCM, Silva BLV, Costa VV, Souza RP, Perretti M, Jonassen TEN, Teixeira MM. Effects of a pro-resolving drug in COVID-19: preclinical studies to a randomized, placebo-controlled, phase Ib/IIa trial in hospitalized patients. Br J Pharmacol 2024; 181:4750-4765. [PMID: 39159951 DOI: 10.1111/bph.17322] [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: 01/17/2024] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Pro-resolving molecules may curb disease caused by viruses without altering the capacity of the host to deal with infection. AP1189 is a melanocortin receptor-biased agonist endowed with pro-resolving and anti-inflammatory activity. We evaluated the preclinical and early clinical effects of treatment with AP1189 in the context of COVID-19. METHODS C57BL/6j mice were infected intranasally with MHV-A59 or hK18-ACE2 mice with SARS-CoV-2. AP1189 (10 mg·kg-1, BID, s.c.) was given to the animals from day 2 and parameters evaluated at day 5. Human PBMCs from health donors were infected with SARS-CoV-2 in presence or absence of AP1189 and production of cytokines quantified. In the clinical study, 6 patients were initially given AP1189 (100 mg daily for 14 days) and this was followed by a randomized (2:1), placebo-controlled, double-blind trial that enrolled 54 hospitalized COVID-19 patients needing oxygen support. The primary outcome was the time in days until respiratory recovery, defined as a SpO2 ≥ 93% in ambient air. RESULTS Treatment with AP1189 attenuated pulmonary inflammation in mice infected with MHV-A59 or SARS-CoV-2 and decreased the release of CXCL10, TNF-α and IL-1β by human PBMCs. Hospitalized COVID-19 patients already taking glucocorticoids took a median time of 6 days until respiratory recovery when given placebo versus 4 days when taking AP1189 (P = 0.017). CONCLUSION Treatment with AP1189 was associated with less disease caused by beta-coronavirus infection both in mice and in humans. This is the first demonstration of the effects of a pro-resolving molecule in the context of severe infection in humans.
Collapse
Affiliation(s)
- Pedro R J Almeida
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre M Periard
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda L Tana
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Larissa B Milhorato
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Katlen M M Alcantara
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carolina B Resende
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Angela V Serufo
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe R Santos
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle C Teixeira
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Talita C M Fonseca
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Barbara L V Silva
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renan P Souza
- Genetics Department, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Thomas E N Jonassen
- Synact Pharma Aps, Holte, Denmark and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mauro M Teixeira
- Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
3
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [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: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
4
|
Farnesi-de-Assunção TS, Oliveira-Scussel ACDM, Rodrigues WF, Matos BS, da Silva DAA, de Andrade E Silva LE, Mundim FV, Helmo FR, Bernardes E Borges AV, Desidério CS, Trevisan RO, Obata MMS, Barbosa LM, Lemes MR, Costa-Madeira JC, Barbosa RM, Cunha ACCH, Pereira LQ, Tanaka SCSV, de Vito FB, Monteiro IB, Ferreira YM, Machado GH, Moraes-Souza H, Rodrigues DBR, de Oliveira CJF, da Silva MV, Júnior VR. COVID-19 Inflammatory Syndrome: Lessons from TNFRI and CRP about the Risk of Death in Severe Disease. Biomedicines 2024; 12:2138. [PMID: 39335653 PMCID: PMC11428742 DOI: 10.3390/biomedicines12092138] [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: 08/02/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Cytokine storm in severe COVID-19 is responsible for irreversible tissue damage and death. Soluble mediators from the TNF superfamily, their correlation with clinical outcome, and the use of TNF receptors as a potent predictor for clinical outcome were evaluated. Methods: Severe COVID-19 patients had the levels of soluble mediators from the TNF superfamily quantified and categorized according to the clinical outcome (death versus survival). Statistical modeling was performed to predict clinical outcomes. Results: COVID-19 patients have elevated serum levels from the TNF superfamily. Regardless of sex and age, the sTNFRI levels were observed to be significantly higher in deceased patients from the first weeks following the onset of symptoms. We analyzed hematological parameters and inflammatory markers, and there was a difference between the groups for the following factors: erythrocytes, hemoglobin, hematocrit, leukocytes, neutrophils, band cells, lymphocytes, monocytes, CRP, IL-8, IFN-γ, IL-10, IL-6, IL-4, IL-2, leptin MIF sCD40L, and sTNFRI (p < 0.05). A post hoc analysis showed an inferential capacity over 70% for some hematological markers, CRP, and inflammatory mediators in deceased patients. sTNFRI was strongly associated with death, and the sTNFRI/sTNFRII ratio differed between outcomes (p < 0.001; power above 90%), highlighting the impact of these proteins on clinical results. The final logistic model, including sTNFRI/sTNFRII and CRP, indicated high sensitivity, specificity, accuracy, and an eight-fold higher odds ratio for an unfavorable outcome. Conclusions: The joint use of the sTNFRI/sTNFRII ratio with CRP proves to be a promising tool to assist in the clinical management of patients hospitalized for COVID-19.
Collapse
Affiliation(s)
| | | | - Wellington Francisco Rodrigues
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Beatriz Sodré Matos
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Djalma Alexandre Alves da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Leonardo Eurípedes de Andrade E Silva
- Clinical Analysis and Pathological Anatomy Laboratory, Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Fabiano Vilela Mundim
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Fernanda Rodrigues Helmo
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | - Chamberttan Souza Desidério
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Rafael Obata Trevisan
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Malu Mateus Santos Obata
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Laís Milagres Barbosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Marcela Rezende Lemes
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Juliana Cristina Costa-Madeira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Rafaela Miranda Barbosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | - Loren Queli Pereira
- Hematological Research Laboratory, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | | | - Ivan Borges Monteiro
- UNIMED São Domingos Hospital, Uberaba 38025-110, MG, Brazil
- Alencar Gomes da Silva Regional Hospital, Uberaba 38060-200, MG, Brazil
| | | | | | - Hélio Moraes-Souza
- Hematological Research Laboratory, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Denise Bertulucci Rocha Rodrigues
- Centro de Formação Especial em Saúde (CEFORES), Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
- Department of Immunology, Medical School, University of Uberaba, Uberaba 38010-200, MG, Brazil
| | - Carlo José Freire de Oliveira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Marcos Vinicius da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Virmondes Rodrigues Júnior
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| |
Collapse
|
5
|
Li X, Luo X, Wang B, Fu L, Chen X, Lu Y. Clofazimine inhibits innate immunity against Mycobacterium tuberculosis by NF-κB. mSphere 2024; 9:e0025424. [PMID: 39046230 PMCID: PMC11351037 DOI: 10.1128/msphere.00254-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Tuberculosis (TB) remains one of the infectious diseases with high incidence and high mortality. About a quarter of the population has been latently infected with Mycobacterium tuberculosis. At present, the available TB treatment strategies have the disadvantages of too long treatment duration and serious adverse reactions. The sustained inflammatory response leads to permanent tissue damage. Unfortunately, the current selection of treatment regimens does not consider the immunomodulatory effects of various drugs. In this study, we preliminarily evaluated the effects of commonly used anti-tuberculosis drugs on innate immunity at the cellular level. The results showed that clofazimine (CFZ) has a significant innate immunosuppressive effect. CFZ significantly inhibited cytokines and type I interferons (IFNα and IFNβ) expression under both lipopolysaccharide stimulation and CFZ-resistant strain infection. In further mechanistic studies, CFZ strongly inhibited the phosphorylation of nuclear factor kappa B (NF-κB) p65 and had no significant effect on the phosphorylation of p38. In conclusion, our study found that CFZ suppresses innate immunity against Mycobacterium tuberculosis by NF-κB, which should be considered in future regimen development. IMPORTANCE The complete elimination of Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, from TB patients is a complicated process that takes a long time. The excessive immune inflammatory response of the host for a long time causes irreversible organic damage to the lungs and liver. Current antibiotic-based treatment options involve multiple complex drug combinations, often targeting different physiological processes of Mtb. Given the high incidence of post-tuberculosis lung disease, we should also consider the immunomodulatory properties of other drugs when selecting drug combinations.
Collapse
Affiliation(s)
- Xinda Li
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoyi Luo
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bin Wang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Lei Fu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xi Chen
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
6
|
Wen Z, Ablimit A. Aquaporin 1 aggravates lipopolysaccharide-induced macrophage polarization and pyroptosis. Sci Rep 2024; 14:18569. [PMID: 39127771 PMCID: PMC11316789 DOI: 10.1038/s41598-024-68899-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Acute respiratory infections (ARIs) are associated with high mortality and morbidity. Acute lung injury (ALI) is caused by the activation of immune cells during ARIs caused by viruses such as SARS-CoV-2. Aquaporin 1 (AQP1) is distributed in a variety of immune cells and is related to the occurrence of ALI, but the mechanism is not clear. A reference map of human single cells was used to identify macrophages in COVID-19 patients at the single-cell level. "FindMarkers" was used to analyze differentially expressed genes (DEGs), and "clusterProfiler" was used to analyze the functions of the DEGs. An M1 macrophage polarization model was established with lipopolysaccharide (LPS) in vitro, and the relationships among AQP1, pyroptosis and M1 polarization were examined by using an AQP1 inhibitor. Transcriptome sequencing and RT-qPCR were used to examine the molecular mechanism by which AQP1 regulates macrophage polarization and pyroptosis. Antigen presentation, M1 polarization, migration and phagocytosis are abnormal in SARS-CoV-2-infected macrophages, which is related to the high expression of AQP1. An M1 polarization model of macrophages was constructed in vitro, and an AQP1 inhibitor was used to examine whether AQP1 could promote M1 polarization and pyroptosis in response to LPS. Transcriptome and cell experiments showed that this effect was related to a decrease in chemokines caused by AQP1 deficiency. AQP1 participates in M1 polarization and pyroptosis in macrophages by increasing the levels of chemokines induced by LPS, which provides new insights for the diagnosis and treatment of ALI.
Collapse
Affiliation(s)
- Zhuman Wen
- Department of Histology and Embryology, Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Abduxukur Ablimit
- Department of Histology and Embryology, Basic Medical College, Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
7
|
Chen S, Zhu H, Lin L, Lu L, Chen L, Zeng L, Yue W, Kong X, Zhang H. Apelin-13 improves pulmonary epithelial barrier function in a mouse model of LPS-induced acute lung injury by inhibiting Chk1-mediated DNA damage. Biochem Pharmacol 2024; 226:116297. [PMID: 38801925 DOI: 10.1016/j.bcp.2024.116297] [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: 10/22/2023] [Revised: 04/10/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Apelin-13, a type of active peptide, can alleviate lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, the specific mechanism is unclear. Cell cycle checkpoint kinase 1 (Chk1) plays an important role in DNA damage. Here, we investigated the regulatory effect of Apelin on Chk1 in ALI. Chk1-knockout and -overexpression mice were used to explore the role of Chk1 in LPS-induced ALI mice treated with or without Apelin-13. In addition, A549 cells were also treated with LPS to establish a cell model. Chk1 knockdown inhibited the destruction of alveolar structure, the damage of lung epithelial barrier function, and DNA damage in the ALI mouse model. Conversely, Chk1 overexpression had the opposite effect. Furthermore, Apelin-13 reduced Chk1 expression and DNA damage to improve the impaired lung epithelial barrier function in the ALI model. However, the high expression of Chk1 attenuated the protective effect of Apelin-13 on ALI. Notably, Apelin-13 promoted Chk1 degradation through autophagy to regulate DNA damage in LPS-treated A549 cells. In summary, Apelin-13 regulates the expression of Chk1 by promoting autophagy, thereby inhibiting epithelial DNA damage and repairing epithelial barrier function.
Collapse
Affiliation(s)
- Siyue Chen
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China; School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Huihui Zhu
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Lidan Lin
- School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Liling Lu
- Children's Hospital, Zhejiang University School of Medicine, Zhejiang 310000, PR China
| | - Lin Chen
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China; School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Luyao Zeng
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Wei Yue
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China.
| | - Hailin Zhang
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China.
| |
Collapse
|
8
|
Zhai G, Fu W, Yuan S, Sun P, Zhu C, Zhao C, Zhang X, Xu J. A fusion protein approach to integrate antiviral and anti-inflammatory activities for developing new therapeutics against influenza A virus infection. Antiviral Res 2024; 228:105924. [PMID: 38862076 DOI: 10.1016/j.antiviral.2024.105924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Human interferon α2 (IFNα2) is a cytokine with broad-spectrum antiviral activity, and its engineered forms are widely used to treat viral infections. However, IFNα2 may trigger proinflammatory responses and underlying side effects during treatment. Trefoil factor 2 (TFF2) is a secreted protein with anti-inflammatory properties. Here, we explored whether coupling IFNα2 to TFF2 in a two-in-one fusion form could combine the beneficial effects of both molecules on viral infections toward a more desirable treatment outcome. We engineered two forms of human IFNα2 and TFF2 fusion proteins, IFNα2-TFF2-Fc (ITF) and TFF2-IFNα2-Fc (TIF), and examined their properties in vitro in comparison to IFNα2 and TFF2 alone. RNA-Seq was further used to explore such comparison on dynamic gene regulation at transriptomic level. These in vitro assessments collectively indicated that TIF largely retained the antiviral activity of IFNα2 while being a weaker inflammation inducer, consistent with the presence of TFF2 activity. We further demonstrated the superiority of TIF over IFNα2 or TFF2 alone in treating influenza infection using a mouse infection model. Together, our study provided evidence supporting that, by possessing antiviral activity conferred by IFNα2 with complementation from TFF2 in suppressing the inflammatory side effects, the fusion proteins, particularly TIF, represent more effective agents against influenza and other respiratory viral infections than IFNα2 or TFF2 alone. It implies that merging two molecules with complementary functions holds potential for developing novel therapeutics against viral infections.
Collapse
Affiliation(s)
- Guanxing Zhai
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Weihui Fu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Songhua Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Peng Sun
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Cuisong Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Chen Zhao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China; Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China; Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Gogoi M, Clark PA, Ferreira ACF, Rodriguez Rodriguez N, Heycock M, Ko M, Murphy JE, Chen V, Luan SL, Jolin HE, McKenzie ANJ. ILC2-derived LIF licences progress from tissue to systemic immunity. Nature 2024; 632:885-892. [PMID: 39112698 PMCID: PMC11338826 DOI: 10.1038/s41586-024-07746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 06/24/2024] [Indexed: 08/17/2024]
Abstract
Migration and homing of immune cells are critical for immune surveillance. Trafficking is mediated by combinations of adhesion and chemokine receptors that guide immune cells, in response to chemokine signals, to specific locations within tissues and the lymphatic system to support tissue-localized immune reactions and systemic immunity1,2. Here we show that disruption of leukaemia inhibitory factor (LIF) production from group 2 innate lymphoid cells (ILC2s) prevents immune cells leaving the lungs to migrate to the lymph nodes (LNs). In the absence of LIF, viral infection leads to plasmacytoid dendritic cells (pDCs) becoming retained in the lungs where they improve tissue-localized, antiviral immunity, whereas chronic pulmonary allergen challenge leads to marked immune cell accumulation and the formation of tertiary lymphoid structures in the lung. In both cases immune cells fail to migrate to the lymphatics, leading to highly compromised LN reactions. Mechanistically, ILC2-derived LIF induces the production of the chemokine CCL21 from lymphatic endothelial cells lining the pulmonary lymphatic vessels, thus licensing the homing of CCR7+ immune cells (including dendritic cells) to LNs. Consequently, ILC2-derived LIF dictates the egress of immune cells from the lungs to regulate tissue-localized versus systemic immunity and the balance between allergen and viral responsiveness in the lungs.
Collapse
Affiliation(s)
- Mayuri Gogoi
- MRC Laboratory of Molecular Biology, Cambridge, UK.
| | | | | | | | | | - Michelle Ko
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | - Victor Chen
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Shi-Lu Luan
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | |
Collapse
|
10
|
Deng L, Ouyang B, Tang W, Wang N, Yang F, Shi H, Zhang Z, Yu H, Chen M, Wei Y, Dong J. Icariside II modulates pulmonary fibrosis via PI3K/Akt/β-catenin pathway inhibition of M2 macrophage program. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155687. [PMID: 38759312 DOI: 10.1016/j.phymed.2024.155687] [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: 12/04/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a debilitating interstitial lung disorder characterized by its limited therapeutic interventions. Macrophages, particularly the alternatively activated macrophages (M2 subtype), have been acknowledged for their substantial involvement in the development of pulmonary fibrosis. Hence, targeting macrophages emerges as a plausible therapeutic avenue for IPF. Icariside II (ISE II) is a natural flavonoid glycoside molecule known for its excellent anti-tumor and anti-fibrotic activities. Nevertheless, the impact of ISE II on pulmonary fibrosis and the intricate mechanisms through which it operates have yet to be elucidated. OBJECTIVE To scrutinize the impact of ISE II on the regulation of M2 macrophage polarization and its inhibitory effect on pulmonary fibrosis, as well as to delve deeper into the underlying mechanisms of its actions. METHODS The effect of ISE II on proliferation and apoptosis in RAW264.7 cells was assessed through the use of EdU-488 labeling and the Annexin V/PI assay. Flow cytometry, western blot, and qPCR were employed to detect markers associated with the M2 polarization phenotype. The anti-fibrotic effects of ISE II in NIH-3T3 cells were investigated in a co-culture with M2 macrophages. Si-Ctnnb1 and pcDNA3.1(+)-Ctnnb1 plasmid were used to investigate the mechanism of targeted intervention. The murine model of pulmonary fibrosis was induced by intratracheal administration of bleomycin (BLM). Pulmonary function, histopathological manifestations, lung M2 macrophage infiltration, and markers associated with pulmonary fibrosis were evaluated. Furthermore, in vivo transcriptomics analysis was employed to elucidate differentially regulated genes in lung tissues. Immunofluorescence, western blot, and immunohistochemistry were conducted for corresponding validation. RESULTS Our investigation demonstrated that ISE II effectively inhibited the proliferation of RAW264.7 cells and mitigated the pro-fibrotic characteristics of M2 macrophages, exemplified by the downregulation of CD206, Arg-1, and YM-1, Fizz1, through the inhibition of the PI3K/Akt/β-catenin signaling pathway. This impact led to the amelioration of myofibroblast activation and the suppression of nuclear translocation of β-catenin of NIH-3T3 cells in a co-culture. Consequently, it resulted in decreased collagen deposition, reduced infiltration of profibrotic macrophages, and a concurrent restoration of pulmonary function in mice IPF models. Furthermore, our RNA sequencing results showed that ISE II could suppress the expression of genes related to M2 polarization, primarily by inhibiting the PI3K/Akt and β-catenin signaling pathway. In essence, our findings suggest that ISE II holds potential as an anti-fibrotic agent by orchestrating macrophage polarization. This may have significant implications in clinical practice. CONCLUSION This study has provided evidence that ISE II exerts a significant anti-fibrotic effect by inhibiting macrophage M2 polarization through the suppression of the PI3K/Akt/β-catenin signaling pathway. These findings underscore the potential of ISE II as a promising candidate for the development of anti-fibrotic pharmaceuticals in the future.
Collapse
Affiliation(s)
- Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Boshu Ouyang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Fangyong Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenhua Zhang
- Shanghai Fifth People's Hospital, Fudan University, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Mengmeng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Papaneophytou C. Breaking the Chain: Protease Inhibitors as Game Changers in Respiratory Viruses Management. Int J Mol Sci 2024; 25:8105. [PMID: 39125676 PMCID: PMC11311956 DOI: 10.3390/ijms25158105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Respiratory viral infections (VRTIs) rank among the leading causes of global morbidity and mortality, affecting millions of individuals each year across all age groups. These infections are caused by various pathogens, including rhinoviruses (RVs), adenoviruses (AdVs), and coronaviruses (CoVs), which are particularly prevalent during colder seasons. Although many VRTIs are self-limiting, their frequent recurrence and potential for severe health complications highlight the critical need for effective therapeutic strategies. Viral proteases are crucial for the maturation and replication of viruses, making them promising therapeutic targets. This review explores the pivotal role of viral proteases in the lifecycle of respiratory viruses and the development of protease inhibitors as a strategic response to these infections. Recent advances in antiviral therapy have highlighted the effectiveness of protease inhibitors in curtailing the spread and severity of viral diseases, especially during the ongoing COVID-19 pandemic. It also assesses the current efforts aimed at identifying and developing inhibitors targeting key proteases from major respiratory viruses, including human RVs, AdVs, and (severe acute respiratory syndrome coronavirus-2) SARS-CoV-2. Despite the recent identification of SARS-CoV-2, within the last five years, the scientific community has devoted considerable time and resources to investigate existing drugs and develop new inhibitors targeting the virus's main protease. However, research efforts in identifying inhibitors of the proteases of RVs and AdVs are limited. Therefore, herein, it is proposed to utilize this knowledge to develop new inhibitors for the proteases of other viruses affecting the respiratory tract or to develop dual inhibitors. Finally, by detailing the mechanisms of action and therapeutic potentials of these inhibitors, this review aims to demonstrate their significant role in transforming the management of respiratory viral diseases and to offer insights into future research directions.
Collapse
Affiliation(s)
- Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| |
Collapse
|
12
|
王 洪, 谢 海, 徐 乌, 李 明. [Urolithin A alleviates respiratory syncytial virus-induced lung infection in neonatal mice by activating miR-136-mediated Sirt1 signaling]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1370-1381. [PMID: 39051083 PMCID: PMC11270657 DOI: 10.12122/j.issn.1673-4254.2024.07.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE To observe the therapeutic effects of urolithin A (UA) on respiratory syncytial virus (RSV)-induced lung infection in neonatal mice and explore the underlying mechanisms. METHODS Babl/c mice (5-7 days old) were subjected to nasal instillation of RSV and received intraperitoneal injection of saline or 2.5, 5 and 10 mg/kg UA 2 h after the infection and then once daily for 2 weeks. Bronchoalveolar lavage fluid (BALF) was then collected for detection of inflammatory cells and mediators, and lung pathology was evaluated with HE staining. RSV-infected BEAS-2B cells were treated with 2.5, 5 or 10 µmol/ L UA. Inflammatory factors, cell viability, apoptosis and autophagy were analyzed using ELISA, CCK-8 assay, TUNEL staining, flow cytometry, Western blotting and immunofluorescence staining. The cellular expressions of miR-136 and Sirt1 mRNAs were detected using qRT-PCR. A dual-luciferase reporter system was used to verify the binding between miR-136 and Sirt1. RESULTS In neonatal Babl/c mice, RSV infection caused obvious lung pathologies, promoted pulmonary cell apoptosis and LC3-Ⅱ/Ⅰ, Beclin-1 and miR-136 expressions, and increased the total cell number, inflammatory cells and factors in the BALF and decreased p62 and Sirt1 expressions. All these changes were alleviated dose-dependently by UA. In BEAS-2B cells, RSV infection significantly increased cell apoptosis, LC3B-positive cells and miR-136 expression and reduced Sirt1 expression (P<0.01), which were dose-dependently attenuated by UA. Dual-luciferase reporter assay confirmed the binding between miR-136 and Sirt1. In RSV-infected BEAS-2B cells with UA treatment, overexpression of miR-136 and Ex527 treatment both significantly increased the inflammatory factors and cell apoptosis but decreased LC3B expression, and these changes were further enhanced by their combined treatment. CONCLUSION UA ameliorates RSV-induced lung infection in neonatal mice by activating miR-136-mediated Sirt1 signaling pathway.
Collapse
|
13
|
Suleri A, Gaiser C, Cecil CAM, Dijkzeul A, Neumann A, Labrecque JA, White T, Bergink V, Muetzel RL. Examining longitudinal associations between prenatal exposure to infections and child brain morphology. Brain Behav Immun 2024; 119:965-977. [PMID: 38750701 PMCID: PMC7616133 DOI: 10.1016/j.bbi.2024.05.014] [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: 10/02/2023] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Maternal infection during pregnancy has been identified as a prenatal risk factor for the later development of psychopathology in exposed offspring. Neuroimaging data collected during childhood has suggested a link between prenatal exposure to maternal infection and child brain structure and function, potentially offering a neurobiological explanation for the emergence of psychopathology. Additionally, preclinical studies utilizing repeated measures of neuroimaging data suggest that effects of prenatal maternal infection on the offspring's brain may normalize over time (i.e., catch-up growth). However, it remains unclear whether exposure to prenatal maternal infection in humans is related to long-term differential neurodevelopmental trajectories. Hence, this study aimed to investigate the association between prenatal exposure to infections on child brain development over time using repeated measures MRI data. METHODS We leveraged data from a population-based cohort, Generation R, in which we examined prospectively assessed self-reported infections at each trimester of pregnancy (N = 2,155). We further used three neuroimaging assessments (at mean ages 8, 10 and 14) to obtain cortical and subcortical measures of the offspring's brain morphology with MRI. Hereafter, we applied linear mixed-effects models, adjusting for several confounding factors, to estimate the association of prenatal maternal infection with child brain development over time. RESULTS We found that prenatal exposure to infection in the third trimester was associated with a slower decrease in volumes of the pars orbitalis, rostral anterior cingulate and superior frontal gyrus, and a faster increase in the middle temporal gyrus. In the temporal pole we observed a divergent pattern, specifically showing an increase in volume in offspring exposed to more infections compared to a decrease in volume in offspring exposed to fewer infections. We further observed associations in other frontal and temporal lobe structures after exposure to infections in any trimester, though these did not survive multiple testing correction. CONCLUSIONS Our results suggest that prenatal exposure to infections in the third trimester may be associated with slower age-related growth in the regions: pars orbitalis, rostral anterior cingulate and superior frontal gyrus, and faster age-related growth in the middle temporal gyrus across childhood, suggesting a potential sensitive period. Our results might be interpreted as an extension of longitudinal findings from preclinical studies, indicating that children exposed to prenatal infections could exhibit catch-up growth. However, given the lack of differences in brain volume between various infection groups at baseline, there may instead be either a longitudinal deviation or a subtle temporal deviation. Subsequent well-powered studies that extend into the period of full brain development (∼25 years) are needed to confirm whether the observed phenomenon is indeed catch-up growth, a longitudinal deviation, or a subtle temporal deviation.
Collapse
Affiliation(s)
- Anna Suleri
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Carolin Gaiser
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biomedical Data Sciences, Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annet Dijkzeul
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeremy A Labrecque
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA; Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
14
|
Mi Y, Liang Y, Liu Y, Bai Z, Li N, Tan S, Hou Y. Integrated network pharmacology and experimental validation-based approach to reveal the underlying mechanisms and key material basis of Jinhua Qinggan granules against acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117920. [PMID: 38373663 DOI: 10.1016/j.jep.2024.117920] [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: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jinhua Qinggan granules (JHQG), the traditional Chinese formula come into the market in 2016, has been proved clinically effective against coronavirus disease. Acute lung injury (ALI) is a major complication of respiratory infection such as coronavirus and influenza virus, with a high clinical fatality rate. Macrophage activation-induced inflammatory response plays a crucial role in the pathogenesis of ALI. However, the participation of inflammatory response in the efficacy of JHQG and its material basis against ALI is still unknown. AIM OF THE STUDY The research aims to investigate the inflammatory response-involved efficacy of JHQG on ALI, explore the "ingredient-target-pathway" mechanisms, and searching for key material basis of JHQG by integrated network pharmacology and experimental validation-based approach. MATERIALS AND METHODS Lipopolysaccharide (LPS)-induced ALI mice was established to assess the protective impact of JHQG. Network pharmacology was utilized to identify potential targets of JHQG and investigate its action mechanisms related to inflammatory response in treating ALI. The therapeutic effect and mechanism of the primary active ingredient in JHQG was verified through high performance liquid chromatography (HPLC) and a combination of wet experiments. RESULTS JHQG remarkably alleviated lung damage in mice model via suppressing macrophage activation, and inhibiting pro-inflammatory mediator level, p-ERK and p-STAT3 expression, TLR4/NF-κB activation. Network pharmacology combined with HPLC found luteolin is the main effective component of JHQG, and it could interact with TLR4/MD2 complex, further exerting the anti-inflammatory property and the protective role against ALI. CONCLUSIONS In summary, our finding clarified the underlying mechanisms and material basis of JHQG therapy for ALI by integrated network pharmacology and experimental validation-based strategy.
Collapse
Affiliation(s)
- Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yusheng Liang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Zisong Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China; School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Shaowen Tan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
15
|
Asadipooya K, Asadipooya A, Adatorwovor R. Combination of spironolactone and DPP-4 inhibitors for treatment of SARS-CoV-2 infection: a literature review. Arch Virol 2024; 169:122. [PMID: 38753071 DOI: 10.1007/s00705-024-06043-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 03/23/2024] [Indexed: 05/21/2024]
Abstract
Coronavirus disease 2019 (COVID-19) is still causing hospitalization and death, and vaccination appears to become less effective with each emerging variant. Spike, non-spike, and other possible unrecognized mutations have reduced the efficacy of recommended therapeutic approaches, including monoclonal antibodies, plasma transfusion, and antivirals. SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) and probably dipeptidyl peptidase 4 (DPP-4) to initiate the process of endocytosis by employing host proteases such as transmembrane serine protease-2 (TMPRSS-2) and ADAM metallopeptidase domain 17 (ADAM17). Spironolactone reduces the amount of soluble ACE2 and antagonizes TMPRSS-2 and ADAM17. DPP-4 inhibitors play immunomodulatory roles and may block viral entry. The efficacy of treatment with a combination of spironolactone and DPP-4 inhibitors does not appear to be affected by viral mutations. Therefore, the combination of spironolactone and DPP-4 inhibitors might improve the clinical outcome for COVID-19 patients by decreasing the efficiency of SARS-CoV-2 entry into cells and providing better anti-inflammatory, antiproliferative, and antifibrotic effects than those achieved using current therapeutic approaches such as antivirals and monoclonal antibodies.
Collapse
Affiliation(s)
- Kamyar Asadipooya
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Barnstable Brown Diabetes and Obesity Center, University of Kentucky, 2195 Harrodsburg Rd, Suite 125, Lexington, KY, 40504, USA.
| | - Artin Asadipooya
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
16
|
Mićanović D, Lazarević M, Kulaš J, Despotović S, Stegnjaić G, Jevtić B, Koprivica I, Mirkov I, Stanisavljević S, Nikolovski N, Miljković Đ, Saksida T. Ethyl pyruvate ameliorates acute respiratory distress syndrome in mice. Eur J Pharmacol 2024; 971:176509. [PMID: 38493914 DOI: 10.1016/j.ejphar.2024.176509] [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: 05/24/2023] [Revised: 02/15/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Acute respiratory distress syndrome (ARDS) became a focus of intensive research due to its death toll during the Covid-19 pandemic. An uncontrolled and excessive inflammatory response mediated by proinflammatory molecules such as high mobility group box protein 1 (HMGB1), IL-6, and TNF mounts as a response to infection. In this study, ethyl pyruvate (EP), a known inhibitor of HMGB1, was tested in the model of murine ARDS induced in C57BL/6 mice by intranasal administration of polyinosinic:polycytidylic acid (poly(I:C)). Intraperitoneal administration of EP ameliorated the ARDS-related histopathological changes in the lungs of poly(I:C)-induced ARDS and decreased numbers of immune cells in the lungs, broncho-alveolar lavage fluid and draining lymph nodes (DLN). Specifically, fewer CD8+ T cells and less activated CD4+ T cells were observed in DLN. Consequently, the lungs of EP-treated animals had fewer damage-inflicting CD8+ cells and macrophages. Additionally, the expression and production of proinflammatory cytokines, IL-17, IFN-γ and IL-6 were downregulated in the lungs. The expression of chemokine CCL5 which recruits immune cells into the lungs was also reduced. Finally, EP downregulated the expression of HMGB1 in the lungs. Our results imply that EP should be further evaluated as a potential candidate for ARDS therapy.
Collapse
Affiliation(s)
- Dragica Mićanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Milica Lazarević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Jelena Kulaš
- Department of Ecology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Sanja Despotović
- Institute of Histology and Embryology "Aleksandar Đ. Kostić", School of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia
| | - Goran Stegnjaić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Ivana Mirkov
- Department of Ecology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Neda Nikolovski
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
17
|
Paukner S, Kimber S, Cumper C, Rea-Davies T, Sueiro Ballesteros L, Kirkham C, Hargreaves A, Gelone SP, Richards C, Wicha WW. In Vivo Immune-Modulatory Activity of Lefamulin in an Influenza Virus A (H1N1) Infection Model in Mice. Int J Mol Sci 2024; 25:5401. [PMID: 38791439 PMCID: PMC11121702 DOI: 10.3390/ijms25105401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Lefamulin is a first-in-class systemic pleuromutilin antimicrobial and potent inhibitor of bacterial translation, and the most recent novel antimicrobial approved for the treatment of community-acquired pneumonia (CAP). It exhibits potent antibacterial activity against the most prevalent bacterial pathogens that cause typical and atypical pneumonia and other infectious diseases. Early studies indicate additional anti-inflammatory activity. In this study, we further investigated the immune-modulatory activity of lefamulin in the influenza A/H1N1 acute respiratory distress syndrome (ARDS) model in BALB/c mice. Comparators included azithromycin, an anti-inflammatory antimicrobial, and the antiviral oseltamivir. Lefamulin significantly decreased the total immune cell infiltration, specifically the neutrophils, inflammatory monocytes, CD4+ and CD8+ T-cells, NK cells, and B-cells into the lung by Day 6 at both doses tested compared to the untreated vehicle control group (placebo), whereas azithromycin and oseltamivir did not significantly affect the total immune cell counts at the tested dosing regimens. Bronchioalveolar lavage fluid concentrations of pro-inflammatory cytokines and chemokines including TNF-α, IL-6, IL-12p70, IL-17A, IFN-γ, and GM-CSF were significantly reduced, and MCP-1 concentrations were lowered (not significantly) by lefamulin at the clinically relevant 'low' dose on Day 3 when the viral load peaked. Similar effects were also observed for oseltamivir and azithromycin. Lefamulin also decreased the viral load (TCID50) by half a log10 by Day 6 and showed positive effects on the gross lung pathology and survival. Oseltamivir and lefamulin were efficacious in the suppression of the development of influenza-induced bronchi-interstitial pneumonia, whereas azithromycin did not show reduced pathology at the tested treatment regimen. The observed anti-inflammatory and immune-modulatory activity of lefamulin at the tested treatment regimens highlights a promising secondary pharmacological property of lefamulin. While these results require confirmation in a clinical trial, they indicate that lefamulin may provide an immune-modulatory activity beyond its proven potent antibacterial activity. This additional activity may benefit CAP patients and potentially prevent acute lung injury (ALI) and ARDS.
Collapse
Affiliation(s)
- Susanne Paukner
- Nabriva Therapeutics GmbH, Leberstrasse 20, 1110 Vienna, Austria;
| | - Sandra Kimber
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Charlotte Cumper
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Tina Rea-Davies
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Lorena Sueiro Ballesteros
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Christopher Kirkham
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | | | | - Claire Richards
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | |
Collapse
|
18
|
Pan T, Guo X, Yang D, Ding J, Chen C. Expression and significance of procalcitonin, leukotriene B4, serum amyloid A, and C-reactive protein in children with different types of pneumonia: An observational study. Medicine (Baltimore) 2024; 103:e37817. [PMID: 38728486 PMCID: PMC11081565 DOI: 10.1097/md.0000000000037817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/15/2024] [Indexed: 05/12/2024] Open
Abstract
This study aimed to investigate the expression and significance of serum procalcitonin (PCT), leukotriene B4 (LTB4), Serum amyloid A (SAA), and C-reactive protein (CRP) in children with different types of pneumonia caused by different pathogenic infections. One hundred and one children with pneumonia admitted to The Fifth People Hospital of Zhuhai from July 2019 to June 2020 were enrolled and divided into 38 cases in the bacterial group, 30 cases in the mycoplasma group, and 33 cases in the virus group according to the different types of pathogens. The patients were divided into 42 cases in the noncritical group, 33 cases in the critical group, and 26 cases in the very critical group according to the pediatric clinical illness score (PCIS), and 30 healthy children were selected as the control group during the same period. Comparison of serum PCT, SAA: bacterial group > mycoplasma group > viral group > control group with significant differences (P < .05). Receiver operator characteristic (ROC) analysis showed that the area under the curves (AUCs) of serum PCT, LTB4, SAA, and CRP for the diagnosis of bacterial pneumonia were 1.000, 0.531, 0.969, and 0.833, respectively, and the AUCs for the diagnosis of mycoplasma pneumonia were 0.653, 0.609, 0.547, and 0.652, respectively, and the AUCs for the diagnosis of viral pneumonia were 0.888, 0.570, 0.955, and 1.000, respectively. Comparison of serum PCT, LTB4, SAA: very critical group > critical group > noncritical group > control group, with significant differences (P < .05). Serum PCT, LTB4, and SAA were negatively correlated with PCIS score by Pearson analysis (P < .05). Serum PCT and SAA showed diagnostic value for bacterial pneumonia, and serum SAA and CRP showed diagnostic value for viral pneumonia; serum PCT, LTB4, and SAA correlate with severity of disease and show higher expression with worsening of the condition.
Collapse
Affiliation(s)
- Tinghui Pan
- Department of Paediatrics, The Fifth People’s Hospital of Zhuhai, Zhuhai, Guangdong, China
| | - Xiwen Guo
- Department of Human Anatomy and Histology & Embryology, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong, China
| | - Dehui Yang
- Department of Paediatrics, The Fifth People’s Hospital of Zhuhai, Zhuhai, Guangdong, China
| | - Jundong Ding
- Department of Paediatrics, The Fifth People’s Hospital of Zhuhai, Zhuhai, Guangdong, China
| | - Chengxian Chen
- Department of Neuromedicine, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Al-Hawary SIS, Jasim SA, Hjazi A, Ullah H, Bansal P, Deorari M, Sapaev IB, Ami AA, Mohmmed KH, Abosaoda MK. A new perspective on therapies involving B-cell depletion in autoimmune diseases. Mol Biol Rep 2024; 51:629. [PMID: 38717637 DOI: 10.1007/s11033-024-09575-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/22/2024] [Indexed: 06/30/2024]
Abstract
It has been rediscovered in the last fifteen years that B-cells play an active role in autoimmune etiology rather than just being spectators. The clinical success of B-cell depletion therapies (BCDTs) has contributed to this. BCDTs, including those that target CD20, CD19, and BAFF, were first developed to eradicate malignant B-cells. These days, they treat autoimmune conditions like multiple sclerosis and systemic lupus erythematosus. Particular surprises have resulted from the use of BCDTs in autoimmune diseases. For example, even in cases where BCDT is used to treat the condition, its effects on antibody-secreting plasma cells and antibody levels are restricted, even though these cells are regarded to play a detrimental pathogenic role in autoimmune diseases. In this Review, we provide an update on our knowledge of the biology of B-cells, examine the outcomes of clinical studies employing BCDT for autoimmune reasons, talk about potential explanations for the drug's mode of action, and make predictions about future approaches to targeting B-cells other than depletion.
Collapse
Affiliation(s)
| | | | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Himayat Ullah
- College of Medicine, Shaqra University, 15526, Shaqra, Saudi Arabia.
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - I B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers National Research University, Tashkent, Uzbekistan
- Scientific Researcher, Western Caspian University, Baku, Azerbaijan
| | - Ahmed Ali Ami
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | | | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Hillah, Iraq
| |
Collapse
|
20
|
Bhat AA, Afzal M, Goyal A, Gupta G, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Shahwan M, Paudel KR, Ali H, Sahu D, Prasher P, Singh SK, Dua K. The impact of formaldehyde exposure on lung inflammatory disorders: Insights into asthma, bronchitis, and pulmonary fibrosis. Chem Biol Interact 2024; 394:111002. [PMID: 38604395 DOI: 10.1016/j.cbi.2024.111002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
Lung inflammatory disorders are a major global health burden, impacting millions of people and raising rates of morbidity and death across many demographic groups. An industrial chemical and common environmental contaminant, formaldehyde (FA) presents serious health concerns to the respiratory system, including the onset and aggravation of lung inflammatory disorders. Epidemiological studies have shown significant associations between FA exposure levels and the incidence and severity of several respiratory diseases. FA causes inflammation in the respiratory tract via immunological activation, oxidative stress, and airway remodelling, aggravating pre-existing pulmonary inflammation and compromising lung function. Additionally, FA functions as a respiratory sensitizer, causing allergic responses and hypersensitivity pneumonitis in sensitive people. Understanding the complicated processes behind formaldehyde-induced lung inflammation is critical for directing targeted strategies aimed at minimizing environmental exposures and alleviating the burden of formaldehyde-related lung illnesses on global respiratory health. This abstract explores the intricate relationship between FA exposure and lung inflammatory diseases, including asthma, bronchitis, allergic inflammation, lung injury and pulmonary fibrosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Mahal Road, Jaipur, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2050, Australia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Dipak Sahu
- Department of Pharmacology, Amity University, Raipur, Chhattisgarh, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
21
|
Chivé C, Martίn-Faivre L, Eon-Bertho A, Alwardini C, Degrouard J, Albinet A, Noyalet G, Chevaillier S, Maisonneuve F, Sallenave JM, Devineau S, Michoud V, Garcia-Verdugo I, Baeza-Squiban A. Exposure to PM 2.5 modulate the pro-inflammatory and interferon responses against influenza virus infection in a human 3D bronchial epithelium model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 348:123781. [PMID: 38492752 DOI: 10.1016/j.envpol.2024.123781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Epidemiological studies showed a positive association between exposure to PM2.5 and the severity of influenza virus infection. However, the mechanisms by which PM2.5 can disrupt antiviral defence are still unclear. From this perspective, the objective of this study was to evaluate the effects of PM2.5 on antiviral signalling in the respiratory epithelium using the bronchial Calu-3 cell line grown at the air-liquid interface. Pre-exposure to PM2.5 before infection with the influenza virus was investigated, as well as a co-exposure. Although a physical interaction between the virus and the particles seems possible, no effect of PM2.5 on viral replication was observed during co-exposure, although a downregulation of IFN-β release was associated to PM2.5 exposure. However, pre-exposure slightly increased the viral nucleoprotein production and the pro-inflammatory response. Conversely, the level of the myxovirus resistance protein A (MxA), an interferon-stimulated gene (ISG) induced by IFN-β, was reduced. Therefore, these results suggest that pre-exposure to PM2.5 could alter the antiviral response of bronchial epithelial cells, increasing their susceptibility to viral infection.
Collapse
Affiliation(s)
- Chloé Chivé
- Université Paris Cité, Functional and Adaptive Biology Unit, UMR8251-CNRS, Paris, France; French Environment and Energy Management Agency 20, Avenue Du Grésillé - BP, 90406 49004, Angers, France
| | - Lydie Martίn-Faivre
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Alice Eon-Bertho
- Université Paris Cité, Functional and Adaptive Biology Unit, UMR8251-CNRS, Paris, France
| | - Christelle Alwardini
- Université Paris Cité, Functional and Adaptive Biology Unit, UMR8251-CNRS, Paris, France
| | - Jéril Degrouard
- Université Paris-Saclay, CNRS, Laboratoire de Physique des Solides, 91405, Orsay, France
| | - Alexandre Albinet
- Institut National de L'Environnement Industriel et des Risques (INERIS), Parc Technologique Alata BP2, 60550, Verneuil en Halatte, France
| | - Gael Noyalet
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, F-75013, Paris, France
| | - Servanne Chevaillier
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, F-75013, Paris, France
| | - Franck Maisonneuve
- Université Paris Est Créteil and Université Paris Cité, CNRS, LISA, F-94010, Créteil, France
| | - Jean-Michel Sallenave
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Stéphanie Devineau
- Université Paris Cité, Functional and Adaptive Biology Unit, UMR8251-CNRS, Paris, France
| | - Vincent Michoud
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, F-75013, Paris, France
| | - Ignacio Garcia-Verdugo
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France.
| | - Armelle Baeza-Squiban
- Université Paris Cité, Functional and Adaptive Biology Unit, UMR8251-CNRS, Paris, France
| |
Collapse
|
22
|
Li G, Yan K, Zhang W, Pan H, Guo P. ARDS and aging: TYMS emerges as a promising biomarker and therapeutic target. Front Immunol 2024; 15:1365206. [PMID: 38558817 PMCID: PMC10978671 DOI: 10.3389/fimmu.2024.1365206] [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: 01/03/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) is a common condition in the intensive care unit (ICU) with a high mortality rate, yet the diagnosis rate remains low. Recent studies have increasingly highlighted the role of aging in the occurrence and progression of ARDS. This study is committed to investigating the pathogenic mechanisms of cellular and genetic changes in elderly ARDS patients, providing theoretical support for the precise treatment of ARDS. Methods Gene expression profiles for control and ARDS samples were obtained from the Gene Expression Omnibus (GEO) database, while aging-related genes (ARGs) were sourced from the Human Aging Genomic Resources (HAGR) database. Differentially expressed genes (DEGs) were subjected to functional enrichment analysis to understand their roles in ARDS and aging. The Weighted Gene Co-expression Network Analysis (WGCNA) and machine learning pinpointed key modules and marker genes, with ROC curves illustrating their significance. The expression of four ARDS-ARDEGs was validated in lung samples from aged mice with ARDS using qRT-PCR. Gene set enrichment analysis (GSEA) investigated the signaling pathways and immune cell infiltration associated with TYMS expression. Single-nucleus RNA sequencing (snRNA-Seq) explored gene-level differences among cells to investigate intercellular communication during ARDS onset and progression. Results ARDEGs are involved in cellular responses to DNA damage stimuli, inflammatory reactions, and cellular senescence pathways. The MEmagenta module exhibited a significant correlation with elderly ARDS patients. The LASSO, RRF, and XGBoost algorithms were employed to screen for signature genes, including CKAP2, P2RY14, RBP2, and TYMS. Further validation emphasized the potential role of TYMS in the onset and progression of ARDS. Immune cell infiltration indicated differential proportion and correlations with TYMS expression. SnRNA-Seq and cell-cell communication analysis revealed that TYMS is highly expressed in endothelial cells, and the SEMA3 signaling pathway primarily mediates cell communication between endothelial cells and other cells. Conclusion Endothelial cell damage associated with aging could contribute to ARDS progression by triggering inflammation. TYMS emerges as a promising diagnostic biomarker and potential therapeutic target for ARDS.
Collapse
Affiliation(s)
- Gang Li
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ke Yan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wanyi Zhang
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haiyan Pan
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Pengxiang Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
23
|
Gheitasi H, Sabbaghian M, Fadaee M, Mohammadzadeh N, Shekarchi AA, Poortahmasebi V. The relationship between autophagy and respiratory viruses. Arch Microbiol 2024; 206:136. [PMID: 38436746 DOI: 10.1007/s00203-024-03838-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 03/05/2024]
Abstract
Respiratory viruses have caused severe global health problems and posed essential challenges to the medical community. In recent years, the role of autophagy as a critical process in cells in viral respiratory diseases has been noticed. One of the vital catabolic biological processes in the body is autophagy. Autophagy contributes to energy recovery by targeting and selectively directing foreign microorganisms, organelles, and senescent intracellular proteins to the lysosome for degradation and phagocytosis. Activation or suppression of autophagy is often initiated when foreign pathogenic organisms such as viruses infect cells. Because of its antiviral properties, several viruses may escape or resist this process by encoding viral proteins. Viruses can also use autophagy to enhance their replication or prolong the persistence of latent infections. Here, we provide an overview of autophagy and respiratory viruses such as coronavirus, rhinovirus, parainfluenza, influenza, adenovirus, and respiratory syncytial virus, and examine the interactions between them and the role of autophagy in the virus-host interaction process and the resulting virus replication strategy.
Collapse
Affiliation(s)
- Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Mohammadzadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Chen W, Zhu Y, Liu R, Kong B, Xia N, Zhao Y, Sun L. Screening Therapeutic Effects of MSC-EVs to Acute Lung Injury Model on A Chip. Adv Healthc Mater 2024; 13:e2303123. [PMID: 38084928 DOI: 10.1002/adhm.202303123] [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: 09/17/2023] [Revised: 11/29/2023] [Indexed: 12/19/2023]
Abstract
Acute lung injury (ALI) is a lethal disease with high mortality rate, and its physiologically relevant models that could mimic human disease processes are urgently needed to study pathophysiology and predict drug efficacy. Here, this work presents a novel lipopolysaccharide (LPS) based ALI model on a microfluidic chip that reconstitutes an air-liquid interface lined by human alveolar epithelium and microvascular endothelium for screening the therapeutic effects of mesenchymal stem cells (MSC) derived extracellular vesicles (MSC-EVs) to the biomimetic ALI. The air-liquid interface is established by coculture of alveolar epithelium and microvascular endothelium on the opposite sides of the porous membrane. The functionalized architecture is characterized by integrate cell layers and suitable permeability. Using this biomimetic microsystem, LPS based ALI model is established, which exhibits the disrupted alveolar-capillary barrier, reduced transepithelial/transendothelial electrical resistance (TEER), and impaired expression of junction proteins. As a reliable disease model, this work examines the effects of MSC-EVs, and the data indicate the therapeutic potential of EVs for severe ALI. MSC-EVs can alleviate barrier disruption by restoring both the epithelial and endothelial barrier integrity. They hope this study can become a unique approach to study human pathophysiology of ALI and advance drug development.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Nan Xia
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, China
| |
Collapse
|
25
|
Wei F, Yin Y, Li J, Chang Y, Zhang S, Zhao W, Ma X. Essential oil from Inula japonica Thunb. And its phenolic constituents ameliorate pulmonary injury and fibrosis in bleomycin-treated mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117169. [PMID: 37704119 DOI: 10.1016/j.jep.2023.117169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary injury and fibrosis can be caused by various factors because of their inflammatory nature, both can lead to serious clinical consequences. Inula japonica Thunb. is used in traditional Chinese medicine for the treatment of lung diseases. However, the effect and mechanism of action of the essential oil of I. japonica (EOI) on pulmonary injury and fibrosis are not well understood. AIM OF THE STUDY To investigate the therapeutic effects of EOI on mice with bleomycin (BLM)-induced acute pulmonary injury and chronic fibrosis formation, as well as its potential mechanism. MATERIALS AND METHODS A short-term mouse model of pulmonary injury was established by intratracheal injection of BLM to investigate the anti-inflammatory effect of EOI, and a long-term model of pulmonary fibrosis was used to explore the anti-fibrosis effect of EOI. High-dose EOI (200 mg/kg) was administered intragastrically, and low-dose (50 mg/kg) was administered by intratracheal injection. Gas chromatography-mass spectrometry (GC-MS) was used to identify the ingredients in EOI, and high-performance liquid chromatography (HPLC) was performed for the preparation of EOI compounds. Western blot and real-time qPCR were used to verify the effects of EOI and its active composition on inflammation, oxidative stress and fibrosis signaling pathway. RESULTS Treatment with EOI significantly reduced the inflammation and oxidative stress by reducing the levels of inflammatory and oxidative cytokines such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and malondialdehyde in BLM-treated mice with acute pulmonary injury. EOI treatment could also suppress the formation of fibrous tissue in mice with BLM-induced pulmonary fibrosis through inhibiting TGF-β/Smad and PI3K/Akt pathways. Chromatographic analysis and preparation suggested that fatty acid and phenol derivatives are present in EOI. Based on cellular inflammation and fibrosis models, the phenolic compounds in EOI can represent the anti-inflammatory and anti-fibrotic effects of EOI by regulating pro-inflammatory and pro-fibrotic cytokines such as NO, TNF-α, IL-6, TGF-β1, and α-SMA. CONCLUSION EOI ameliorated BLM-induced pulmonary injury and fibrosis in mice by inhibiting the inflammatory response and regulating the redox equilibrium, as well as by mediating TGFβ/Smad and PI3K/Akt, which suggested that EOI has potential to treat pulmonary diseases.
Collapse
Affiliation(s)
- Fan Wei
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuzhen Yin
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Li
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yibo Chang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuyuan Zhang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wenyu Zhao
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Xiaochi Ma
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
26
|
Guo K, Yombo DJK, Wang Z, Navaeiseddighi Z, Xu J, Schmit T, Ahamad N, Tripathi J, De Kumar B, Mathur R, Hur J, Sun J, Olszewski MA, Khan N. The chemokine receptor CXCR3 promotes CD8 + T cell-dependent lung pathology during influenza pathogenesis. SCIENCE ADVANCES 2024; 10:eadj1120. [PMID: 38170765 PMCID: PMC10776024 DOI: 10.1126/sciadv.adj1120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
The dual role of CD8+ T cells in influenza control and lung pathology is increasingly appreciated. To explore whether protective and pathological functions can be linked to specific subsets, we dissected CD8+ T responses in influenza-infected murine lungs. Our single-cell RNA-sequencing (scRNA-seq) analysis revealed notable diversity in CD8+ T subpopulations during peak viral load and infection-resolved state. While enrichment of a Cxcr3hi CD8+ T effector subset was associated with a more robust cytotoxic response, both CD8+ T effector and central memory exhibited equally potent effector potential. The scRNA-seq analysis identified unique regulons regulating the cytotoxic response in CD8+ T cells. The late-stage CD8+ T blockade in influenza-cleared lungs or continuous CXCR3 blockade mitigated lung injury without affecting viral clearance. Furthermore, adoptive transfer of wild-type CD8+ T cells exacerbated influenza lung pathology in Cxcr3-/- mice. Collectively, our data imply that CXCR3 interception could have a therapeutic effect in preventing influenza-linked lung injury.
Collapse
Affiliation(s)
- Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan J. K. Yombo
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Zhihan Wang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | | | - Jintao Xu
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, MI 48109, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Taylor Schmit
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Nassem Ahamad
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA
| | - Jitendra Tripathi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Bony De Kumar
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Ramkumar Mathur
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
| | - Michal A. Olszewski
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, MI 48109, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Nadeem Khan
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
27
|
Pontiroli AE, Scovenna F, Carlini V, Tagliabue E, Martin-Delgado J, Sala LL, Tanzi E, Zanoni I. Vaccination against influenza viruses reduces infection, not hospitalization or death, from respiratory COVID-19: A systematic review and meta-analysis. J Med Virol 2024; 96:e29343. [PMID: 38163281 PMCID: PMC10924223 DOI: 10.1002/jmv.29343] [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: 09/05/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 and has brought a huge burden in terms of human lives. Strict social distance and influenza vaccination have been recommended to avoid co-infections between influenza viruses and SARS-CoV-2. Scattered reports suggested a protective effect of influenza vaccine on COVID-19 development and severity. We analyzed 51 studies on the capacity of influenza vaccination to affect infection with SARS-CoV-2, hospitalization, admission to Intensive Care Units (ICU), and mortality. All subjects taken into consideration did not receive any anti-SARS-CoV-2 vaccine, although their status with respect to previous infections with SARS-CoV-2 is not known. Comparison between vaccinated and not-vaccinated subjects for each of the four endpoints was expressed as odds ratio (OR), with 95% confidence intervals (CIs); all analyses were performed by DerSimonian and Laird model, and Hartung-Knapp model when studies were less than 10. In a total of 61 029 936 subjects from 33 studies, influenza vaccination reduced frequency of SARS-CoV-2 infection [OR plus 95% CI = 0.70 (0.65-0.77)]. The effect was significant in all studies together, in health care workers and in the general population; distance from influenza vaccination and the type of vaccine were also of importance. In 98 174 subjects from 11 studies, frequency of ICU admission was reduced with influenza vaccination [OR (95% CI) = 0.71 (0.54-0.94)]; the effect was significant in all studies together, in pregnant women and in hospitalized subjects. In contrast, in 4 737 328 subjects from 14 studies hospitalization was not modified [OR (95% CI) = 1.05 (0.82-1.35)], and in 4 139 660 subjects from 19 studies, mortality was not modified [OR (95% CI) = 0.76 (0.26-2.20)]. Our study emphasizes the importance of influenza vaccination in the protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Antonio E. Pontiroli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Francesco Scovenna
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Valentina Carlini
- IRCCS MultiMedica, Laboratory of Cardiovascular and Dysmetabolic Disease, 20138 Milan, Italy
| | - Elena Tagliabue
- IRCCS MultiMedica, Value-Based Healthcare Unit, 20099 Milan, Italy
| | - Jimmy Martin-Delgado
- Hospital Luis Vernaza, Junta de Beneficiencia de Guayaquil 090603, Ecuador
- Instituto de Investigacion e Innovacion en Salud Integral, Universidad Catolica de Santiago de Guayaquil, Guayaquil 090603, Ecuador
| | - Lucia La Sala
- IRCCS MultiMedica, Laboratory of Cardiovascular and Dysmetabolic Disease, 20138 Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Tanzi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School, Boston Children’s Hospital, Division of Immunology and Division of Gastroenterology, Boston, MA 02115, USA
| |
Collapse
|
28
|
Daskou M, Fotooh Abadi L, Gain C, Wong M, Sharma E, Kombe Kombe AJ, Nanduri R, Kelesidis T. The Role of the NRF2 Pathway in the Pathogenesis of Viral Respiratory Infections. Pathogens 2023; 13:39. [PMID: 38251346 PMCID: PMC10819673 DOI: 10.3390/pathogens13010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
In humans, acute and chronic respiratory infections caused by viruses are associated with considerable morbidity and mortality. Respiratory viruses infect airway epithelial cells and induce oxidative stress, yet the exact pathogenesis remains unclear. Oxidative stress activates the transcription factor NRF2, which plays a key role in alleviating redox-induced cellular injury. The transcriptional activation of NRF2 has been reported to affect both viral replication and associated inflammation pathways. There is complex bidirectional crosstalk between virus replication and the NRF2 pathway because virus replication directly or indirectly regulates NRF2 expression, and NRF2 activation can reversely hamper viral replication and viral spread across cells and tissues. In this review, we discuss the complex role of the NRF2 pathway in the regulation of the pathogenesis of the main respiratory viruses, including coronaviruses, influenza viruses, respiratory syncytial virus (RSV), and rhinoviruses. We also summarize the scientific evidence regarding the effects of the known NRF2 agonists that can be utilized to alter the NRF2 pathway.
Collapse
Affiliation(s)
- Maria Daskou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Leila Fotooh Abadi
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.F.A.); (R.N.)
| | - Chandrima Gain
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Wong
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Eashan Sharma
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Arnaud John Kombe Kombe
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.F.A.); (R.N.)
| | - Ravikanth Nanduri
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.F.A.); (R.N.)
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.F.A.); (R.N.)
| |
Collapse
|
29
|
Sellarès-Nadal J, Burgos J, Velasquez F, Martin-Gómez MT, Antón A, Romero-Herrera D, Eremiev S, Bosch-Nicolau P, Rodriguez-Pardo D, Len O, Falcó V. Impact of viral detection in patients with community-acquired pneumonia: An observational cohort study. Med Clin (Barc) 2023; 161:523-529. [PMID: 37598051 DOI: 10.1016/j.medcli.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 08/21/2023]
Abstract
PURPOSE The presence of a respiratory virus in patients with community-acquired pneumonia (CAP) may have an impact on the bacterial etiology and clinical presentation. In this study we aimed to assess the role of viral infection in the bacterial etiology and outcomes of patients with CAP. METHODS We performed a retrospective study of all adults hospitalized with CAP between November 2017 and October 2018. Patients were classified according to the presence of viral infection. An unvaried and a multivaried analysis were performed to identify variables associated with viral infection and clinical outcomes. RESULTS Overall 590 patients were included. A microorganism was documented in 375 cases (63.5%). A viral infection was demonstrated in 118 (20%). The main pathogens were Streptococcus pneumoniae (35.8%), Staphylococcus aureus (2.9%) and influenza virus (10.8%). A trend to a higher rate of S. aureus (p=0.06) in patients with viral infection was observed. Patients with viral infection had more often bilateral consolidation patterns (17.8% vs 10.8%, p=0.04), respiratory failure (59.3% vs 42.8%, p=0.001), ICU admission (17.8% vs 7%, p=0.001) and invasive mechanical ventilation (9.3% vs 2.8%, p=0.003). Risk factors for respiratory failure were chronic lung disease, age >65 years, positive blood cultures and viral infection. Influenza, virus but no other respiratory viruses, was associated with respiratory failure (OR, 3.72; 95% CI, 2.06-6.73). CONCLUSIONS Our study reinforces the idea that co-viral infection has an impact in the clinical presentation of CAP causing a more severe clinical picture. This impact seems to be mainly due to influenza virus infection.
Collapse
Affiliation(s)
- Julia Sellarès-Nadal
- Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain; Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain; Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Joaquin Burgos
- Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain; Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain.
| | - Fernando Velasquez
- Microbiology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | | | - Andrés Antón
- Microbiology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Dani Romero-Herrera
- Microbiology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Simeón Eremiev
- Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain
| | - Pau Bosch-Nicolau
- Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain; Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain
| | - Dolors Rodriguez-Pardo
- Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain
| | - Oscar Len
- Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain
| | - Vicenç Falcó
- Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain; Infectious Diseases Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona, Spain
| |
Collapse
|
30
|
He Q, Liu Y, Yin P, Gao Y, Kan H, Zhou M, Chen R, Li Y. Differentiating the impacts of ambient temperature on pneumonia mortality of various infectious causes: a nationwide, individual-level, case-crossover study. EBioMedicine 2023; 98:104854. [PMID: 38251462 PMCID: PMC10628343 DOI: 10.1016/j.ebiom.2023.104854] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND It remains unknown how ambient temperature impact pneumonia of various infectious causes. METHODS Based on the national death registry covering all counties in Chinese mainland, we conducted an individual-level case-crossover study in China from 2013 to 2019. Exposures were assigned at residential addresses for each decedent. Conditional logistic regression model combined with distributed lag non-linear models were used to estimate the exposure-response associations. The attributable fractions due to non-optimum temperature were calculated after accounting for spatial and temporal patterns for the excess risks. FINDINGS The exposure-response curves were inversely J-shaped with both low and high temperature increasing the risks, and the effect of low temperature was stronger. Extremely low temperature was associated with higher magnitude of influenza-related pneumonia [relative risk (RR): 2.46, 95% confidence interval (CI): 1.62-3.74], than viral pneumonia (RR: 1.89, 95% CI: 1.55-2.30) and bacterial pneumonia (RR: 1.81, 95% CI: 1.56-2.09). The magnitudes of RRs associated with extremely high temperature were similar among the three categories of pneumonia. The mortality attributable fraction for influenza-related pneumonia (29.78%) was the highest. The effects were stronger in people of low education level or residence in the north. INTERPRETATION This nationwide study presents findings on the varied risk and burden of pneumonia mortality of various infectious causes, and highlights the susceptibility of influenza-related pneumonia to ambient low temperature. FUNDING This study is supported by the National Key Research and Development Program (2022YFC3702701), the Shanghai Municipal Science and Technology Commission (21TQ015) and Shanghai International Science and Technology Partnership Project (21230780200).
Collapse
Affiliation(s)
- Qinglin He
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Yunning Liu
- National Center for Chronic Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Peng Yin
- National Center for Chronic Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Ya Gao
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Maigeng Zhou
- National Center for Chronic Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200032, China.
| | - Yanming Li
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
31
|
Maina JN. A critical assessment of the cellular defences of the avian respiratory system: are birds in general and poultry in particular relatively more susceptible to pulmonary infections/afflictions? Biol Rev Camb Philos Soc 2023; 98:2152-2187. [PMID: 37489059 DOI: 10.1111/brv.13000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023]
Abstract
In commercial poultry farming, respiratory diseases cause high morbidities and mortalities, begetting colossal economic losses. Without empirical evidence, early observations led to the supposition that birds in general, and poultry in particular, have weak innate and adaptive pulmonary defences and are therefore highly susceptible to injury by pathogens. Recent findings have, however, shown that birds possess notably efficient pulmonary defences that include: (i) a structurally complex three-tiered airway arrangement with aerodynamically intricate air-flow dynamics that provide efficient filtration of inhaled air; (ii) a specialised airway mucosal lining that comprises air-filtering (ciliated) cells and various resident phagocytic cells such as surface and tissue macrophages, dendritic cells and lymphocytes; (iii) an exceptionally efficient mucociliary escalator system that efficiently removes trapped foreign agents; (iv) phagocytotic atrial and infundibular epithelial cells; (v) phagocytically competent surface macrophages that destroy pathogens and injurious particulates; (vi) pulmonary intravascular macrophages that protect the lung from the vascular side; and (vii) proficiently phagocytic pulmonary extravasated erythrocytes. Additionally, the avian respiratory system rapidly translocates phagocytic cells onto the respiratory surface, ostensibly from the subepithelial space and the circulatory system: the mobilised cells complement the surface macrophages in destroying foreign agents. Further studies are needed to determine whether the posited weak defence of the avian respiratory system is a global avian feature or is exclusive to poultry. This review argues that any inadequacies of pulmonary defences in poultry may have derived from exacting genetic manipulation(s) for traits such as rapid weight gain from efficient conversion of food into meat and eggs and the harsh environmental conditions and severe husbandry operations in modern poultry farming. To reduce pulmonary diseases and their severity, greater effort must be directed at establishment of optimal poultry housing conditions and use of more humane husbandry practices.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park Campus, Kingsway Avenue, Johannesburg, 2006, South Africa
| |
Collapse
|
32
|
Hegazy GE, Abu-Serie MM, Soliman NA, Teleb M, Abdel-Fattah YR. Superior anti-pulmonary viral potential of Natrialba sp. M6-producing surfactin and C50 carotenoid pigment with unveiling its action modes. Virol J 2023; 20:249. [PMID: 37904234 PMCID: PMC10614327 DOI: 10.1186/s12985-023-02215-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/20/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Respiratory viruses, particularly adenoviruses (ADV), influenza A virus (e.g., H1N1), and coronaviruses (e.g., HCoV-229E and SARS-CoV-2) pose a global public health problem. Therefore, developing natural wide-spectrum antiviral compounds for disrupting the viral life cycle with antioxidant activity provides an efficient treatment approach. Herein, biosurfactant (Sur) and C50 carotenoid pigment (Pig) of haloalkaliphilic archaeon Natrialba sp. M6 which exhibited potent efficacy against hepatitis and anti-herpes simplex viruses, were investigated against pulmonary viruses. METHODS The cytotoxicity of the extracted Sur and Pig was examined on susceptible cell lines for ADV, HIN1, HCoV-229E, and SARS-CoV-2. Their potential against the cytopathic activity of these viruses was detected with investigating the action modes (including, virucidal, anti-adsorption, and anti-replication), unveiling the main mechanisms, and using molecular docking analysis. Radical scavenging activity was determined and HPLC analysis for potent extract (Sur) was performed. RESULTS All current investigations stated higher anti-pulmonary viruses of Sur than Pig via mainly virucidal and/or anti-replicative modes. Moreover, Sur had stronger ADV's capsid protein binding, ADV's DNA polymerase inhibition, suppressing hemagglutinin and neuraminidase of H1N1, and inhibiting chymotrypsin-like (3CL) protease of SARS-CoV-2, supporting with in-silico analysis, as well as radical scavenging activity than Pig. HPLC analysis of Sur confirmed the predominate presence of surfactin in it. CONCLUSION This study declared the promising efficacy of Sur as an efficient pharmacological treatment option for these pulmonary viruses and considered as guide for further in vivo research.
Collapse
Affiliation(s)
- Ghada E Hegazy
- National Institute of Oceanography and Fisheries, NIOF, Cairo, Egypt.
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt.
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt.
| | - Nadia A Soliman
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt.
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Yasser R Abdel-Fattah
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt.
| |
Collapse
|
33
|
Jaafar A, Albarazanchi A, Kadhim MJ, Darvin ME, Váczi T, Tuchin VV, Veres M. Impact of e-cigarette liquid on porcine lung tissue-Ex vivo confocal Raman micro-spectroscopy study. JOURNAL OF BIOPHOTONICS 2023:e202300336. [PMID: 37851480 DOI: 10.1002/jbio.202300336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023]
Abstract
Ex vivo porcine lung immersed in e-liquid was investigated in-depth using confocal Raman micro-spectroscopy to assess the e-liquid influence on the lung. It was found that lung-related Raman band intensities at 1002, 1548, 1618 and 1655 cm-1 increased after first and second treatments except the surface, which was attributed to the well-known optical clearing (OC) effect due to alveoli filling with e-liquid resulting in light scattering reduction. The autofluorescence enhancement was explained by oxidative stress induced in lung during exposure to e-liquid. Moreover, e-liquid induced collagen dehydration was revealed by the I937 /I926 Raman band intensity ratio change. The effect was enhanced after the second treatment of the same lung tissue that indicates the possibility of multi-step OC treatment. We hypothesize that the nicotine-flavour-free e-liquids containing glycerol and propylene glycol could potentially be used in clinical protocols as OC agent for enhanced in-depth Raman-guided bronchoscopy.
Collapse
Affiliation(s)
- Ali Jaafar
- Institute for Solid State Physics and Optics, Wigner Research Center for Physics, Budapest, Hungary
- Institute of Physics, University of Szeged, Szeged, Hungary
- Ministry of Higher Education and Scientific Research, Baghdad, Iraq
| | - Abbas Albarazanchi
- Department of Physics, College of Science, Mustansiriyah University, Baghdad, Iraq
| | | | | | - Tamás Váczi
- Institute for Solid State Physics and Optics, Wigner Research Center for Physics, Budapest, Hungary
| | - Valery V Tuchin
- Institute of Physics and Science Medical Center, Saratov State University, Saratov, Russia
- Laboratory of Laser Diagnostics of Technical and Living Systems, Institute of Precision Mechanics and Control, FRC "Saratov Scientific Centre of the Russian Academy of Sciences", Saratov, Russia
| | - Miklós Veres
- Institute for Solid State Physics and Optics, Wigner Research Center for Physics, Budapest, Hungary
| |
Collapse
|
34
|
Bermúdez-Barrezueta L, López-Casillas P, Rojo-Rello S, Sáez-García L, Marugán-Miguelsanz JM, Pino-Vázquez MDLA. Outcomes of viral coinfections in infants hospitalized for acute bronchiolitis. Virol J 2023; 20:235. [PMID: 37845714 PMCID: PMC10577995 DOI: 10.1186/s12985-023-02197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The clinical relevance of the detection of multiple respiratory viruses in acute bronchiolitis (AB) has not been established. Our goal was to evaluate the effect of viral coinfections on the progression and severity of AB. METHODS A retrospective observational study was conducted in a tertiary hospital in Spain from September 2012 to March 2020. Infants admitted for AB with at least one respiratory virus identified by molecular diagnostic techniques were included. A comparison was made between single-virus infections and viral coinfections. The evolution and severity of AB were determined based on the days of hospitalization and admission to the pediatric intensive care unit (PICU). RESULTS Four hundred forty-five patients were included (58.4% male). The median weight was 5.2 kg (IQR 4.2-6.5), and the median age was 2.5 months (IQR 1.4-4.6). A total of 105 patients (23.6%) were admitted to the PICU. Respiratory syncytial virus (RSV) was the most frequent etiological agent (77.1%). A single virus was detected in 270 patients (60.7%), and viral coinfections were detected in 175 (39.3%), of which 126 (28.3%) had two viruses and 49 (11%) had three or more viruses. Hospital length of stay (LOS) increased in proportion to the number of viruses detected, with a median of 6 days (IQR 4-8) for single infections, 7 days (IQR 4-9) for coinfections with two viruses and 8 days (IQR 5-11) for coinfections with ≥ 3 viruses (p = 0.003). The adjusted Cox regression model showed that the detection of ≥ 3 viruses was an independent risk factor for a longer hospital LOS (HR 0.568, 95% CI 0.410-0.785). No significant association was observed between viral coinfections and the need for PICU admission (OR 1.151; 95% CI 0.737-1.797). CONCLUSIONS Viral coinfections modified the natural history of AB, prolonging the hospital LOS in proportion to the number of viruses detected without increasing the need for admission to the PICU.
Collapse
Affiliation(s)
- Lorena Bermúdez-Barrezueta
- División of Pediatric and Neonatal Intensive Care, Department of Pediatrics, Hospital Clínico Universitario de Valladolid, Av. Ramón y Cajal, 3, 47003, Valladolid, Spain.
- Department of Pediatrics, Faculty of Medicine, Valladolid University, Valladolid, Spain.
| | - Pablo López-Casillas
- División of Pediatric and Neonatal Intensive Care, Department of Pediatrics, Hospital Clínico Universitario de Valladolid, Av. Ramón y Cajal, 3, 47003, Valladolid, Spain
| | - Silvia Rojo-Rello
- Microbiology and Immunology Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Laura Sáez-García
- Division of Pediatric Intensive Care, Reina Sofía Hospital, Córdoba, Spain
| | - José Manuel Marugán-Miguelsanz
- Department of Pediatrics, Faculty of Medicine, Valladolid University, Valladolid, Spain
- Division of Gastroenterology and Pediatric Nutrition, Head of Department of Pediatrics, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - María de la Asunción Pino-Vázquez
- División of Pediatric and Neonatal Intensive Care, Department of Pediatrics, Hospital Clínico Universitario de Valladolid, Av. Ramón y Cajal, 3, 47003, Valladolid, Spain
| |
Collapse
|
35
|
Rutkowska E, Kwiecień I, Pietruszka-Wałęka E, Więsik-Szewczyk E, Rzepecki P, Jahnz-Różyk K. Analysis of Leukocyte Subpopulations by Flow Cytometry during Hospitalization Depending on the Severity of COVID-19 Course. Biomedicines 2023; 11:2728. [PMID: 37893102 PMCID: PMC10604221 DOI: 10.3390/biomedicines11102728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The mechanisms underlying the immune response to coronavirus disease 2019 (COVID-19) and the recovery process have not been fully elucidated. The aim of the study was to analyze leukocyte subpopulations in patients at significant time points (at diagnosis, and 3 and 6 months after infection) selected according to the analysis of changes in the lungs by the CT classification system, considering the severity of the disease. The study groups consisted of severe and non-severe COVID-19 patients. There was a significant decrease in CD8+ T cells, NK and eosinophils, with an increasing percentage of neutrophils during hospitalization. We noticed lower levels of CD4 and CD8 T lymphocytes, eosinophils, basophils, and CD16+ monocytes and elevated neutrophil levels in severe patients relative to non-severe patients. Three months after infection, we observed higher levels of basophils, and after 6 months, higher CD4/CD8 ratios and T cell levels in the severe compared to non-severe group. Non-severe patients showed significant changes in the leukocyte populations studied at time of hospitalization and both within 3 months and 6 months of onset. The CT CSS classification with parameters of the flow cytometry method used for COVID-19 patients determined changes that proved useful in the initial evaluation of patients.
Collapse
Affiliation(s)
- Elżbieta Rutkowska
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Iwona Kwiecień
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Ewa Pietruszka-Wałęka
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Piotr Rzepecki
- Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| |
Collapse
|
36
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [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: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
37
|
Adhikari S, Baral P. Protocol for neonatal respiratory syncytial virus infection in mice and immune analysis of infected lungs and bronchoalveolar lavage fluid. STAR Protoc 2023; 4:102434. [PMID: 37432860 PMCID: PMC10362174 DOI: 10.1016/j.xpro.2023.102434] [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: 04/13/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection in infants and toddlers is a major public health problem. Here, we provide a protocol for neonatal RSV infection in mice and immune analysis of infected lungs and bronchoalveolar lavage (BAL) fluid. We describe steps for anesthesia and intranasal inoculation, weight monitoring, and whole lung collection. We then detail BAL fluid immune and whole lung analyses. This protocol can be used for neonatal pulmonary infection with other viruses or bacteria.
Collapse
Affiliation(s)
- Sandeep Adhikari
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | - Pankaj Baral
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
38
|
Belizário J, Garay-Malpartida M, Faintuch J. Lung microbiome and origins of the respiratory diseases. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100065. [PMID: 37456520 PMCID: PMC10339129 DOI: 10.1016/j.crimmu.2023.100065] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/08/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
The studies on the composition of the human microbiomes in healthy individuals, its variability in the course of inflammation, infection, antibiotic therapy, diets and different pathological conditions have revealed their intra and inter-kingdom relationships. The lung microbiome comprises of major species members of the phylum Bacteroidetes, Firmicutes, Actinobacteria, Fusobacteria and Proteobacteria, which are distributed in ecological niches along nasal cavity, nasopharynx, oropharynx, trachea and in the lungs. Commensal and pathogenic species are maintained in equilibrium as they have strong relationships. Bacterial overgrowth after dysbiosis and/or imbalanced of CD4+ helper T cells, CD8+ cytotoxic T cells and regulatory T cells (Treg) populations can promote lung inflammatory reactions and distress, and consequently acute and chronic respiratory diseases. This review is aimed to summarize the latest advances in resident lung microbiome and its participation in most common pulmonary infections and pneumonia, community-acquired pneumonia (CAP), ventilator-associated pneumonia (VAP), immunodeficiency associated pneumonia, SARS-CoV-2-associated pneumonia, acute respiratory distress syndrome (ARDS) and chronic obstructive pulmonary disease (COPD). We briefly describe physiological and immunological mechanisms that selectively create advantages or disadvantages for relative growth of pathogenic bacterial species in the respiratory tract. At the end, we propose some directions and analytical methods that may facilitate the identification of key genera and species of resident and transient microbes involved in the respiratory diseases' initiation and progression.
Collapse
Affiliation(s)
- José Belizário
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo, CEP 03828-000, Brazil
| | - Miguel Garay-Malpartida
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo, CEP 03828-000, Brazil
| | - Joel Faintuch
- Department of Gastroenterology of the Clinics Hospital of the University of São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 255, São Paulo, CEP 05403-000, Brazil
| |
Collapse
|
39
|
Latreille E, Lee WL. Modulation of the Host Response as a Therapeutic Strategy in Severe Lung Infections. Viruses 2023; 15:1462. [PMID: 37515150 PMCID: PMC10386155 DOI: 10.3390/v15071462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Respiratory pathogens such as influenza and SARS-CoV-2 can cause severe lung infections leading to acute respiratory distress syndrome (ARDS). The pathophysiology of ARDS includes an excessive host immune response, lung epithelial and endothelial cell death and loss of the epithelial and endothelial barrier integrity, culminating in pulmonary oedema and respiratory failure. Traditional approaches for the treatment of respiratory infections include drugs that exert direct anti-pathogen effects (e.g., antivirals). However, such agents are typically ineffective or insufficient after the development of ARDS. Modulation of the host response has emerged as a promising alternative therapeutic approach to mitigate damage to the host for the treatment of respiratory infections; in principle, this strategy should also be less susceptible to the development of pathogen resistance. In this review, we discuss different host-targeting strategies against pathogen-induced ARDS. Developing therapeutics that enhance the host response is a pathogen-agnostic approach that will help prepare for the next pandemic.
Collapse
Affiliation(s)
- Elyse Latreille
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Warren L Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
- Department of Medicine, Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
40
|
Li L, Liu T, Wang Q, Ding Y, Jiang Y, Wu Z, Wang X, Dou H, Jia Y, Jiao B. Genetic characterization and whole-genome sequencing-based genetic analysis of influenza virus in Jining City during 2021-2022. Front Microbiol 2023; 14:1196451. [PMID: 37426015 PMCID: PMC10324579 DOI: 10.3389/fmicb.2023.1196451] [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: 03/29/2023] [Accepted: 05/02/2023] [Indexed: 07/11/2023] Open
Abstract
Background The influenza virus poses a significant threat to global public health due to its high mutation rate. Continuous surveillance, development of new vaccines, and public health measures are crucial in managing and mitigating the impact of influenza outbreaks. Methods Nasal swabs were collected from individuals with influenza-like symptoms in Jining City during 2021-2022. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect influenza A viruses, followed by isolation using MDCK cells. Additionally, nucleic acid detection was performed to identify influenza A H1N1, seasonal H3N2, B/Victoria, and B/Yamagata strains. Whole-genome sequencing was conducted on 24 influenza virus strains, and subsequent analyses included characterization, phylogenetic construction, mutation analysis, and assessment of nucleotide diversity. Results A total of 1,543 throat swab samples were collected. The study revealed the dominance of the B/Victoria influenza virus in Jining during 2021-2022. Whole-genome sequencing showed co-prevalence of B/Victoria influenza viruses in the branches of Victoria clade 1A.3a.1 and Victoria clade 1A.3a.2, with a higher incidence observed in winter and spring. Comparative analysis demonstrated lower similarity in the HA, MP, and PB2 gene segments of the 24 sequenced influenza virus strains compared to the Northern Hemisphere vaccine strain B/Washington/02/2019. Mutations were identified in all antigenic epitopes of the HA protein at R133G, N150K, and N197D, and the 17-sequence antigenic epitopes exhibited more than 4 amino acid variation sites, resulting in antigenic drift. Moreover, one sequence had a D197N mutation in the NA protein, while seven sequences had a K338R mutation in the PA protein. Conclusion This study highlights the predominant presence of B/Victoria influenza strain in Jining from 2021 to 2022. The analysis also identified amino acid site variations in the antigenic epitopes, contributing to antigenic drift.
Collapse
Affiliation(s)
- Libo Li
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Tiantian Liu
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Qingchuan Wang
- Department of Medicine, Jining Municipal Government Hospital, Jining, China
| | - Yi Ding
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Yajuan Jiang
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Zengding Wu
- Department of AI and Bioinformatics, Nanjing Chengshi BioTech (TheraRNA) Co., Ltd., Nanjing, China
| | - Xiaoyu Wang
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Huixin Dou
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Yongjian Jia
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| | - Boyan Jiao
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining, China
| |
Collapse
|
41
|
Mattoli S, Schmidt M. Investigational Use of Mesenchymal Stem/Stromal Cells and Their Secretome as Add-On Therapy in Severe Respiratory Virus Infections: Challenges and Perspectives. Adv Ther 2023; 40:2626-2692. [PMID: 37069355 PMCID: PMC10109238 DOI: 10.1007/s12325-023-02507-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/24/2023] [Indexed: 04/19/2023]
Abstract
Serious manifestations of respiratory virus infections such as influenza and coronavirus disease 2019 (COVID-19) are associated with a dysregulated immune response and systemic inflammation. Treating the immunological/inflammatory dysfunction with glucocorticoids, Janus kinase inhibitors, and monoclonal antibodies against the interleukin-6 receptor has significantly reduced the risk of respiratory failure and death in hospitalized patients with severe COVID-19, but the proportion of those requiring invasive mechanical ventilation (IMV) and dying because of respiratory failure remains elevated. Treatment of severe influenza-associated pneumonia and acute respiratory distress syndrome (ARDS) with available immunomodulators and anti-inflammatory compounds is still not recommended. New therapies are therefore needed to reduce the use of IMV and the risk of death in hospitalized patients with rapidly increasing oxygen demand and systemic inflammation who do not respond to the current standard of care. This paper provides a critical assessment of the published clinical trials that have tested the investigational use of intravenously administered allogeneic mesenchymal stem/stromal cells (MSCs) and MSC-derived secretome with putative immunomodulatory/antiinflammatory/regenerative properties as add-on therapy to improve the outcome of these patients. Increased survival rates are reported in 5 of 12 placebo-controlled or open-label comparative trials involving patients with severe and critical COVID-19 and in the only study concerning patients with influenza-associated ARDS. Results are encouraging but inconclusive for the following reasons: small number of patients tested in each trial; differences in concomitant treatments and respiratory support; imbalances between study arms; differences in MSC source, MSC-derived product, dosing and starting time of the investigational therapy; insufficient/inappropriate reporting of clinical data. Solutions are proposed for improving the clinical development plan, with the aim of facilitating regulatory approval of the MSC-based investigational therapy for life-threatening respiratory virus infections in the future. Major issues are the absence of a biomarker predicting responsiveness to MSCs and MSC-derived secretome and the lack of pharmacoeconomic evaluations.
Collapse
Affiliation(s)
- Sabrina Mattoli
- Center of Expertise in Research and Innovation of the International Network for the Advancement of Viable and Applicable Innovations in Life Sciences (InAvail), InAvail at Rosental Nexxt, 4058 Basel, Switzerland
- Avail Biomedical Research Institute, 80539 Munich, Germany
| | - Matthias Schmidt
- Avail Biomedical Research Institute, 80539 Munich, Germany
- Discovery and Translational Research Center, 80539 Munich, Germany
| |
Collapse
|
42
|
Ouyang B, Deng L, Yang F, Shi H, Wang N, Tang W, Huang X, Zhou Y, Yu H, Wei Y, Dong J. Albumin-based formononetin nanomedicines for lung injury and fibrosis therapy via blocking macrophage pyroptosis. Mater Today Bio 2023; 20:100643. [PMID: 37214555 PMCID: PMC10193015 DOI: 10.1016/j.mtbio.2023.100643] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/23/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Pulmonary fibrosis that occurs following lung injury is a progressive and fatal disease since continual damage to lung tissue triggers the dysregulated inflammation response and accompanying abnormal healing process. Pyroptosis of alveolar macrophages has been found to play an essential role in the deterioration of lung injury and fibrosis. However, the lack of inhibitors against this inflammatory cell death in macrophages and the dense stroma pose major barriers to lung injury and fibrosis treatment. Herein, we developed an albumin-based nanoformulation to realize active delivery of formononetin (FMN) to improve the treatment of lung injury and fibrosis. The obtained nanoparticle, FMN@BSA NPs, could efficiently accumulate at the impaired lesion benefiting from the leaky vasculatures and the affinity between albumin and the overexpressed SPARC protein. Through blocking the NLRP3 inflammasome-involved pyroptosis process of macrophages, FMN@BSA NPs remarkably improved lung function and prolonged animal survival in the bleomycin (BLM)-induced lung injury and fibrosis model without noticeable side effects. Meanwhile, we proved FMN as a pyroptosis inhibitor and the corresponding lipid metabolism-related mechanisms through multi-omics analysis. This study first employed an albumin-based nanoparticle to deliver the pyroptosis inhibitor to the impaired lung tissue actively, providing a promising strategy for lung injury and fibrosis treatment.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Fangyong Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| |
Collapse
|
43
|
Caceres CJ, Susta L, Rajao DS. Editorial: Pathogenesis, vaccines, and antivirals against respiratory viruses. Front Cell Infect Microbiol 2023; 13:1202251. [PMID: 37223845 PMCID: PMC10200974 DOI: 10.3389/fcimb.2023.1202251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/02/2023] [Indexed: 05/25/2023] Open
Affiliation(s)
- C. Joaquin Caceres
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Ontario, ON, Canada
| | - Daniela S. Rajao
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
44
|
Liu C, Wu X, Bing X, Qi W, Zhu F, Guo N, Li C, Gao X, Cao X, Zhao M, Xia M. H1N1 influenza virus infection through NRF2-KEAP1-GCLC pathway induces ferroptosis in nasal mucosal epithelial cells. Free Radic Biol Med 2023; 204:226-242. [PMID: 37146698 DOI: 10.1016/j.freeradbiomed.2023.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Influenza A virus can induce nasal inflammation by stimulating the death of nasal mucosa epithelium, however, the mechanism is not clear. In this study, to study the causes and mechanisms of nasal mucosa epithelial cell death caused by Influenza A virus H1N1, we isolated and cultured human nasal epithelial progenitor cells (hNEPCs) and exposed them to H1N1 virus after leading differentiation. Then we performed high-resolution untargeted metabolomics and RNAseq analysis of human nasal epithelial cells (hNECs) infected with H1N1 virus. Surprisingly, H1N1 virus infection caused the differential expression of a large number of ferroptosis related genes and metabolites in hNECs. Furthermore, we have observed a significant reduction in Nrf2/KEAP1 expression, GCLC expression, and abnormal glutaminolysis. By constructing overexpression vector of GCLC and the shRNAs of GCLC and Keap1, we determined the role of NRF2-KEAP1-GCLC signaling pathway in H1N1 virus-induced ferroptosis. In addition, A glutaminase antagonist, JHU-083, also demonstrated that glutaminolysis can regulate the NRF2-KEAP1-GCLC signal pathway and ferroptosis. According to this study, H1N1 virus can induce the ferroptosis of hNECs via the NRF2-KEAP1-GCLC signal pathway and glutaminolysis, leading to nasal mucosal epithelial inflammation. This discovery is expected to provide an attractive therapeutic target for viral-induced nasal inflammation.
Collapse
Affiliation(s)
- Chengcheng Liu
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xinhao Wu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, China
| | - Xin Bing
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, China
| | - Wenwen Qi
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, China
| | - Fangyuan Zhu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Na Guo
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Chengzhilin Li
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaochen Gao
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xue Cao
- Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Miaoqing Zhao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, China; Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, China; NHC Key Laboratory of Otorhinolaryngology, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
45
|
Zhang J, Yang T, Zou M, Wang L, Sai L. The epidemiological features of respiratory tract infection using the multiplex panels detection during COVID-19 pandemic in Shandong province, China. Sci Rep 2023; 13:6319. [PMID: 37072619 PMCID: PMC10112310 DOI: 10.1038/s41598-023-33627-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 04/16/2023] [Indexed: 05/03/2023] Open
Abstract
Respiratory tract infection is one of the most common reasons for both morbidity and mortality worldwide. High attention has been paid to the etiological tracing of respiratory tract infection since the advent of COVID-19. In this study, we aimed to evaluate the epidemiological features of pathogens in respiratory tract infection, especially during COVID-19 pandemic. A total of 7668 patients with respiratory tract infection who admitted to Qilu Hospital of Shandong University from March 2019 to Dec 2021 were retrospectively included. The respiratory tract specimens were detected using a commercial multiplex PCR-based panel assay for common respiratory pathogens including influenza A virus (Flu-A), influenza A virus H1N1 (H1N1), influenza A virus H3N2 (H3N2), influenza B virus (Flu-B), parainfluenza virus (PIV), respiratory syncytial virus (RSV), adenovirus (ADV), Boca virus (Boca), human Rhinovirus (HRV), Metapneumovirus (MPV), Coronavirus (COV), Mycoplasma pneumoniae (MP), and Chlamydia (Ch). The positive rates were compared using a chi-square test. Compared with 2019, the positive rate of pathogen detection during from January 2020 to December 2021 was significantly lower, especially the detection of Flu-A. The positive rate of respiratory pathogen strains was 40.18% during COVID-19 pandemic, and a total of 297 cases (4.69%) of mixed infection with two or more pathogens were detected. There was no statistical difference in the positive rate between male and female patients. However, the positive rates of infection were different among different age groups, with higher incidence of RSV in infancy and toddler group, and MP infection in children and teenager group. While, HRV was the most common pathogen in the adult patients. Moreover, Flu-A and Flu-B were higher in winter, and MP and RSV were higher in spring, autumn and winter. The pathogens such as ADV, BOCA, PIV, and COV were detected without significant seasonal distribution. In conclusion, respiratory pathogen infection rates may vary by age and season, regardless of gender. During the COVID-19 epidemic, blocking transmission routes could help reduce the incidence of respiratory tract infection. The current prevalence of respiratory tract infection pathogens is of great significance for clinical prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong, China
| | - Tao Yang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Mingjin Zou
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong, China.
| | - Lintao Sai
- Department of Infectious Diseases, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
46
|
Geppe NA, Zaplatnikov AL, Kondyurina EG, Chepurnaya MM, Kolosova NG. The Common Cold and Influenza in Children: To Treat or Not to Treat? Microorganisms 2023; 11:microorganisms11040858. [PMID: 37110281 PMCID: PMC10146091 DOI: 10.3390/microorganisms11040858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
The common cold, which is mostly caused by respiratory viruses and clinically represented by the symptoms of acute respiratory viral infections (ARVI) with mainly upper respiratory tract involvement, is an important problem in pediatric practice. Due to the high prevalence, socio-economic burden, and lack of effective prevention measures (except for influenza and, partially, RSV infection), ARVI require strong medical attention. The purpose of this descriptive literature review was to analyze the current practical approaches to the treatment of ARVI to facilitate the choice of therapy in routine practice. This descriptive overview includes information on the causative agents of ARVI. Special attention is paid to the role of interferon gamma as a cytokine with antiviral and immunomodulatory effects on the pathogenesis of ARVI. Modern approaches to the treatment of ARVI, including antiviral, pathogenesis-directed and symptomatic therapy are presented. The emphasis is on the use of antibody-based drugs in the immunoprophylaxis and immunotherapy of ARVI. The data presented in this review allow us to conclude that a modern, balanced and evidence-based approach to the choice of ARVI treatment in children should be used in clinical practice. The published results of clinical trials and systematic reviews with meta-analyses of ARVI in children allow us to conclude that it is possible and expedient to use broad-spectrum antiviral drugs in complex therapy. This approach can provide an adequate response of the child’s immune system to the virus without limiting the clinical possibilities of using only symptomatic therapy.
Collapse
|
47
|
Lim MJ, Lakshminrusimha S, Hedriana H, Albertson T. Pregnancy and Severe ARDS with COVID-19: Epidemiology, Diagnosis, Outcomes and Treatment. Semin Fetal Neonatal Med 2023; 28:101426. [PMID: 36964118 PMCID: PMC9990893 DOI: 10.1016/j.siny.2023.101426] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Pregnancy-related acute respiratory distress syndrome (ARDS) is fast becoming a growing and clinically relevant subgroup of ARDS amidst global outbreaks of various viral respiratory pathogens that include H1N1-influenza, severe acute respiratory syndrome (SARS), middle east respiratory syndrome (MERS), and the most recent COVID-19 pandemic. Pregnancy is a risk factor for severe viral-induced ARDS and commonly associated with poor maternal and fetal outcomes including fetal growth-restriction, preterm birth, and spontaneous abortion. Physiologic changes of pregnancy further compounded by mechanical and immunologic alterations are theorized to impact the development of ARDS from viral pneumonia. The COVID-19 sub-phenotype of ARDS share overlapping molecular features of maternal pathogenicity of pregnancy with respect to immune-dysregulation and endothelial/microvascular injury (i.e., preeclampsia) that may in part explain a trend toward poor maternal and fetal outcomes seen with severe COVID-19 maternal infections. To date, current ARDS diagnostic criteria and treatment management fail to include and consider physiologic adaptations that are unique to maternal physiology of pregnancy and consideration of maternal-fetal interactions. Treatment focused on lung-protective ventilation strategies have been shown to improve clinical outcomes in adults with ARDS but may have adverse maternal-fetal interactions when applied in pregnancy-related ARDS. No specific pharmacotherapy has been identified to improve outcomes in pregnancy with ARDS. Adjunctive therapies aimed at immune-modulation and anti-viral treatment with COVID-19 infection during pregnancy have been reported but data in regard to its efficacy and safety is currently lacking.
Collapse
Affiliation(s)
- Michelle J Lim
- UC Davis School of Medicine, UC Davis Children's Hospital, Department of Pediatrics, Division of Critical Care and Neonatology, Sacramento, CA, USA.
| | - Satyan Lakshminrusimha
- UC Davis School of Medicine, UC Davis Children's Hospital, Department of Pediatrics, Division of Critical Care and Neonatology, Sacramento, CA, USA
| | - Herman Hedriana
- UC Davis School of Medicine, UC Davis Medical Center, Department of Obstetrics and Gynecology, Sacramento, CA, USA
| | - Timothy Albertson
- UC Davis School of Medicine, UC Davis Medical Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Sacramento, CA, USA
| |
Collapse
|
48
|
Liu YN, Zhang YF, Xu Q, Qiu Y, Lu QB, Wang T, Zhang XA, Lin SH, Lv CL, Jiang BG, Li H, Li ZJ, Gao GF, Yang WZ, Hay SI, Wang LP, Fang LQ, Liu W. Infection and co-infection patterns of community-acquired pneumonia in patients of different ages in China from 2009 to 2020: a national surveillance study. THE LANCET MICROBE 2023; 4:e330-e339. [PMID: 37001538 DOI: 10.1016/s2666-5247(23)00031-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Severe community-acquired pneumonia (SCAP) is associated with a substantial number of hospitalisations and deaths worldwide. Infection or co-infection patterns, along with their age dependence and clinical effects are poorly understood. We aimed to explore the causal and epidemiological characteristics by age, to better describe patterns of community-acquired pneumonia (CAP) and their association with severe disease. METHODS National surveillance of CAP was conducted through a network of hospitals in 30 provinces in China from 2009-20 inclusive. Patients with CAP were included if they had evidence of acute respiratory tract, had evidence of pneumonia by chest radiography, diagnosis of pneumonia within 24 h of hospital admission, and resided in the study catchment area. For the enrolled patients with CAP, nasopharyngeal and oral swabs were taken and tested for eight viral pathogens; and blood, urine, or expectorated sputum was tested for six bacterial pathogens. Clinical outcomes, including SCAP, were investigated with respect to age and patterns of infections or co-infections by performing binary logistic regression and multivariate analysis. FINDINGS Between January, 2009, and December, 2020, 18 807 patients with CAP (3771 [20·05%] with SCAP) were enrolled. For both children (aged ≤5 years) and older adults (aged >60 years), a higher overall rate of viral and bacterial infections, as well as viral-bacterial co-infections were seen in patients with SCAP than in patients with non-SCAP. For adults (aged 18-60 years), however, only a higher rate of bacterial-bacterial co-infection was observed. The most frequent pathogens associated with SCAP were respiratory syncytial virus (RSV; 21·30%) and Streptococcus pneumoniae (12·61%) among children, and influenza virus (10·94%) and Pseudomonas aeruginosa (15·37%) among older adults. Positive rates of detection of most of the tested pathogens decreased during 2020 compared with the 2009-19 period, except for RSV, P aeruginosa, and Klebsiella pneumoniae. Multivariate analyses showed SCAP was significantly associated with infection with human adenovirus, human rhinovirus, K pneumoniae, or co-infection of RSV and Haemophilus influenzae or RSV and Staphylococcus aureus in children and adolescents (aged <18 years), and significantly associated with infection with P aeruginosa, K pneumoniae, or S pneumoniae, or co-infection with P aeruginosa and K pneumoniae in adults (aged ≥18 years). INTERPRETATION Both prevalence and infection pattern of respiratory pathogens differed between patients with SCAP and patients with non-SCAP in an age-dependent manner. These findings suggest potential advantages to age-related strategies for vaccine schedules, as well as clinical diagnosis, treatment, and therapy. FUNDING China Mega-Project on Infectious Disease Prevention and The National Natural Science Funds of China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
|
49
|
Zhang M, Zhang X, Pei J, Guo B, Zhang G, Li M, Huang L. Identification of phytochemical compounds of Fagopyrum dibotrys and their targets by metabolomics, network pharmacology and molecular docking studies. Heliyon 2023; 9:e14029. [PMID: 36911881 PMCID: PMC9977108 DOI: 10.1016/j.heliyon.2023.e14029] [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: 07/24/2022] [Revised: 01/30/2023] [Accepted: 02/19/2023] [Indexed: 03/05/2023] Open
Abstract
Acute lung injury (ALI) is a clinically severe lung illness with high incidence rate and mortality. Especially, coronavirus disease 2019 (COVID-19) poses a serious threat to world wide governmental fitness. It has distributed to almost from corner to corner of the universe, and the situation in the prevention and control of COVID-19 remains grave. Traditional Chinese medicine plays a vital role in the precaution and therapy of sicknesses. At present, there is a lack of drugs for treating these diseases, so it is necessary to develop drugs for treating COVID-19 related ALI. Fagopyrum dibotrys (D. Don) Hara is an annual plant of the Polygonaceae family and one of the long-history used traditional medicine in China. In recent years, its rhizomes (medicinal parts) have attracted the attention of scholars at home and abroad due to their significant anti-inflammatory, antibacterial and anticancer activities. It can work on SARS-COV-2 with numerous components, targets, and pathways, and has a certain effect on coronavirus disease 2019 (COVID-19) related acute lung injury (ALI). However, there are few systematic studies on its aerial parts (including stems and leaves) and its potential therapeutic mechanism has not been studied. The phytochemical constituents of rhizome of F. dibotrys were collected using TCMSP database. And metabolites of F. dibotrys' s aerial parts were detected by metabonomics. The phytochemical targets of F. dibotrys were predicted by the PharmMapper website tool. COVID-19 and ALI-related genes were retrieved from GeneCards. Cross targets and active phytochemicals of COVID-19 and ALI related genes in F. dibotrys were enriched by gene ontology (GO) and KEGG by metscape bioinformatics tools. The interplay network entre active phytochemicals and anti COVID-19 and ALI targets was established and broke down using Cytoscape software. Discovery Studio (version 2019) was used to perform molecular docking of crux active plant chemicals with anti COVID-19 and ALI targets. We identified 1136 chemicals from the aerial parts of F. dibotrys, among which 47 were active flavonoids and phenolic chemicals. A total of 61 chemicals were searched from the rhizome of F. dibotrys, and 15 of them were active chemicals. So there are 6 commonly key active chemicals at the aerial parts and the rhizome of F. dibotrys, 89 these phytochemicals's potential targets, and 211 COVID-19 and ALI related genes. GO enrichment bespoken that F. dibotrys might be involved in influencing gene targets contained numerous biological processes, for instance, negative regulation of megakaryocyte differentiation, regulation of DNA metabolic process, which could be put down to its anti COVID-19 associated ALI effects. KEGG pathway indicated that viral carcinogenesis, spliceosome, salmonella infection, coronavirus disease - COVID-19, legionellosis and human immunodeficiency virus 1 infection pathway are the primary pathways obsessed in the anti COVID-19 associated ALI effects of F. dibotrys. Molecular docking confirmed that the 6 critical active phytochemicals of F. dibotrys, such as luteolin, (+) -epicatechin, quercetin, isorhamnetin, (+) -catechin, and (-) -catechin gallate, can combine with kernel therapeutic targets NEDD8, SRPK1, DCUN1D1, and PARP1. In vitro activity experiments showed that the total antioxidant capacity of the aerial parts and rhizomes of F. dibotrys increased with the increase of concentration in a certain range. In addition, as a whole, the antioxidant capacity of the aerial part of F. dibotrys was stronger than that of the rhizome. Our research afford cues for farther exploration of the anti COVID-19 associated ALI chemical compositions and mechanisms of F. dibotrys and afford scientific foundation for progressing modern anti COVID-19 associated ALI drugs based on phytochemicals in F. dibotrys. We also fully developed the medicinal value of F. dibotrys' s aerial parts, which can effectively avoid the waste of resources. Meanwhile, our work provides a new strategy for integrating metabonomics, network pharmacology, and molecular docking techniques which was an efficient way for recognizing effective constituents and mechanisms valid to the pharmacologic actions of traditional Chinese medicine.
Collapse
Key Words
- ARDS, acute respiratory distress syndrome
- BC, BetweennessCentrality
- CC, ClosenessCentrality
- CHM, Chinese herbal medicines
- COVID-19 related ALI, Coronavirus disease 2019 related acute lung injury
- Coronavirus disease 2019 related acute lung injury
- DL, drug-like properties
- Fagopyrum dibotrys
- GO, Gene Ontology
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LC-MS, liquid chromatography-mass spectrometry
- Metabolomics
- Molecular docking
- NC, NeighborhoodConnectivity
- NSCLC, Non-small cell lung carcinoma
- Network pharmacology
- OB, oral bioavailability
- PARP-1, Poly(ADP-ribose)polymerase-1
- PDB, Protein Data Bank database
- PPI network, protein-protein interaction network
- RMSD, Root mean square deviation
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- TCM, traditional Chinese medicine
- TCMSP, traditional Chinese medicine systems pharmacology database and analysis platform
- WTM, widely targeted metabolome
Collapse
Affiliation(s)
- Min Zhang
- A Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- College of Pharmacy, Baotou Medical College, Baotou, 014040, China
- Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, 010020, China
- Inner Mongolia Academy of Chinese and Mongolian Medicine, Hohhot, 010010, China
| | - Xinke Zhang
- A Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baolin Guo
- A Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Guoshuai Zhang
- A Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Minhui Li
- College of Pharmacy, Baotou Medical College, Baotou, 014040, China
- Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, 010020, China
- Inner Mongolia Academy of Chinese and Mongolian Medicine, Hohhot, 010010, China
- Corresponding author. College of Pharmacy, Baotou Medical College, Baotou, 014040, China.
| | - Linfang Huang
- A Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Corresponding author.
| |
Collapse
|
50
|
Application Potential of Luteolin in the Treatment of Viral Pneumonia. J Food Biochem 2023. [DOI: 10.1155/2023/1810503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Aim of the Review. This study aims to summarize the therapeutic effect of luteolin on the pathogenesis of viral pneumonia, explore its absorption and metabolism in the human body, evaluate the possibility of luteolin as a drug to treat viral pneumonia, and provide a reference for future research. Materials and Methods. We searched MEDLINE/PubMed, Web of Science, China National Knowledge Infrastructure, and Google Scholar and collected research on luteolin in the treatment of viral pneumonia and related diseases since 2003. Then, we summarized the efficacy and potential of luteolin in directly inhibiting viral activity, limiting inflammatory storms, reducing pulmonary inflammation, and treating pneumonia complications. Results and Conclusion. Luteolin has the potential to treat viral pneumonia in multiple ways. Luteolin has a direct inhibitory effect on coronavirus, influenza virus, and respiratory syncytial virus. Luteolin can alleviate the inflammatory factor storm induced by multiple factors by inhibiting the function of macrophages or mast cells. Luteolin can reduce pulmonary inflammation, pulmonary edema, or pulmonary fibrosis induced by multiple factors. In addition, viral pneumonia may cause multisystem complications, while luteolin has extensive protective effects on the gastrointestinal system, cardiovascular system, and nervous system. However, due to the first-pass metabolism mediated by phase II enzymes, the bioavailability of oral luteolin is low. The bioavailability of luteolin can be improved, and its potential value can be further developed by changing the dosage form or route of administration.
Collapse
|