1
|
Wu YJ, Feng WW, Wu ZL, Zhang YY, Liu JY, Xu PP. Prim-O-glucosylcimifugin alleviates influenza virus-induced pneumonia in mice by inhibiting the TGF-β1/PI3KCD/MSK2/RELA signalling pathway. Arch Virol 2024; 169:232. [PMID: 39467851 DOI: 10.1007/s00705-024-06158-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/28/2024] [Indexed: 10/30/2024]
Abstract
Prim-O-glucosylcimifugin (POG) is a chromone derived primarily from Saposhnikovia divaricata (Turcz) Schischk and Cimicifuga simplex. Previous research has shown that POG possesses antibacterial, anticancer, anti-inflammatory, antioxidant, anticonvulsant, antipyretic, and analgesic properties. However, the specific impact of POG on influenza-virus-induced pneumonia is not well understood. In this study, we investigated the protective effects and underlying mechanisms of POG in pneumonia caused by influenza A virus (IAV). In vitro, POG was found to have a protective effect against infections caused by the respiratory viruses respiratory syncytial virus (RSV), human coronavirus OC43, and influenza A virus. POG inhibited A/FM/1/1947(H1N1) infection with an EC50 ranging from 3.01 to 10.43 in vitro. Intraperitoneal infection of mice with POG at a dose of 5 or 10 mg/kg resulted in a reduction in IAV-induced pneumonia, as evidenced by decreased pulmonary edema, improved lung histopathology, and reduced inflammatory cell accumulation. At the higher dose (10 mg/kg), POG treatment significantly increased survival rates, decreased viral titres in the lungs, improved lung histology, and reduced lung inflammation in IAV-infected mice. POG also effectively alleviated pulmonary fibrosis by reducing the levels of fibrotic markers (hydroxyproline [Hyp] and transforming growth factor β1 [TGF-β1]) and suppressing the expression of alpha smooth muscle actin (α-SMA), p focal adhesion kinase (p-FAK), and TGF-β1 in lung tissues. In addition, POG inhibited the expression of the RELA proto-oncogene (RELA), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta (PIK3CD), and mitogen- and stress-activated protein kinase 2 (MSK2) in lung tissues. These results indicate that POG may have a protective effect against IAV-induced pneumonia by downregulating the TGF-β1/PI3KCD/MSK2/RELA signalling pathway in the lungs.
Collapse
Affiliation(s)
- Yu-Jia Wu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist, Guangzhou, Guangdong, 510405, People's Republic of China
| | - Wen-Wen Feng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist, Guangzhou, Guangdong, 510405, People's Republic of China
| | - Zhen-Lin Wu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist, Guangzhou, Guangdong, 510405, People's Republic of China
| | - Yue-Yao Zhang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist, Guangzhou, Guangdong, 510405, People's Republic of China
| | - Jin-Yuan Liu
- Basic Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Pei-Ping Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist, Guangzhou, Guangdong, 510405, People's Republic of China.
| |
Collapse
|
2
|
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
|
3
|
Yang S, Wang L, Pan X, Liang Y, Zhang Y, Li J, Zhou B. 5-Methoxyflavone-induced AMPKα activation inhibits NF-κB and P38 MAPK signaling to attenuate influenza A virus-mediated inflammation and lung injury in vitro and in vivo. Cell Mol Biol Lett 2022; 27:82. [PMID: 36180831 PMCID: PMC9524045 DOI: 10.1186/s11658-022-00381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/02/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza-related acute lung injury (ALI) is a life-threatening condition that results mostly from uncontrolled replication of influenza virus (IV) and severe proinflammatory responses. The methoxy flavonoid compound 5-methoxyflavone (5-MF) is believed to have superior biological activity in the treatment of cancer. However, the effects and underlying mechanism of 5-MF on IV-mediated ALI are still unclear. Here, we showed that 5-MF significantly improved the survival of mice with lethal IV infection and ameliorated IV-mediated lung edema, lung histological changes, and inflammatory cell lung recruitment. We found that 5-MF has antiviral activity against influenza A virus (IAV), which was probably associated with increased expression of radical S-adenosyl methionine domain containing 2 (RSAD2) and suppression of endosomal acidification. Moreover, IV-infected A549 cells with 5-MF treatment markedly reduced proinflammatory mediator expression (IL-6, CXCL8, TNF-α, CXCL10, CCL2, CCL3, CCL4, GM-CSF, COX-2, and PGE2) and prevented P-IKBα, P-P65, and P-P38 activation. Interestingly, we demonstrated that 5-MF treatment could trigger activation of AMP-activated protein kinase (AMPK)α in IV-infected A549 cells, as evidenced by activation of the AMPKα downstream molecule P53. Importantly, the addition of AMPKα blocker compound C dramatically abolished 5-MF-mediated increased levels of RSAD2, the inhibitory effects on H1N1 virus-elicited endosomal acidification, and the suppression expression of proinflammatory mediators (IL-6, TNF-α, CXCL10, COX-2 and PGE2), as well as the inactivation of P-IKBα, P-P65, and P-P38 MAPK signaling pathways. Furthermore, inhibition of AMPKα abrogated the protective effects of 5-MF on H1N1 virus-mediated lung injury and excessive inflammation in vivo. Taken together, these results indicate that 5-MF alleviated IV-mediated ALI and suppressed excessive inflammatory responses through activation of AMPKα signaling.
Collapse
Affiliation(s)
- Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | | | | | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China. .,Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou, 525200, China.
| |
Collapse
|
4
|
Wei X, Lan Y, Nong Z, Li C, Feng Z, Mei X, Zhai Y, Zou M. Ursolic acid represses influenza A virus-triggered inflammation and oxidative stress in A549 cells by modulating the miR-34c-5p/TLR5 axis. Cytokine 2022; 157:155947. [PMID: 35780710 DOI: 10.1016/j.cyto.2022.155947] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ursolic acid (UA) is a pentacyclic triterpenoid compound with a wide range of anti-tumor, anti-inflammatory, hypotensive and other pharmacological effects. Here, the biological roles and regulatory mechanisms of UA in influenza A virus (IAV)-treated A549 cells were investigated. METHOD The cytotoxic impacts of UA on A549 cells with or without IAV treatment were determined using MTT and LDH assays. The inflammatory responses and oxidative stress of IAV-treated A549 cells were measured by RT-qPCR, ELISA, DCFH-DA probe, and colorimetric assays. A dual luciferase assay was carried out to validate the molecular interaction between miR-34c-5p and TLR5. Promoter methylation was detected by MSP experiment. Methylation-related proteins were quantified by western blot. Virus replication was assessed by TCID50 and western blot assays. RESULTS UA significantly ameliorated IAV-triggered cell injury and inflammatory response, virus replication and oxidative stress by elevating cell viability, ROS level and the activities of SOD and GSH-Px but reducing the LDH, MDA, and TCID50 values and the expression of virus-related proteins (NP) and cytokines (TNF-α, IL-1β, IL-6, and IL-18). Moreover, UA promoted miR-34c-5p expression by repressing DNMTs-mediated methylation. TLR5 was verified to be a direct target of miR-34c-5p and could be downregulated by UA. Rescue experiments revealed that silencing miR-34c-5p diminished the regulatory roles of UA in IAV-treated A549 cells. CONCLUSION Our data elucidated that UA attenuated IAV-triggered inflammatory responses and oxidative stress in A549 cells by regulating the miR-34c-5p/TLR5 axis, suggesting that UA plays a protective role in IAV-induced pneumonia.
Collapse
Affiliation(s)
- Xing Wei
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China
| | - Yuying Lan
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China
| | - Zhifei Nong
- Department of Pediatrics, Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530022, Guangxi Province, China
| | - Chongjin Li
- Department of Pediatrics, Maoming Hospital of Traditional Chinese Medicine, Maoming 525000, Guangdong Province, China
| | - Zhiqiong Feng
- Department of Pediatrics, Jinshazhou Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Xiaoping Mei
- Department of Pediatrics, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China
| | - Yang Zhai
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Nanning 530200, Guangxi Province, China; Department of International Medical, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China
| | - Min Zou
- Department of Pediatrics, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China.
| |
Collapse
|
5
|
Ikonomou L, Magnusson M, Dries R, Herzog EL, Hynds RE, Borok Z, Park JA, Skolasinski S, Burgess JK, Turner L, Mojarad SM, Mahoney JE, Lynch T, Lehmann M, Thannickal VJ, Hook JL, Vaughan AE, Hoffman ET, Weiss DJ, Ryan AL. Stem cells, cell therapies, and bioengineering in lung biology and disease 2021. Am J Physiol Lung Cell Mol Physiol 2022; 323:L341-L354. [PMID: 35762622 PMCID: PMC9484991 DOI: 10.1152/ajplung.00113.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
The 9th biennial conference titled "Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases" was hosted virtually, due to the ongoing COVID-19 pandemic, in collaboration with the University of Vermont Larner College of Medicine, the National Heart, Lung, and Blood Institute, the Alpha-1 Foundation, the Cystic Fibrosis Foundation, and the International Society for Cell & Gene Therapy. The event was held from July 12th through 15th, 2021 with a pre-conference workshop held on July 9th. As in previous years, the objectives remained to review and discuss the status of active research areas involving stem cells (SCs), cellular therapeutics, and bioengineering as they relate to the human lung. Topics included 1) technological advancements in the in situ analysis of lung tissues, 2) new insights into stem cell signaling and plasticity in lung remodeling and regeneration, 3) the impact of extracellular matrix in stem cell regulation and airway engineering in lung regeneration, 4) differentiating and delivering stem cell therapeutics to the lung, 5) regeneration in response to viral infection, and 6) ethical development of cell-based treatments for lung diseases. This selection of topics represents some of the most dynamic and current research areas in lung biology. The virtual workshop included active discussion on state-of-the-art methods relating to the core features of the 2021 conference, including in situ proteomics, lung-on-chip, induced pluripotent stem cell (iPSC)-airway differentiation, and light sheet microscopy. The conference concluded with an open discussion to suggest funding priorities and recommendations for future research directions in basic and translational lung biology.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, State University of New York, Buffalo, New York
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ruben Dries
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Erica L Herzog
- Yale Interstitial Lung Disease Center of Excellence, Pulmonary and Critical Care Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Robert E Hynds
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Janette K Burgess
- Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Leigh Turner
- Department of Health, Society, and Behavior, University of California, Irvine Program In Public Health, Irvine, California
| | - Sarah M Mojarad
- Engineering in Society Program, Viterbi School of Engineering, University of Southern California, Los Angeles, California
| | | | - Thomas Lynch
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Mareike Lehmann
- Institute of Lung Health and Immunity, Comprehensive Pneumology Center Munich, Helmholtz Zentrum München, Munich, Germany
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jamie L Hook
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evan T Hoffman
- Department of Medicine, University of Vermont, Burlington, Vermont
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont
| | - Amy L Ryan
- Hastings Center for Pulmonary Research, Department of Medicine, University of Southern California, Los Angeles, California
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
6
|
Jang WY, Lee HP, Kim SA, Huang L, Yoon JH, Shin CY, Mitra A, Kim HG, Cho JY. Angiopteris cochinchinensis de Vriese Ameliorates LPS-Induced Acute Lung Injury via Src Inhibition. PLANTS 2022; 11:plants11101306. [PMID: 35631731 PMCID: PMC9143704 DOI: 10.3390/plants11101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
Growing demand for treatment options against acute lung injury (ALI) emphasizes studies on plant extracts harboring anti-inflammatory effects. According to GC-MS analysis, Angiopteris cochinchinensis de Vriese consists of various flavonoids with anti-inflammatory activities. Thus, in this study, the anti-inflammatory effects of an extract of Angiopteris cochinchinensis de Vriese (Ac-EE) were assessed using RAW264.6 murine macrophages and a lipopolysaccharide (LPS)-induced ALI model. Ac-EE reduced the nitric oxide production in murine macrophages increased by LPS induction. Moreover, protective effects of Ac-EE on lung tissue were demonstrated by shrinkage of edema and lung injury. Reduced neutrophil infiltration and formation of hyaline membranes were also detected in lung tissues after H&E staining. Semiquantitative RT-PCR, quantitative real-time PCR, and ELISA showed that Ac-EE inhibits the production of proinflammatory mediators, including iNOS and COX-2, and cytokines, such as TNF-α, IL-1β, and IL-6. An Ac-EE-mediated anti-inflammatory response was derived from inhibiting the NF-κB signaling pathway, which was evaluated by luciferase reporter assay and Western blotting analysis. A cellular thermal shift assay revealed that the prime target of Ac-EE in alleviating inflammation was Src. With its direct binding with Src, Angiopteris cochinchinensis de Vriese significantly mitigates lung injury, showing possibilities of its potential as an effective botanical drug.
Collapse
Affiliation(s)
- Won Young Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Seung A Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Lei Huang
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Ji Hye Yoon
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Chae Yun Shin
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Ankita Mitra
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
- Correspondence: (H.G.K.); (J.Y.C.); Tel.: +82-31-290-7878 (H.G.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
- Correspondence: (H.G.K.); (J.Y.C.); Tel.: +82-31-290-7878 (H.G.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
7
|
Spencer JA, Shutt DP, Moser SK, Clegg H, Wearing HJ, Mukundan H, Manore CA. Distinguishing viruses responsible for influenza-like illness. J Theor Biol 2022; 545:111145. [PMID: 35490763 DOI: 10.1016/j.jtbi.2022.111145] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022]
Abstract
The many respiratory viruses that cause influenza-like illness (ILI) are reported and tracked as one entity, defined by the CDC as a group of symptoms that include a fever of 100 degrees Fahrenheit, a cough, and/or a sore throat. In the United States alone, ILI impacts 9-49 million people every year. While tracking ILI as a single clinical syndrome is informative in many respects, the underlying viruses differ in parameters and outbreak properties. Most existing models treat either a single respiratory virus or ILI as a whole. However, there is a need for models capable of comparing several individual viruses that cause respiratory illness, including ILI. To address this need, here we present a flexible model and simulations of epidemics for influenza, RSV, rhinovirus, seasonal coronavirus, adenovirus, and SARS/MERS, parameterized by a systematic literature review and accompanied by a global sensitivity analysis. We find that for these biological causes of ILI, their parameter values, timing, prevalence, and proportional contributions differ substantially. These results demonstrate that distinguishing the viruses that cause ILI will be an important aspect of future work on diagnostics, mitigation, modeling, and preparation for future pandemics.
Collapse
Affiliation(s)
- Julie A Spencer
- A-1 Information Systems and Modeling, Los Alamos National Laboratory, NM87545, USA.
| | - Deborah P Shutt
- A-1 Information Systems and Modeling, Los Alamos National Laboratory, NM87545, USA
| | - S Kane Moser
- B-10 Biosecurity and Public Health, Los Alamos National Laboratory, NM87545, USA
| | - Hannah Clegg
- A-1 Information Systems and Modeling, Los Alamos National Laboratory, NM87545, USA
| | - Helen J Wearing
- Department of Biology, University of New Mexico, NM87131, USA; Department of Mathematics and Statistics, University of New Mexico, NM87102, USA
| | - Harshini Mukundan
- C-PCS Physical Chemistry and Applied Spectroscopy, Los Alamos National Laboratory, NM87545, USA
| | - Carrie A Manore
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, NM87545, USA
| |
Collapse
|
8
|
Carvallo FR, Stevenson VB. Interstitial pneumonia and diffuse alveolar damage in domestic animals. Vet Pathol 2022; 59:586-601. [DOI: 10.1177/03009858221082228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Classification of pneumonia in animals has been controversial, and the most problematic pattern is interstitial pneumonia. This is true from the gross and histologic perspectives, and also from a mechanistic point of view. Multiple infectious and noninfectious diseases are associated with interstitial pneumonia, all of them converging in the release of inflammatory mediators that generate local damage and attract inflammatory cells that inevitably trigger a second wave of damage. Diffuse alveolar damage is one of the more frequently identified histologic types of interstitial pneumonia and involves injury to alveolar epithelial and/or endothelial cells, with 3 distinct stages. The first is the “exudative” stage, with alveolar edema and hyaline membranes. The second is the “proliferative” stage, with hyperplasia and reactive atypia of type II pneumocytes, infiltration of lymphocytes, plasma cells, and macrophages in the interstitium and early proliferation of fibroblasts. These stages are reversible and often nonfatal. If damage persists, there is a third “fibrosing” stage, characterized by fibrosis of the interstitium due to proliferation of fibroblasts/myofibroblasts, persistence of type II pneumocytes, segments of squamous metaplasia of alveolar epithelium, plus inflammation. Understanding the lesion patterns associated with interstitial pneumonias, their causes, and the underlying mechanisms aid in accurate diagnosis that involves an interdisciplinary collaborative approach involving pathologists, clinicians, and radiologists.
Collapse
Affiliation(s)
- Francisco R. Carvallo
- Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA
- Virginia Department of Agriculture and Consumer Services, Harrisonburg, VA
| | | |
Collapse
|
9
|
Rubey KM, Brenner JS. Nanomedicine to fight infectious disease. Adv Drug Deliv Rev 2021; 179:113996. [PMID: 34634395 PMCID: PMC8665093 DOI: 10.1016/j.addr.2021.113996] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/09/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022]
Abstract
The ubiquity and potency of antibiotics may give the false impression that infection is a solved problem. Unfortunately, even bacterial infections, the target of antibiotics, remain a major cause of illness and death. Several major unmet needs persist: biofilms, such as those on implanted hardware, largely resist antibiotics; the inflammatory host response to infection often produces more damage than the infection itself; and systemic antibiotics often decimate the gut microbiome, which can predispose to additional infections and even predispose to non-infectious diseases. Additionally, there is an increasing threat from multi-drug resistant microorganisms, though market forces may continue to inhibit innovation in this realm. These numerous unmet infection-related needs provide attractive goals for innovation of targeted drug delivery technologies, especially those of nanomedicine. Here we review several of those innovations in pre-clinical development, the two such therapies which have made it to clinical use, and the opportunities for further technology development for treating infections.
Collapse
Affiliation(s)
- Kathryn M Rubey
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Gotts JE, Maishan M, Chun L, Fang X, Han C, Chiueh V, Khakoo AY, Lee T, Stolina M, Matthay MA. Delayed angiopoietin-2 blockade reduces influenza-induced lung injury and improves survival in mice. Physiol Rep 2021; 9:e15081. [PMID: 34755490 PMCID: PMC8578883 DOI: 10.14814/phy2.15081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/24/2022] Open
Abstract
Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.
Collapse
Affiliation(s)
- Jeffrey E. Gotts
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Mazharul Maishan
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lauren Chun
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Xiaohui Fang
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Chun‐Ya Han
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Venice Chiueh
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Aarif Y. Khakoo
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - TaeWeon Lee
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Marina Stolina
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Michael A. Matthay
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
11
|
Yu W, Zeng M, Xu P, Liu J, Wang H. Effect of paeoniflorin on acute lung injury induced by influenza A virus in mice. Evidences of its mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153724. [PMID: 34509953 DOI: 10.1016/j.phymed.2021.153724] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Influenza often leads to acute lung injury (ALI). Few therapeutics options such as vaccines and other antiviral drugs are available. Paeoniflorin is a monoterpene glucoside isolated from the roots of Paeonia lactiflora Pall. that has showed good anti-inflammatory and anti-fibrotic effects. However, it is not known whether paeoniflorin has an effect on influenza virus-induced ALI. PURPOSE To investigative the protective effect and potential mechanism of paeoniflorin on ALI induced by influenza A virus (IAV). STUDY DESIGN AND METHODS The anti-influenza activity of paeoniflorin in vitro was investigated. Influenza virus A/FM/1/47 was intranasally infected in mice to induce ALI, and paeoniflorin (50 and 100 mg/kg) was given orally to mice during 5 days, beginning 2 h after infection. On day 6 post-infection, body and lung weights, histology and survival were observed, and the lungs were examined for viral load, cytokine and cellular pathway protein expression. RESULTS Results showed that paeoniflorin (50 and 100 mg/kg) reduced IAV-induced ALI. It reduces pulmonary oedema and improves histopathological changes in the lung, and also diminishes the accumulation of inflammatory cells in the lung. It was shown that paeoniflorin (50 and 100 mg/kg) alleviated IAV-induced ALI, as evidenced by improved survival in infected mice (40% and 50%, respectively), reduced viral titer in lung tissue, improved histological changes, and reduced lung inflammation. Paeoniflorin also improves pulmonary fibrosis by reducing the levels of pulmonary fibrotic markers (collagen type IV, alpha-smooth muscle actin, hyaluronic acid, laminin, and procollagen type III) and downregulating the expression levels of type I collagen (Col I) and type III collagen (Col III) in the lung tissues. Additionally, paeoniflorin inhibits the expression of αvβ3, TGF-β1, Smad2, NF-κB, and p38MAPK in the lung tissues. CONCLUSION The results showed that paeoniflorin (50 and 100 mg/kg) protected against IAV-induced ALI, and the underlying mechanism may be related to the reduction of pro-inflammatory cytokine production and lung collagen deposition through down-regulation of activation of αvβ3/TGF-β1 pathway in lung tissue.
Collapse
Affiliation(s)
- Wendi Yu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist., Guangzhou, Guangdong 510405, PR China
| | - Maosen Zeng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist., Guangzhou, Guangdong 510405, PR China
| | - Peiping Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist., Guangzhou, Guangdong 510405, PR China.
| | - Jinyuan Liu
- Basic Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| | - Huixian Wang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Rd., San Yuanli St., Bai Yun Dist., Guangzhou, Guangdong 510405, PR China
| |
Collapse
|
12
|
Aboushanab SA, El-Far AH, Narala VR, Ragab RF, Kovaleva EG. Potential therapeutic interventions of plant-derived isoflavones against acute lung injury. Int Immunopharmacol 2021; 101:108204. [PMID: 34619497 DOI: 10.1016/j.intimp.2021.108204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 09/25/2021] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a life-threatening syndrome that possibly leads to high morbidity and mortality as no therapy exists. Several natural ingredients with negligible adverse effects have recently been investigated to possibly inhibit the inflammatory pathways associated with ALI at the molecular level. Isoflavones, as phytoestrogenic compounds, are naturally occurring bioactive compounds that represent the most abundant category of plant polyphenols (Leguminosae family). A broad range of therapeutic activities of isoflavones, including antioxidants, chemopreventive, anti-inflammatory, antiallergic and antibacterial potentials, have been extensively documented in the literature. Our review exclusively focuses on the possible anti-inflammatory, antioxidant role of botanicals'-derived isoflavones against ALI and their immunomodulatory effect in experimentally induced ALI. Despite the limited scope covering their molecular mechanisms, isoflavones substantially contributed to protecting from ALI via inhibiting toll-like receptor 4 (TLR4)/Myd88/NF-κB pathway and subsequent cytokines, chemokines, and adherent proteins. Nonetheless, future research is suggested to fill the gap in elucidating the protective roles of isoflavones to alleviate ALI concerning antioxidant potentials, inhibition of the inflammatory pathways, and associated molecular mechanisms.
Collapse
Affiliation(s)
- Saied A Aboushanab
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt; Scientific Chair of Yousef Abdullatif Jameel of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | | | - Rokia F Ragab
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Elena G Kovaleva
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| |
Collapse
|
13
|
Garg SS, Sharma A, Gupta J. Immunomodulation and immunotherapeutics of COVID-19. Clin Immunol 2021; 231:108842. [PMID: 34461289 PMCID: PMC8393504 DOI: 10.1016/j.clim.2021.108842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 causes coronavirus disease 2019, a pandemic which was originated from Wuhan city of China. The pandemic has affected millions of people worldwide. The pathogenesis of SARS-CoV-2 is characterized by a cytokine storm in the blood (cytokinemia) and tissues, especially the lungs. One of the major repercussions of this inflammatory process is the endothelial injury-causing intestinal bleeding, coagulopathy, and thromboembolism which result in various sudden and unexpected post-COVID complications including kidney failure, myocardial infarction, or multiorgan failure. In this review, we have summarized the immune responses, biochemical changes, and inflammatory responses in the human body after infection with the SARS-CoV-2 virus. The increased amount of inflammatory cytokines, chemokines, and involvement of complement proteins in inflammatory reaction increase the risk of occurrence of disease.
Collapse
Affiliation(s)
- Sourbh Suren Garg
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Atulika Sharma
- Department of Chemistry, School of Chemical Engineering and Physical Science, Lovely Professional University, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India.
| |
Collapse
|
14
|
Al-Gabri NA, Saghir SAM, Al-Hashedi SA, El-Far AH, Khafaga AF, Swelum AA, Al-Wajeeh AS, Mousa SA, Abd El-Hack ME, Naiel MAE, El-Tarabily KA. Therapeutic Potential of Thymoquinone and Its Nanoformulations in Pulmonary Injury: A Comprehensive Review. Int J Nanomedicine 2021; 16:5117-5131. [PMID: 34349511 PMCID: PMC8326280 DOI: 10.2147/ijn.s314321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
As a crucial organ, the lung is exposed to various harmful agents that may induce inflammation and oxidative stress, which may cause chronic or acute lung injury. Nigella sativa, also known as black seed, has been widely used to treat various diseases and is one of the most extensively researched medicinal plants. Thymoquinone (TQ) is the main component of black seed volatile oil and has been proven to have antioxidant, anti-inflammatory, and antineoplastic properties. The potential therapeutic properties of TQ against various pulmonary disorders have been studied in both in vitro and in vivo studies. Furthermore, the application of nanotechnology may increase drug solubility, cellular absorption, drug release (sustained or control), and drug delivery to lung tissue target sites. As a result, fabricating TQ as nanoparticles (NPs) is a potential therapeutic approach against a variety of lung diseases. In this current review, we summarize recent findings on the efficacy of TQ and its nanotypes in lung disorders caused by immunocompromised conditions such as cancer, diabetes, gastric ulcers, and other neurodegenerative diseases. It is concluded that TQ nanoparticles with anti-inflammatory, antioxidant, antiasthma, and antitumor activity may be safely applied to treat lung disorders. However, more research is required before TQ nanoparticles can be used as pharmaceutical preparations in human studies.
Collapse
Affiliation(s)
- Naif A Al-Gabri
- Department of Pathology, Faculty of Veterinary Medicine, Thamar University, Dhamar, Yemen.,Laboratory of Regional Djibouti Livestock Quarantine, Abu Yasar international Est. 1999, Arta, Djibouti
| | - Sultan A M Saghir
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Medical Sciences, AlHussein Bin Talal University, Ma'an, 71111, Jordan
| | | | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Ayman A Swelum
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | | | - Shaker A Mousa
- Department of Pharmaceutical Sciences, the Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, 12144, USA
| | - Mohamed E Abd El-Hack
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Mohammed A E Naiel
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, 15551, United Arab Emirates.,Biosecurity and One Health Research Centre, Harry Butler Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
| |
Collapse
|
15
|
Chen X, Hu Mt W, Yang M, Ling J, Zhang Y, Deng L, Li J, Lundkvist Å, Lindahl JF, Xiong Y. Risk factors for the delayed viral clearance in COVID-19 patients. J Clin Hypertens (Greenwich) 2021; 23:1483-1489. [PMID: 34171164 PMCID: PMC8420571 DOI: 10.1111/jch.14308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/13/2021] [Accepted: 05/22/2021] [Indexed: 12/19/2022]
Abstract
Comorbidities are important for the disease outcome of COVID‐19, however, which underlying diseases that contribute the most to aggravate the conditions of COVID‐19 patients are still unclear. Viral clearance is the most important laboratory test for defining the recovery of COVID‐19 infections. To better understand which underlying diseases that are risk factors for delaying the viral clearance, we retrospectively analyzed 161 COVID‐19 clinical cases in the Zhongnan Hospital of Wuhan University, Wuhan, China between January 5 and March 13, 2020. The demographic, clinical and laboratory data, as well as patient treatment records were collected. Univariable and multivariable analysis were performed to explore the association between delayed viral clearance and other factors by using logistic regression. Survival analyses by Kaplan‐Meier and Cox regression modeling were employed to identify factors negatively influencing the viral clearance negatively. We found that hypertension and intravenous immunoglobulin adversely affected the time of viral RNA shedding. Hypertension was the most important risk factor to delay the SARS‐CoV‐2 virus clearance, however, the use of Angiotensin‐Converting Enzyme Inhibitors(ACEI)/Angiotensin Receptor Blockers(ARB) did not shorten the time for virus clearance in these hypertensive patients’ virus clearance. We conclude that patients having hypertension and intravenous immunoglobulin may delay the viral clearance in COVID‐19 patients.
Collapse
Affiliation(s)
- Xiaoping Chen
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenjia Hu Mt
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miao Yang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaxin Ling
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Yongxi Zhang
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liping Deng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, University of Uppsala, Uppsala, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Johanna F Lindahl
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, University of Uppsala, Uppsala, Sweden.,Department of Biosciences, International Livestock Research Institute, Hanoi, Vietnam.,Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Yong Xiong
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Chu C, Zeng S, Hasan AA, Hocher C, Krämer BK, Hocher B. Comparison of infection risks and clinical outcomes in patients with and without SARS-CoV-2 lung infection under renin-angiotensin-aldosterone system blockade: Systematic review and meta-analysis. Br J Clin Pharmacol 2021; 87:2475-2492. [PMID: 33217033 PMCID: PMC7753617 DOI: 10.1111/bcp.14660] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/28/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
AIMS Angiotensin-converting enzyme-2 (ACE2) is the receptor for SARS-CoV-2. Animal studies suggest that renin-angiotensin-aldosterone system (RAAS) blockers might increase the expression of ACE2 and potentially increase the risk of SARS-CoV-2 infection. METHODS AND RESULTS The effect of ACE inhibitor (ACEI) treatment on the pneumonia incidence in non-COVID-19 patients (25 studies, 330 780 patients) was associated with a 26% reduction of pneumonia risk (odds ratio [OR]: 0.74, P < .001). Pneumonia-related death cases in ACEI-treated non-COVID-19 patients were reduced by 27% (OR: 0.73, P = .004). However, angiotensin II receptor blockers (ARB) treatment (10 studies, 275 621 non-COVID-19 patients) did not alter pneumonia risk in patients. Pneumonia-related death cases in ARB-treated non-COVID-19 patients was analysed only in 1 study and was significantly reduced (OR, 0.47; 95% confidence interval, 0.30 to 0.72). Results from 11 studies (8.4 million patients) showed that the risk of getting infected with the SARS-CoV-2 virus was reduced by 13% (OR: 0.87, P = .014) in patients treated with ACEI, whereas analysis from 10 studies (8.4 million patients) treated with ARBs showed no effect (OR, 0.92, P = .354). Results from 34 studies in 67 644 COVID-19 patients showed that RAAS blockade reduces all-cause mortality by 24% (OR = 0.76, P = .04). CONCLUSION ACEIs reduce the risk of getting infected with the SARS-CoV-2 virus. Blocking the RAAS may decrease all-cause mortality in COVID-19 patients. ACEIs also reduce the risk of non-COVID pneumonia. All-cause mortality due to non-COVID pneumonia is reduced by ACEI and potentially by ARBs.
Collapse
Affiliation(s)
- Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
- Department of NephrologyCharité‐Universitätsmedizin Berlin, Campus MitteBerlinGermany
| | - Shufei Zeng
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
- Department of NephrologyCharité‐Universitätsmedizin Berlin, Campus MitteBerlinGermany
| | - Ahmed A. Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
- Department of Nutritional Toxicology, Institute of Nutritional ScienceUniversity of PotsdamNuthetalGermany
- Department of Biochemistry, Faculty of PharmacyZagazig UniversityEgypt
| | - Carl‐Friedrich Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
- European Center of Angioscience, Medical Faculty MannheimUniversity of HeidelbergGermany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology)University Medical Centre Mannheim, University of HeidelbergGermany
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of MedicineHunan Normal UniversityChangshaChina
- Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- IMD Institut für Medizinische Diagnostik Berlin‐Potsdam GbRBerlinGermany
| |
Collapse
|
17
|
Ali J, Khan FR, Ullah R, Hassan Z, Khattak S, Lakhta G, Zad Gul N, Ullah R. Cardiac Troponin I Levels in Hospitalized COVID-19 Patients as a Predictor of Severity and Outcome: A Retrospective Cohort Study. Cureus 2021; 13:e14061. [PMID: 33898144 PMCID: PMC8061753 DOI: 10.7759/cureus.14061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Introduction The COVID-19 (coronavirus disease) has affected millions of people, wreaking havoc worldwide. World Health Organization (WHO) labelled this disease as a serious threat to public health since its rapid spread from Wuhan, China. The respiratory manifestations of COVID-19 are common, but myocardium involvement causing myocardial injury and rise in cardiac markers is much less discussed. Materials and methods We conducted this retrospective cohort study from 1st April 2020 to 1st October 2020. Data was collected from the Hospital Management and Information System (HMIS) based on inclusion criteria. We used the Cox proportional hazard regression model for survival analysis, estimated the probability curves of survival using the Kaplan-Meier method, and contrasted it with the log-rank test. Results Among the 466 patients, 280 (69%) were male; the rest were female. The majority were both hypertensive and diabetic, and one-third had a myocardial injury on arrival. The most frequent symptoms in more than half of the patients (51.90%) included a combination of fever, dry cough, and shortness of breath. Out of 466 patients, 266 patients were discharged, and 200 did not survive. In our study, 168 (36.05%) patients had a cardiac injury; among them, 38 (22.61%) were in the discharge group, and the remaining 130 (77.39%) patients were in the nonsurvivor group. Our study results showed that the mortality rate was higher in patients with high cardiac troponin I (cTnI) levels (hazard ratio [HR] 3.61) on admission. Conclusion Our result concluded that measuring cTnI levels on presentation could help predict the severity and outcome in COVID-19 patients. It will allow physicians to triage patients and decrease mortality.
Collapse
Affiliation(s)
- Jabar Ali
- Cardiology/Interventional Cardiology, Lady Reading Hospital, Peshawar, PAK
| | - Fahad R Khan
- Cardiology, Lady Reading Hospital, Peshawar, PAK
| | - Rizwan Ullah
- Cardiology, Lady Reading Hospital, Peshawar, PAK
| | - Zair Hassan
- Cardiology, Lady Reading Hospital, Peshawar, PAK
| | | | - Gul Lakhta
- Gynecology and Obstetrics, Lady Reading Hospital, Peshawar, PAK
| | - Nooh Zad Gul
- Cardiology, Lady Reading Hospital, Peshawar, PAK
| | - Rahman Ullah
- Cardiology, Lady Reading Hospital, Peshawar, PAK
| |
Collapse
|
18
|
Fosse JH, Haraldsen G, Falk K, Edelmann R. Endothelial Cells in Emerging Viral Infections. Front Cardiovasc Med 2021; 8:619690. [PMID: 33718448 PMCID: PMC7943456 DOI: 10.3389/fcvm.2021.619690] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
There are several reasons to consider the role of endothelial cells in COVID-19 and other emerging viral infections. First, severe cases of COVID-19 show a common breakdown of central vascular functions. Second, SARS-CoV-2 replicates in endothelial cells. Third, prior deterioration of vascular function exacerbates disease, as the most common comorbidities of COVID-19 (obesity, hypertension, and diabetes) are all associated with endothelial dysfunction. Importantly, SARS-CoV-2's ability to infect endothelium is shared by many emerging viruses, including henipaviruses, hantavirus, and highly pathogenic avian influenza virus, all specifically targeting endothelial cells. The ability to infect endothelium appears to support generalised dissemination of infection and facilitate the access to certain tissues. The disturbed vascular function observed in severe COVID-19 is also a prominent feature of many other life-threatening viral diseases, underscoring the need to understand how viruses modulate endothelial function. We here review the role of vascular endothelial cells in emerging viral infections, starting with a summary of endothelial cells as key mediators and regulators of vascular and immune responses in health and infection. Next, we discuss endotheliotropism as a possible virulence factor and detail features that regulate viruses' ability to attach to and enter endothelial cells. We move on to review how endothelial cells detect invading viruses and respond to infection, with particular focus on pathways that may influence vascular function and the host immune system. Finally, we discuss how endothelial cell function can be dysregulated in viral disease, either by viral components or as bystander victims of overshooting or detrimental inflammatory and immune responses. Many aspects of how viruses interact with the endothelium remain poorly understood. Considering the diversity of such mechanisms among different emerging viruses allows us to highlight common features that may be of general validity and point out important challenges.
Collapse
Affiliation(s)
| | - Guttorm Haraldsen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Department of Pathology, University of Oslo, Oslo, Norway
| | - Knut Falk
- Norwegian Veterinary Institute, Oslo, Norway.,AquaMed Consulting AS, Oslo, Norway
| | - Reidunn Edelmann
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
| |
Collapse
|
19
|
Harper A, Vijayakumar V, Ouwehand AC, ter Haar J, Obis D, Espadaler J, Binda S, Desiraju S, Day R. Viral Infections, the Microbiome, and Probiotics. Front Cell Infect Microbiol 2021; 10:596166. [PMID: 33643929 PMCID: PMC7907522 DOI: 10.3389/fcimb.2020.596166] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/23/2020] [Indexed: 01/07/2023] Open
Abstract
Viral infections continue to cause considerable morbidity and mortality around the world. Recent rises in these infections are likely due to complex and multifactorial external drivers, including climate change, the increased mobility of people and goods and rapid demographic change to name but a few. In parallel with these external factors, we are gaining a better understanding of the internal factors associated with viral immunity. Increasingly the gastrointestinal (GI) microbiome has been shown to be a significant player in the host immune system, acting as a key regulator of immunity and host defense mechanisms. An increasing body of evidence indicates that disruption of the homeostasis between the GI microbiome and the host immune system can adversely impact viral immunity. This review aims to shed light on our understanding of how host-microbiota interactions shape the immune system, including early life factors, antibiotic exposure, immunosenescence, diet and inflammatory diseases. We also discuss the evidence base for how host commensal organisms and microbiome therapeutics can impact the prevention and/or treatment of viral infections, such as viral gastroenteritis, viral hepatitis, human immunodeficiency virus (HIV), human papilloma virus (HPV), viral upper respiratory tract infections (URTI), influenza and SARS CoV-2. The interplay between the gastrointestinal microbiome, invasive viruses and host physiology is complex and yet to be fully characterized, but increasingly the evidence shows that the microbiome can have an impact on viral disease outcomes. While the current evidence base is informative, further well designed human clinical trials will be needed to fully understand the array of immunological mechanisms underlying this intricate relationship.
Collapse
Affiliation(s)
- Ashton Harper
- ADM Health & Wellness, Medical Affairs Department, Somerset, United Kingdom
| | | | - Arthur C. Ouwehand
- Global Health and Nutrition Sciences, DuPont Nutrition and Biosciences, Kantvik, Finland
| | | | - David Obis
- Innovation Science & Nutrition Department, Danone Nutricia Research, Palaiseau, France
| | | | - Sylvie Binda
- Lallemand Health Solutions, Montreal, QC, Canada
| | | | - Richard Day
- ADM Health & Wellness, Medical Affairs Department, Somerset, United Kingdom
| |
Collapse
|
20
|
SARS-CoV-2 and Obesity: "CoVesity"-a Pandemic Within a Pandemic. Obes Surg 2021; 31:1745-1754. [PMID: 33479921 PMCID: PMC7819768 DOI: 10.1007/s11695-020-04919-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022]
Abstract
Individuals who are overweight or suffering from obesity are in a chronic state of low-grade inflammation, making them particularly susceptible to developing severe forms of respiratory failure. Studies conducted in past pandemics link obesity with worse health outcomes. This population is thus of particular concern within the context of the COVID-19 pandemic, considering the cessation of obesity management services. This systematic review highlights [1] the reciprocal link between the obesity and COVID-19 pandemics, [2] obesity as a risk factor for more severe disease in past pandemics, [3] potential mechanisms that make individual’s suffering from obesity more susceptible to severe disease and higher viral load, and [4] the need to safely resume bariatric services as recommended by expert guidelines, in order to mitigate the health outcomes of an already vulnerable population.
Collapse
|
21
|
Xu F, Gao J, Bergmann S, Sims AC, Ashbrook DG, Baric RS, Cui Y, Jonsson CB, Li K, Williams RW, Schughart K, Lu L. Genetic Dissection of the Regulatory Mechanisms of Ace2 in the Infected Mouse Lung. Front Immunol 2021; 11:607314. [PMID: 33488611 PMCID: PMC7819859 DOI: 10.3389/fimmu.2020.607314] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Acute lung injury (ALI) is an important cause of morbidity and mortality after viral infections, including influenza A virus H1N1, SARS-CoV, MERS-CoV, and SARS-CoV-2. The angiotensin I converting enzyme 2 (ACE2) is a key host membrane-bound protein that modulates ALI induced by viral infection, pulmonary acid aspiration, and sepsis. However, the contributions of ACE2 sequence variants to individual differences in disease risk and severity after viral infection are not understood. In this study, we quantified H1N1 influenza-infected lung transcriptomes across a family of 41 BXD recombinant inbred strains of mice and both parents—C57BL/6J and DBA/2J. In response to infection Ace2 mRNA levels decreased significantly for both parental strains and the expression levels was associated with disease severity (body weight loss) and viral load (expression levels of viral NA segment) across the BXD family members. Pulmonary RNA-seq for 43 lines was analyzed using weighted gene co-expression network analysis (WGCNA) and Bayesian network approaches. Ace2 not only participated in virus-induced ALI by interacting with TNF, MAPK, and NOTCH signaling pathways, but was also linked with high confidence to gene products that have important functions in the pulmonary epithelium, including Rnf128, Muc5b, and Tmprss2. Comparable sets of transcripts were also highlighted in parallel studies of human SARS-CoV-infected primary human airway epithelial cells. Using conventional mapping methods, we determined that weight loss at two and three days after viral infection maps to chromosome X—the location of Ace2. This finding motivated the hierarchical Bayesian network analysis, which defined molecular endophenotypes of lung infection linked to Ace2 expression and to a key disease outcome. Core members of this Bayesian network include Ace2, Atf4, Csf2, Cxcl2, Lif, Maml3, Muc5b, Reg3g, Ripk3, and Traf3. Collectively, these findings define a causally-rooted Ace2 modulatory network relevant to host response to viral infection and identify potential therapeutic targets for virus-induced respiratory diseases, including those caused by influenza and coronaviruses.
Collapse
Affiliation(s)
- Fuyi Xu
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jun Gao
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States.,Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Silke Bergmann
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David G Ashbrook
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yan Cui
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Colleen B Jonsson
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kui Li
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Robert W Williams
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Klaus Schughart
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,University of Veterinary Medicine Hannover, Hannover, Germany
| | - Lu Lu
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
22
|
Lonati C, Gatti S, Catania A. Activation of Melanocortin Receptors as a Potential Strategy to Reduce Local and Systemic Reactions Induced by Respiratory Viruses. Front Endocrinol (Lausanne) 2020; 11:569241. [PMID: 33362713 PMCID: PMC7758465 DOI: 10.3389/fendo.2020.569241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
The clinical hallmarks of infections caused by critical respiratory viruses consist of pneumonia, which can progress to acute lung injury (ALI), and systemic manifestations including hypercoagulopathy, vascular dysfunction, and endotheliitis. The disease outcome largely depends on the immune response produced by the host. The bio-molecular mechanisms underlying certain dire consequences of the infection partly arise from an aberrant production of inflammatory molecules, an event denoted as "cytokine storm". Therefore, in addition to antiviral therapies, molecules able to prevent the injury caused by cytokine excess are under investigation. In this perspective, taking advantage of melanocortin peptides and their receptors, components of an endogenous modulatory system that exerts marked anti-inflammatory and immunomodulatory influences, could be an effective therapeutic strategy to control disease evolution. Exploiting the melanocortin system using natural or synthetic ligands can form a realistic basis to counteract certain deleterious effects of respiratory virus infections. The central and peripheral protective actions exerted following melanocortin receptor activation could allow dampening the harmful events that trigger the cytokine storm and endothelial dysfunction while sustaining the beneficial signals required to elicit repair mechanisms. The long standing evidence for melanocortin safety encourages this approach.
Collapse
Affiliation(s)
- Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | |
Collapse
|
23
|
Mujica G, Sternberg Z, Solis J, Wand T, Carrasco P, Henao-Martínez AF, Franco-Paredes C. Defusing COVID-19: Lessons Learned from a Century of Pandemics. Trop Med Infect Dis 2020; 5:E182. [PMID: 33266051 PMCID: PMC7709642 DOI: 10.3390/tropicalmed5040182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022] Open
Abstract
Amidst the COVID-19 global pandemic of 2020, identifying and applying lessons learned from previous influenza and coronavirus pandemics may offer important insight into its interruption. Herein, we conducted a review of the literature of the influenza pandemics of the 20th century; and of the coronavirus and influenza pandemics of the 21st century. Influenza and coronavirus pandemics are zoonoses that spread rapidly in consistent seasonal patterns during an initial wave of infection and subsequent waves of spread. For all of their differences in the state of available medical technologies, global population changes, and social and geopolitical factors surrounding each pandemic, there are remarkable similarities among them. While vaccination of high-risk groups is advocated as an instrumental mode of interrupting pandemics, non-pharmacological interventions including avoidance of mass gatherings, school closings, case isolation, contact tracing, and the implementation of infection prevention strategies in healthcare settings represent the cornerstone to halting transmission. In conjunction with lessons learned from previous pandemics, the public health response to the COVID-19 pandemic constitutes the basis for delineating best practices to confront future pandemics.
Collapse
Affiliation(s)
- Graciela Mujica
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA; (G.M.); (Z.S.); (J.S.); (T.W.)
| | - Zane Sternberg
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA; (G.M.); (Z.S.); (J.S.); (T.W.)
| | - Jamie Solis
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA; (G.M.); (Z.S.); (J.S.); (T.W.)
| | - Taylor Wand
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA; (G.M.); (Z.S.); (J.S.); (T.W.)
| | - Peter Carrasco
- International Association for Immunization Managers, Washington, DC 20037, USA;
- Department of Immunization and Vaccines, World Health Organization, 1202 Geneva, Switzerland
| | - Andrés F. Henao-Martínez
- Department of Medicine, Division of Infectious Diseases, Anschutz Medical Center, University of Colorado, Aurora, CO 80045, USA;
| | - Carlos Franco-Paredes
- Department of Medicine, Division of Infectious Diseases, Anschutz Medical Center, University of Colorado, Aurora, CO 80045, USA;
- Hospital Infantil de México, Federico Gomez, México City 06720, Mexico
| |
Collapse
|
24
|
Zhou HX, Li RF, Wang YF, Shen LH, Cai LH, Weng YC, Zhang HR, Chen XX, Wu X, Chen RF, Jiang HM, Wang C, Yang M, Lu J, Luo XD, Jiang Z, Yang ZF. Total alkaloids from Alstonia scholaris inhibit influenza a virus replication and lung immunopathology by regulating the innate immune response. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 77:153272. [PMID: 32702592 DOI: 10.1016/j.phymed.2020.153272] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/01/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Alstonia scholaris is a folk medicine used to treat cough, asthma and chronic obstructive pulmonary disease in China. Total alkaloids (TA) from A. scholaris exhibit anti-inflammatory properties in acute respiratory disease, which suggests their possible anti-inflammatory effect on influenza virus infection. PURPOSE To assess the clinical use of TA by demonstrating their anti-influenza and anti-inflammatory effects and the possible mechanism underlying the effect of TA on influenza A virus (IAV) infection in vitro and to reveal the inhibitory effect of TA on lung immunopathology caused by IAV infection. METHODS Antiviral and anti-inflammatory activities were assessed in Madin-Darby canine kidney (MDCK) and A549 cells and U937-derived macrophages infected with influenza A/PR/8/34 (H1N1) virus. Proinflammatory cytokine levels were measured by real-time quantitative PCR and Bio-Plex assays. The activation of innate immune signaling induced by H1N1 virus in the absence or presence of TA was detected in A549 cells by Western blot. Furthermore, mice were infected intranasally with H1N1 virus and treated with TA (50, 25 and 12.5 mg/kg/d) or oseltamivir (60 mg/kg/d) for 5 days in vivo. The survival rates and body weight were recorded, and the viral titer, proinflammatory cytokine levels, innate immune cell populations and histopathological changes in the lungs were analyzed. RESULTS TA significantly inhibited viral replication in A549 cells and U937-derived macrophages and markedly reduced cytokine and chemokine production at the mRNA and protein levels. Furthermore, TA blocked the activation of pattern recognition receptor (PRR)- and IFN-activated signal transduction in A549 cells. Critically, TA also increased the survival rate, reduced the viral titer, suppressed proinflammatory cytokine production and innate immune cell infiltration and improved lung histopathology in a lethal PR8 mouse model. CONCLUSION TA exhibits anti-viral and anti-inflammatory effects against IAV infection by interfering with PRR- and IFN-activated signal transduction.
Collapse
Affiliation(s)
- Hong-Xia Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China; Dongguan People's Hospital, Dongguan, 523000, China
| | - Run-Feng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yi-Feng Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Li-Han Shen
- Dongguan People's Hospital, Dongguan, 523000, China
| | - Li-Hua Cai
- Dongguan People's Hospital, Dongguan, 523000, China
| | - Yun-Ceng Weng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | | | - Xin-Xin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiao Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Rui-Feng Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Hai-Ming Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Caiyun Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China
| | - Mingrong Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China
| | - Jingguang Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China
| | - Xiao-Dong Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Zhihong Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China; Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, 510000, China
| | - Zi-Feng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China; KingMed Virology Diagnostic & Translational Center, 510000, China; Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, 510000, China.
| |
Collapse
|
25
|
Coupled CRC 2D and ALI 3D Cultures Express Receptors of Emerging Viruses and Are More Suitable for the Study of Viral Infections Compared to Conventional Cell Lines. Stem Cells Int 2020; 2020:2421689. [PMID: 32695180 PMCID: PMC7368225 DOI: 10.1155/2020/2421689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 02/02/2023] Open
Abstract
Infections of emerging and reemerging viruses (SARS-CoVs, influenza H1N1, etc.) largely and globally affect human health. Animal models often fail to reflect a physiological status because of species tropism of virus infection. Conventional cell lines are usually genetically and phenotypically different from primary cells. Developing an in vitro physiological model to study the infection of emerging viruses will facilitate our understanding of virus-host cell interactions, thereby benefiting antiviral drug discovery. In the current work, we first established normal airway epithelial cells (upper and lower airway track) in 2D and 3D culture systems using conditional reprogramming (CR) and air-liquid interface (ALI) techniques. These long-term cultures maintained differentiation potential. More importantly, these cells express two types of influenza virus receptors, α2-6-Gal- and α2-3-Gal-linked sialic acids, and angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoVs as well. These cells were permissive to the infection of pandemic influenza H1N1 (H1N1pdm). In contrast, the lung cancer cell line A549 and immortalized airway epithelial cells (16HBE) were not susceptible to H1N1 infection. A virus-induced cytopathic effect (CPE) on 2D CRC cultures developed in a time-dependent manner. The pathological effects were also readily observed spreading from the apical layer to the basal layer of the 3D ALI culture. This integrated 2D CRC and 3D ALI cultures provide a physiological and personalized in vitro model to study the infection of emerging viruses. This novel model can be used for studying virus biology and host response to viral infection and for antiviral drug discovery.
Collapse
|
26
|
Abstract
It has been over 100 years since the 1918 influenza pandemic, one of the most infamous examples of viral immunopathology. Since that time, there has been an inevitable repetition of influenza pandemics every few decades and yearly influenza seasons, which have a significant impact on human health. Recently, noteworthy progress has been made in defining the cellular and molecular mechanisms underlying pathology induced by an exuberant host response to influenza virus infection. Infection with influenza viruses is associated with a wide spectrum of disease, from mild symptoms to severe complications including respiratory failure, and the severity of influenza disease is driven by a complex interplay of viral and host factors. This chapter will discuss mechanisms of infection severity using concepts of disease resistance and tolerance as a framework for understanding the balance between viral clearance and immunopathology. We review mechanistic studies in animal models of infection and correlational studies in humans that have begun to define these factors and discuss promising host therapeutic targets to improve outcomes from severe influenza disease.
Collapse
Affiliation(s)
- David F Boyd
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Taylor L Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
27
|
Liu X, Wu Y, Rong L. Conditionally Reprogrammed Human Normal Airway Epithelial Cells at ALI: A Physiological Model for Emerging Viruses. Virol Sin 2020; 35:280-289. [PMID: 32557270 PMCID: PMC7298165 DOI: 10.1007/s12250-020-00244-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer cell lines have been used widely in cancer biology, and as biological or functional cell systems in many biomedical research fields. These cells are usually defective for many normal activities or functions due to significant genetic and epigenetic changes. Normal primary cell yields and viability from any original tissue specimens are usually relatively low or highly variable. These normal cells cease after a few passages or population doublings due to very limited proliferative capacity. Animal models (ferret, mouse, etc.) are often used to study virus-host interaction. However, viruses usually need to be adapted to the animals by several passages due to tropism restrictions including viral receptors and intracellular restrictions. Here we summarize applications of conditionally reprogrammed cells (CRCs), long-term cultures of normal airway epithelial cells from human nose to lung generated by conditional cell reprogramming (CR) technology, as an ex vivo model in studies of emerging viruses. CR allows to robustly propagate cells from non-invasive or minimally invasive specimens, for example, nasal or endobronchial brushing. This process is rapid (2 days) and conditional. The CRCs maintain their differentiation potential and lineage functions, and have been used for studies of adenovirus, rhinovirus, respiratory syncytial virus, influenza viruses, parvovirus, and SARS-CoV. The CRCs can be easily used for air-liquid interface (ALI) polarized 3D cultures, and these coupled CRC/ALI cultures mimic physiological conditions and are suitable for studies of viral entry including receptor binding and internalization, innate immune responses, viral replications, and drug discovery as an ex vivo model for emerging viruses.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA.
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| | - Yuntao Wu
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinoi at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
28
|
Yang L, Wang S, Wang Y, Zhao P, Cui C, Tu L, Li X, Yu Y, Li H, Wang L. Diversity of locally produced IFN-α subtypes in human nasopharyngeal epithelial cells and mouse lung tissues during influenza virus infection. Appl Microbiol Biotechnol 2020; 104:6351-6361. [PMID: 32472176 DOI: 10.1007/s00253-020-10676-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/30/2020] [Accepted: 05/10/2020] [Indexed: 11/24/2022]
Abstract
The excessively expressed interferon-α (IFN-α) might contribute to the uncontrolled inflammatory responses, causing pathological damage during influenza virus infection. However, the correlation of the pathological damage with the expression profile of IFN-α subtypes in the focus of infection with influenza viruses is poorly understood. To investigate this, we detected the IFN-α subtype dominance in human respiratory epithelial cells and mouse lungs, both of which were infected with influenza viruses. It was found that IFN-α1, IFN-α6, IFN-α14, and IFN-α16 were dominantly expressed in respiratory epithelial cells from the patients infected with IAV, whereas IFN-α5, IFN-α8, and IFN-α21 were dominantly expressed in respiratory epithelial cells from the patients infected with less pathogenic IBV and that IFN-α1, IFN-α9, and IFN-α15 were dominantly expressed in lungs of the mice infected with H1N1 IAV, and IFN-α2, IFN-α12, and IFN-α13 were dominantly expressed in lungs of the mice infected with less pathogenic H9N2 IAV. Compared with H9N2 IAV, H1N1 IAV induced higher mortality rates and more obvious body weight loss in the mice. In addition, IAV or H1N1 IAV induced a significantly higher level of CXCL10 mRNA in the human respiratory epithelial cells or the mouse lungs, respectively. In mice, the high level of Cxcl10 mRNA was accompanied by the abundant infiltrated neutrophils and more severe pathological changes in the lungs. Together, the data presented here indicate that the pathogenicity of influenza viruses is correlated with the IFN-α subtypes induced by influenza viruses. KEY POINTS: • Different influenza viruses induce differential inflammation responses. • Various influenza viruses induce diverse expression profiles of IFN-α subtypes. • The locally produced IFN-α subtypes correlated to the differential inflammation. Graphical abstract.
Collapse
Affiliation(s)
- Lei Yang
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Shengnan Wang
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Ying Wang
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Peiyan Zhao
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Cuiyun Cui
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Liqun Tu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Xin Li
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Haibo Li
- Department of Pediatric Clinic, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| | - Liying Wang
- Institute of Pediatrics in The First Hospital of Jilin University and Department of Molecular Biology in College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
29
|
COVID-19 and ACE -inhibitors and angiotensin receptor blockers-: The need to differentiate between early infection and acute lung injury. REVISTA COLOMBIANA DE CARDIOLOGÍA 2020. [PMCID: PMC7174165 DOI: 10.1016/j.rccar.2020.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
30
|
Smirnov VS, Totolyan AA. Innate immunity in coronavirus infection. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2020. [DOI: 10.15789/2220-7619-iii-1440] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Coronaviruses (CoVs) comprise a polymorphic group of respiratory viruses causing acute inflammatory diseases in domestic and agricultural animals (chicken, pig, buffalo, cat, dog). Until recently, this infection in humans was mainly observed during the autumn-winter period and characterized by a mild, often asymptomatic, course. The situation changed dramatically in 2003, when SARS outbreak caused by pathogenic CoV (SARS-CoV) was recorded in China. A decade later, a new CoV outbreak occurred in the form of the Middle East respiratory syndrome (MERS-CoV), whereas in December 2019, SARS-CoV-2 (COVID-19) cases were recorded, which transformed within the first months of 2020 into the pandemic. In all three cases, CoV disease led to severe bronchopulmonary lesions, varying from dry, debilitating cough to acute respiratory distress syndrome (ARDS). At the same time, multiple changes in innate immunity were noted most often manifested as a pronounced inflammatory reaction in the lower respiratory tract, featured by damaged type II pneumocytes, apoptosis, hyalinization of alveolar membranes, focal or generalized pulmonary edema. Destructive processes in the respiratory tract were accompanied by migration of monocytes/macrophages and granulocyte neutrophils to the inflammatory focus. Such events were accompanied by production of pro-inflammatory cytokines, which magnitude could ascend up to a cytokine storm. SARS-CoV is characterized by symptoms of secondary immunosuppression, manifested by the late onset of interferon production and activation of NLRP3 inflammasomes – the key inflammatory factor. The reason for such reaction may be accounted for by CoV arsenal containing extensive set of structural and non-structural proteins exerting pro-inflammatory and immunosuppressive properties. Delayed IFN production allowed CoV to replicate actively and freely, and when type I IFN synthesis was eventually triggered, its activity was detrimental and accompanied by an aggravated infection course. Thus, SARS can surely be referred to immune-dependent infections with a marked immunopathological component. The purpose of this review was to describe some mechanisms underlying formation of innate immune response to infection caused by pathogenic coronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2 (COVID-19).
Collapse
|
31
|
Fontes CAP, Dos Santos AASMD, de Oliveira SA, Aidê MA. Influenza A virus H1N1 associated pneumonia - acute and late aspects evaluated with high resolution tomography in hospitalized patients. Multidiscip Respir Med 2020; 15:692. [PMID: 33117533 PMCID: PMC7542991 DOI: 10.4081/mrm.2020.692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
Background Influenza A (H1N1) virus often compromises the respiratory tract, leading to pneumonia, which is the principal cause of death in these patients. The purpose of this study was to review the acute and late phase pulmonary findings in influenza A(H1N1) associated pneumonia using high resolution computed tomography (HRCT), and to determine the importance of performing end expiration series. Methods Between July and August 2009, 140 patients presented with influenza A (H1N1) confirmed by real-timepolymerase chain reaction. Out of these, 27 patients underwent HRCT in the acute and late phases of pneumonia, allowing for a comparative study. Late phase exams were performed due to clinical worsening and up to 120 days later in patients with persistent complaints of dyspnea. Results Ground glass opacities, consolidations, and the combination of both were associated with the acute phase, whereas persistence or worsening of the lesions, lesion improvement, and air trapping in the end expiration series (as seen using HRCT, n=6) were observed in the late phase. Conclusions In the HRCT end expiration series, air trapping was found in the late phase of H1N1 associated pneumonia. Generally, these exams are not evaluated in research articles, and air trapping has not previously been studied using the end expiration series. Our study brings more scientific knowledge about aspects of pulmonary involvement by influenza A (H1N1), through evaluation with end expiration series, which makes the CT exam dynamic, translating the respiratory movement, and showing bronchial alteration.
Collapse
Affiliation(s)
| | - Alair Augusto Sarmet Moreira Damas Dos Santos
- Radiology Service, Department of Radiology, Faculty of Medicine, Antônio Pedro University Hospital, Federal Fluminense University, Niterói.,Imaging Center of the Complex Hospital of Niterói (CHN), Niterói
| | - Solange Artimos de Oliveira
- Department of Clinical Medicine, Faculty of Medicine, Antônio Pedro University Hospital, Federal Fluminense University, Niterói, Brazil
| | - Miquel Abdon Aidê
- Department of Clinical Medicine, Faculty of Medicine, Antônio Pedro University Hospital, Federal Fluminense University, Niterói, Brazil
| |
Collapse
|
32
|
Mehrbod P, Ebrahimi SN, Fotouhi F, Eskandari F, Eloff JN, McGaw LJ, Fasina FO. Experimental validation and computational modeling of anti-influenza effects of quercetin-3-O-α-L-rhamnopyranoside from indigenous south African medicinal plant Rapanea melanophloeos. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:346. [PMID: 31791311 PMCID: PMC6888925 DOI: 10.1186/s12906-019-2774-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/27/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Influenza A virus (IAV) is still a major health threat. The clinical manifestations of this infection are related to immune dysregulation, which causes morbidity and mortality. The usage of traditional medication with immunomodulatory properties against influenza infection has been increased recently. Our previous study showed antiviral activity of quercetin-3-O-α-L-rhamnopyranoside (Q3R) isolated from Rapanea melanophloeos (RM) (L.) Mez (family Myrsinaceae) against H1N1 (A/PR/8/34) infection. This study aimed to confirm the wider range of immunomodulatory effect of Q3R on selective pro- and anti-inflammatory cytokines against IAV in vitro, to evaluate the effect of Q3R on apoptosis pathway in combination with H1N1, also to assess the physical interaction of Q3R with virus glycoproteins and RhoA protein using computational docking. METHODS MDCK cells were exposed to Q3R and 100CCID50/100 μl of H1N1 in combined treatments (co-, pre- and post-penetration treatments). The treatments were tested for the cytokines evaluation at RNA and protein levels by qPCR and ELISA, respectively. In another set of treatment, apoptosis was examined by detecting RhoA GTPase protein and caspase-3 activity. Molecular docking was used as a tool for evaluation of the potential anti-influenza activity of Q3R. RESULTS The expressions of cytokines in both genome and protein levels were significantly affected by Q3R treatment. It was shown that Q3R was much more effective against influenza when it was applied in co-penetration treatment. Q3R in combination with H1N1 increased caspase-3 activity while decreasing RhoA activation. The molecular docking results showed strong binding ability of Q3R with M2 transmembrane, Neuraminidase of 2009 pandemic H1N1, N1 and H1 of PR/8/1934 and Human RhoA proteins, with docking energy of - 10.81, - 10.47, - 9.52, - 9.24 and - 8.78 Kcal/mol, respectively. CONCLUSIONS Quercetin-3-O-α-L-rhamnopyranoside from RM was significantly effective against influenza infection by immunomodulatory properties, affecting the apoptosis pathway and binding ability to viral receptors M2 transmembrane and Neuraminidase of 2009 pandemic H1N1 and human RhoA cellular protein. Further research will focus on detecting the detailed specific mechanism of Q3R in virus-host interactions.
Collapse
Affiliation(s)
- Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran, Iran
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Samad Nejad Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Fotouhi
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Eskandari
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran, Iran
| | - Jacobus N. Eloff
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Lyndy J. McGaw
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Folorunso O. Fasina
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
- ECTAD, Food and Agriculture Organization of the United Nations (FAO), Dar es Salaam, Tanzania
| |
Collapse
|
33
|
Localization Analysis of Heterophilic Antigen Epitopes of H1N1 Influenza Virus Hemagglutinin. Virol Sin 2019; 34:306-314. [PMID: 31020574 DOI: 10.1007/s12250-019-00100-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/23/2019] [Indexed: 10/26/2022] Open
Abstract
Previous studies have indicated that two monoclonal antibodies (mAbs; A1-10 and H1-84) of the hemagglutinin (HA) antigen on the H1N1 influenza virus cross-react with human brain tissue. It has been proposed that there are heterophilic epitopes between the HA protein and human brain tissue (Guo et al. in Immunobiology 220:941-946, 2015). However, characterisation of the two mAbs recognising the heterophilic epitope on HA has not yet been performed. In the present study, the common antigens of influenza virus HA were confirmed using indirect enzyme-linked immunosorbent assays and analysed with DNAMAN software. The epitopes were localized to nine peptides in the influenza virus HA sequence and the distribution of the peptides in the three-dimensional structure of HA was determined using PyMOL software. Key amino acids and variable sequences of the antibodies were identified using abYsis software. The results demonstrated that there were a number of common antigens among the five influenza viruses studied that were recognised by the mAbs. One of the peptides, P2 (LVLWGIHHP191-199), bound both of the mAbs and was located in the head region of HA. The key amino acids of this epitope and the variable regions in the heavy and light chain sequences of the mAbs that recognised the epitope are described. A heterophilic epitope on H1N1 influenza virus HA was also introduced. The existence of this epitope provides a novel perspective for the occurrence of nervous system diseases that could be caused by influenza virus infection, which might aid in influenza prevention and control.
Collapse
|
34
|
Henry C, Zaizafoun M, Stock E, Ghamande S, Arroliga AC, White HD. Impact of angiotensin-converting enzyme inhibitors and statins on viral pneumonia. Proc (Bayl Univ Med Cent) 2018; 31:419-423. [PMID: 30948970 DOI: 10.1080/08998280.2018.1499293] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 01/15/2023] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors and statins may potentially benefit patients with viral infections and pneumonia. Our study aimed to evaluate the impact of ACE inhibitors and statins on the rates of intubation and death in viral pneumonia. We retrospectively studied 1055 adult patients admitted to a tertiary care center in central Texas with a positive respiratory viral polymerase chain reaction test. Of these, 539 had clinical presentation and imaging consistent with pneumonia. We collected information on demographic characteristics, microbiology, comorbid conditions, medication use, and outcomes. ACE inhibitors given prior to admission were associated with an increased risk of death or intubation (odds ratio [OR] = 3.02; 95% confidence interval [CI], 1.30-7.01), whereas statin use prior to admission did not change rates of death or intubation. Lower rates of death and intubation were noted with continued use of ACE inhibitors (OR =0.25; 95% CI, 0.09-0.64) and statins (OR =0.26; 95% CI, 0.08-0.81) throughout the hospital stay. We added further evidence of the beneficial effect of continued use of ACE inhibitors and statins in viral pneumonia.
Collapse
Affiliation(s)
- Christopher Henry
- Division of Pulmonary, Critical Care, and Sleep Medicine, Baylor Scott & White HealthTempleTexas
| | - Manaf Zaizafoun
- Division of Pulmonary and Critical Care Medicine, University Hospitals, Ahuja Medical CenterBedfordOhio
| | - Eileen Stock
- Perry Point/Baltimore Coordinating Center, Cooperative Studies Program, Office of Research and Development, US Department of Veterans AffairsPerry PointMaryland
| | - Shekhar Ghamande
- Division of Pulmonary, Critical Care, and Sleep Medicine, Baylor Scott & White HealthTempleTexas
| | - Alejandro C Arroliga
- Division of Pulmonary, Critical Care, and Sleep Medicine, Baylor Scott & White HealthTempleTexas
| | - Heath D White
- Division of Pulmonary, Critical Care, and Sleep Medicine, Baylor Scott & White HealthTempleTexas
| |
Collapse
|
35
|
Yip TF, Selim ASM, Lian I, Lee SMY. Advancements in Host-Based Interventions for Influenza Treatment. Front Immunol 2018; 9:1547. [PMID: 30042762 PMCID: PMC6048202 DOI: 10.3389/fimmu.2018.01547] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
Influenza is a major acute respiratory infection that causes mortality and morbidity worldwide. Two classes of conventional antivirals, M2 ion channel blockers and neuraminidase inhibitors, are mainstays in managing influenza disease to lessen symptoms while minimizing hospitalization and death in patients with severe influenza. However, the development of viral resistance to both drug classes has become a major public health concern. Vaccines are prophylaxis mainstays but are limited in efficacy due to the difficulty in matching predicted dominant viral strains to circulating strains. As such, other potential interventions are being explored. Since viruses rely on host cellular functions to replicate, recent therapeutic developments focus on targeting host factors involved in virus replication. Besides controlling virus replication, potential targets for drug development include controlling virus-induced host immune responses such as the recently suggested involvement of innate lymphoid cells and NADPH oxidases in influenza virus pathogenesis and immune cell metabolism. In this review, we will discuss the advancements in novel host-based interventions for treating influenza disease.
Collapse
Affiliation(s)
- Tsz-Fung Yip
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Aisha Sami Mohammed Selim
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ida Lian
- School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, Singapore, Singapore
| | - Suki Man-Yan Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
36
|
Shao X, Li X, Ma X, Zhu F. Long-term prediction of dynamic distribution of passive contaminant in complex recirculating ventilation system. BUILDING AND ENVIRONMENT 2017; 121:49-66. [PMID: 32287971 PMCID: PMC7126526 DOI: 10.1016/j.buildenv.2017.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 06/11/2023]
Abstract
Recirculating ventilation systems may act as carriers of hazardous substances. The long-term prediction of the dynamic distribution of contaminants in this type of system is crucial for the evaluation of pollution and further design of more efficient ventilation systems. However, few convenient methods can predict the dynamic distribution of contaminants, because the dynamic supply air concentrations resulting from air recirculation are unknown, especially over long time periods, such as months or years. In this study, a novel method is proposed to predict the dynamic distribution of contaminants over a long time period in a complex recirculating ventilation system, where an algebraic expression based on the indices of the response coefficient is applied to account for the relationship between the contaminant distribution inside the room and various boundary conditions. The method is established by obtaining comprehensive mathematical descriptions of the relationships between concentrations of contaminants in the air handling units, supply air inlets, return air outlets, and fresh air. Hourly supply air concentrations can be easily obtained by solving a matrix, and the dynamic distribution of contaminants is then calculated using an expression based on the response coefficient. The reliability of the proposed method is analyzed by both experimental and numerical methods. A simplified method is suggested to accelerate the time-consuming calculation of the response coefficient. The proposed method is beneficial for predicting three-dimensional dynamic distribution of contaminants in complex ventilation systems with an acceptable accuracy and time cost.
Collapse
Affiliation(s)
- Xiaoliang Shao
- School of Civil and Resource Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xianting Li
- Department of Building Science, School of Architecture, Tsinghua University, Beijing 100084, China
| | - Xiaojun Ma
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Fenfei Zhu
- Department of Building Science, School of Architecture, Tsinghua University, Beijing 100084, China
| |
Collapse
|
37
|
Nuclear translocation of HIF-1α induced by influenza A (H1N1) infection is critical to the production of proinflammatory cytokines. Emerg Microbes Infect 2017; 6:e39. [PMID: 28536432 PMCID: PMC5520484 DOI: 10.1038/emi.2017.21] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/25/2017] [Accepted: 03/02/2017] [Indexed: 02/08/2023]
Abstract
Infection with the influenza A (H1N1) virus is a major challenge for public health because it can cause severe morbidity and even mortality in humans. The over-secretion of inflammatory cytokines (cytokine storm) is considered to be a key contributor to the severe pneumonia caused by H1N1 infection. It has been reported that hypoxia-inducible factor 1-alpha (HIF-1α) is associated with the production of proinflammatory molecules, but whether HIF-1α participates in the acute inflammatory responses against H1N1 infection is still unclear. To investigate the role of HIF-1α in H1N1 infection, the expression and nuclear translocation of HIF-1α in A549 and THP-1 cell lines infected with H1N1 virus were observed. The results showed that without altering the intracellular mRNA or protein expression of HIF-1α, H1N1 infection only induced nuclear translocation of HIF-1α under normal oxygen concentrations. The use of 2-methoxyestradiol (2ME2), a HIF-1α inhibitor that blocks HIF-1α nuclear accumulation, in H1N1-infected cells decreased the mRNA and protein expression of tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6 and increased the levels of IL-10. In contrast, H1N1-infected cells under hypoxic conditions had increased HIF-1α nuclear accumulation, increased expression of TNF-α and IL-6 and decreased levels of IL-10. In conclusion, our data implied that in vitro H1N1 infection induced nuclear translocation of HIF-1α without altering the expression of HIF-1α, which may promote the secretion of proinflammatory cytokines during H1N1 infection.
Collapse
|
38
|
Affiliation(s)
- Raj D Shah
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, 676 North St. Clair Street, Arkes 14-045, Chicago, IL 60611, USA
| | - Richard G Wunderink
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, 676 North St. Clair Street, Arkes 14-045, Chicago, IL 60611, USA.
| |
Collapse
|
39
|
Allyn PR, Duffy EL, Humphries RM, Injean P, Weigt SS, Saggar R, Shino MY, Lynch JP, Ardehali A, Kubak B, Tseng CH, Belperio JA, Ross DJ, Gregson AL. Graft Loss and CLAD-Onset Is Hastened by Viral Pneumonia After Lung Transplantation. Transplantation 2016; 100:2424-2431. [PMID: 27467538 PMCID: PMC5077663 DOI: 10.1097/tp.0000000000001346] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Community-acquired respiratory virus (CARV) infections occur frequently after lung transplantation and may adversely impact outcomes. We hypothesized that while asymptomatic carriage would not increase the risk of chronic lung allograft dysfunction (CLAD) and graft loss, severe infection would. METHODS All lung transplant cases between January 2000 and July 2013 performed at our center were reviewed for respiratory viral samples. Each isolation of virus was classified according to clinical level of severity: asymptomatic, symptomatic without pneumonia, and viral pneumonia. Multivariate Cox modeling was used to assess the impact of CARV isolation on progression to CLAD and graft loss. RESULTS Four thousand four hundred eight specimens were collected from 563 total patients, with 139 patients producing 324 virus-positive specimens in 245 episodes of CARV infection. Overall, the risk of CLAD was elevated by viral infection (hazard ratio [HR], 1.64; P < 0.01). This risk, however, was due to viral pneumonia alone (HR, 3.94; P < 0.01), without significant impact from symptomatic viral infection (HR, 0.97; P = 0.94) nor from asymptomatic viral infection (HR, 0.99; P = 0.98). The risk of graft loss was not increased by asymptomatic CARV infection (HR, 0.74; P = 0.37) nor symptomatic CARV infection (HR, 1.39; P = 0.41). Viral pneumonia did, however, significantly increase the risk of graft loss (HR, 2.78; P < 0.01). CONCLUSIONS With respect to CARV, only viral pneumonia increased the risk of both CLAD and graft loss after lung transplantation. In the absence of pneumonia, respiratory viruses had no impact on measured outcomes.
Collapse
Affiliation(s)
- Paul R. Allyn
- Division of Infectious Diseases, Department of Medicine, University of California Los Angeles
| | - Erin L. Duffy
- Department of Medicine Statistics Core, University of California Los Angeles
| | - Romney M. Humphries
- Department of Pathology and Laboratory Medicine, University of California Los Angeles
| | - Patil Injean
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - S. Samuel Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - Michael Y. Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - Joseph P. Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - Abbas Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, University of California Los Angeles
| | - Bernard Kubak
- Division of Infectious Diseases, Department of Medicine, University of California Los Angeles
| | - Chi-Hong Tseng
- Department of Medicine Statistics Core, University of California Los Angeles
| | - John A. Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - David J. Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles
| | - Aric L. Gregson
- Division of Infectious Diseases, Department of Medicine, University of California Los Angeles
| |
Collapse
|
40
|
Tavares LP, Teixeira MM, Garcia CC. The inflammatory response triggered by Influenza virus: a two edged sword. Inflamm Res 2016; 66:283-302. [PMID: 27744631 DOI: 10.1007/s00011-016-0996-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV) is a relevant respiratory tract pathogen leading to a great number of deaths and hospitalizations worldwide. Secondary bacterial infections are a very common cause of IAV associated morbidity and mortality. The robust inflammatory response that follows infection is important for the control of virus proliferation but is also associated with lung damage, morbidity and death. The role of the different components of immune response underlying protection or disease during IAV infection is not completely elucidated. Overall, in the context of IAV infection, inflammation is a 'double edge sword' necessary to control infection but causing disease. Therefore, a growing number of studies suggest that immunomodulatory strategies may improve disease outcome without affecting the ability of the host to deal with infection. This review summarizes recent aspects of the inflammatory responses triggered by IAV that are preferentially involved in causing severe pulmonary disease and the anti-inflammatory strategies that have been suggested to treat influenza induced immunopathology.
Collapse
Affiliation(s)
- Luciana P Tavares
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristiana C Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil. .,Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, 21040360, Rio de Janeiro, Brazil.
| |
Collapse
|
41
|
Abstract
Survival after lung transplantation is limited in large part due to the high incidence of chronic rejection, known as chronic lung allograft dysfunction (CLAD). Pulmonary infections are a frequent complication in lung transplant recipients, due both to immunosuppressive medications and constant exposure of the lung allograft to the external environment via the airways. Infection is a recognized risk factor for the development of CLAD, and both acute infection and chronic lung allograft colonization with microorganisms increase the risk for CLAD. Acute infection by community acquired respiratory viruses, and the bacteria Pseudomonas aeruginosa and Staphylococcus aureus are increasingly recognized as important risk factors for CLAD. Colonization by the fungus Aspergillus may also augment the risk of CLAD. Fostering this transition from healthy lung to CLAD in each of these infectious episodes is the persistence of an inflammatory lung allograft environment.
Collapse
Affiliation(s)
- Aric L Gregson
- Division of Infectious Diseases, Department of Medicine, University of California, Box 957119, Warren Hall 14-154, Los Angeles, CA, 90995-7119, USA.
| |
Collapse
|
42
|
Nedel WL, Nora DG, Salluh JIF, Lisboa T, Póvoa P. Corticosteroids for severe influenza pneumonia: A critical appraisal. World J Crit Care Med 2016; 5:89-95. [PMID: 26855898 PMCID: PMC4733461 DOI: 10.5492/wjccm.v5.i1.89] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/24/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Influenza pneumonia is associated with high number of severe cases requiring hospital and intensive care unit (ICU) admissions with high mortality. Systemic steroids are proposed as a valid therapeutic option even though its effects are still controversial. Heterogeneity of published data regarding study design, population demographics, severity of illness, dosing, type and timing of corticosteroids administered constitute an important limitation for drawing robust conclusions. However, it is reasonable to admit that, as it was not found any advantage of corticosteroid therapy in so diverse conditions, such beneficial effects do not exist at all. Its administration is likely to increase overall mortality and such trend is consistent regardless of the quality as well as the sample size of studies. Moreover it was shown that corticosteroids might be associated with higher incidence of hospital-acquired pneumonia and longer duration of mechanical ventilation and ICU stay. Finally, it is reasonable to conclude that corticosteroids failed to demonstrate any beneficial effects in the treatment of patients with severe influenza infection. Thus its current use in severe influenza pneumonia should be restricted to very selected cases and in the setting of clinical trials.
Collapse
|
43
|
Abstract
The outbreak of Enterovirus D-68 (EV-D68) in the United States in 2014 raised great interest due to it affecting large numbers of people and because patients presented with severe respiratory and/or central nervous system involvement. Many studies have tried to evaluate the biologic and genetic characteristics of this virus, its association with disease development and the possibility of infection prevention and therapy. The main aim of this paper is to discuss what is presently known and what might be expected in the future regarding EV-D68. We highlight that further studies are needed to precisely define the epidemiology and total burden of EV-D68, the real age prevalence, and the factors that may lead to negative outcomes in some patients and not in others. Moreover, if recently reported clinical data are confirmed, specific efficacious prophylactic and therapeutic measures should be urgently developed.
Collapse
Affiliation(s)
- Nicola Principi
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Susanna Esposito
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
44
|
The role of C5a in acute lung injury induced by highly pathogenic viral infections. Emerg Microbes Infect 2015; 4:e28. [PMID: 26060601 PMCID: PMC4451266 DOI: 10.1038/emi.2015.28] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/12/2015] [Accepted: 03/31/2015] [Indexed: 12/14/2022]
Abstract
The complement system, an important part of innate immunity, plays a critical role in pathogen clearance. Unregulated complement activation is likely to play a crucial role in the pathogenesis of acute lung injury (ALI) induced by highly pathogenic virus including influenza A viruses H5N1, H7N9, and severe acute respiratory syndrome (SARS) coronavirus. In highly pathogenic virus-induced acute lung diseases, high levels of chemotactic and anaphylatoxic C5a were produced as a result of excessive complement activaiton. Overproduced C5a displays powerful biological activities in activation of phagocytic cells, generation of oxidants, and inflammatory sequelae named "cytokine storm", and so on. Blockade of C5a signaling have been implicated in the treatment of ALI induced by highly pathogenic virus. Herein, we review the literature that links C5a and ALI, and review our understanding of the mechanisms by which C5a affects ALI during highly pathogenic viral infection. In particular, we discuss the potential of the blockade of C5a signaling to treat ALI induced by highly pathogenic viruses.
Collapse
|
45
|
Micro-RNAs in regenerating lungs: an integrative systems biology analysis of murine influenza pneumonia. BMC Genomics 2014; 15:587. [PMID: 25015185 PMCID: PMC4108790 DOI: 10.1186/1471-2164-15-587] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 06/27/2014] [Indexed: 12/20/2022] Open
Abstract
Background Tissue regeneration in the lungs is gaining increasing interest as a potential influenza management strategy. In this study, we explored the role of microRNAs, short non-coding RNAs involved in post-transcriptional regulation, during pulmonary regeneration after influenza infection. Results We profiled miRNA and mRNA expression levels following lung injury and tissue regeneration using a murine influenza pneumonia model. BALB/c mice were infected with a sub-lethal dose of influenza A/PR/8(H1N1) virus, and their lungs were harvested at 7 and 15 days post-infection to evaluate the expression of ~300 miRNAs along with ~36,000 genes using microarrays. A global network was constructed between differentially expressed miRNAs and their potential target genes with particular focus on the pulmonary repair and regeneration processes to elucidate the regulatory role of miRNAs in the lung repair pathways. The miRNA arrays revealed a global down-regulation of miRNAs. TargetScan analyses also revealed specific miRNAs highly involved in targeting relevant gene functions in repair such as miR-290 and miR-505 at 7 dpi; and let-7, miR-21 and miR-30 at 15 dpi. Conclusion The significantly differentially regulated miRNAs are implicated in the activation or suppression of cellular proliferation and stem cell maintenance, which are required during the repair of the damaged lungs. These findings provide opportunities in the development of novel repair strategies in influenza-induced pulmonary injury. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-587) contains supplementary material, which is available to authorized users.
Collapse
|
46
|
Klumpp K, Crépin T. Capsid proteins of enveloped viruses as antiviral drug targets. Curr Opin Virol 2014; 5:63-71. [PMID: 24607800 DOI: 10.1016/j.coviro.2014.02.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 01/10/2014] [Accepted: 02/02/2014] [Indexed: 01/01/2023]
Abstract
Viral proteins have enabled the design of selective and efficacious treatments for viral diseases. While focus in this area has been on viral enzymes, it appears that multifunctional viral proteins may be even more susceptible to small molecule interference. As exemplified by HIV capsid, small molecule inhibitors can bind to multiple binding sites on the capsid protein and induce or prevent protein interactions and conformational changes. Resistance selection is complicated by the fact that the capsid proteins have to engage in different protein interactions at different times of the life cycle. Viral capsid assembly and disassembly have therefore emerged as highly sensitive processes that could deliver a new generation of antiviral agents across viral diseases.
Collapse
Affiliation(s)
- Klaus Klumpp
- Novira Therapeutics, Inc., 3805 Old Easton Road, Doylestown, PA 18902, United States.
| | - Thibaut Crépin
- University of Grenoble Alpes-EMBL-CNRS, Unit for Virus Host-Cell Interactions, 6 rue Jules Horowitz, 38042, France
| |
Collapse
|