1
|
Li H, Wang A, Zhang Y, Wei F. Diverse roles of lung macrophages in the immune response to influenza A virus. Front Microbiol 2023; 14:1260543. [PMID: 37779697 PMCID: PMC10534047 DOI: 10.3389/fmicb.2023.1260543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/16/2023] [Indexed: 10/03/2023] Open
Abstract
Influenza viruses are one of the major causes of human respiratory infections and the newly emerging and re-emerging strains of influenza virus are the cause of seasonal epidemics and occasional pandemics, resulting in a huge threat to global public health systems. As one of the early immune cells can rapidly recognize and respond to influenza viruses in the respiratory, lung macrophages play an important role in controlling the severity of influenza disease by limiting viral replication, modulating the local inflammatory response, and initiating subsequent adaptive immune responses. However, influenza virus reproduction in macrophages is both strain- and macrophage type-dependent, and ineffective replication of some viral strains in mouse macrophages has been observed. This review discusses the function of lung macrophages in influenza virus infection in order to better understand the pathogenesis of the influenza virus.
Collapse
Affiliation(s)
- Haoning Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Aoxue Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yuying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Fanhua Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
2
|
Samaras C, Kyriazopoulou E, Poulakou G, Reiner E, Kosmidou M, Karanika I, Petrakis V, Adamis G, Gatselis NK, Fragkou A, Rapti A, Taddei E, Kalomenidis I, Chrysos G, Bertoli G, Kainis I, Alexiou Z, Castelli F, Saverio Serino F, Bakakos P, Nicastri E, Tzavara V, Kostis E, Dagna L, Koukidou S, Tzatzagou G, Chini M, Bassetti M, Trakatelli C, Tsoukalas G, Selmi C, Samarkos M, Pyrpasopoulou A, Masgala A, Antonakis E, Argyraki A, Akinosoglou K, Sympardi S, Panagopoulos P, Milionis H, Metallidis S, Syrigos KN, Angel A, Dalekos GN, Netea MG, Giamarellos-Bourboulis EJ. Interferon gamma-induced protein 10 (IP-10) for the early prognosis of the risk for severe respiratory failure and death in COVID-19 pneumonia. Cytokine 2023; 162:156111. [PMID: 36529030 PMCID: PMC9747699 DOI: 10.1016/j.cyto.2022.156111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/18/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Elevated concentrations of soluble urokinase plasminogen activator receptor (suPAR) predict progression to severe respiratory failure (SRF) or death among patients with COVID-19 pneumonia and guide early anakinra treatment. As suPAR testing may not be routinely available in every health-care setting, alternative biomarkers are needed. We investigated the performance of C-reactive protein (CRP), interferon gamma-induced protein-10 (IP-10) and TNF-related apoptosis-inducing ligand (TRAIL) for predicting SRF or death in COVID-19. METHODS Two cohorts were studied; one discovery cohort with 534 patients from the SAVE-MORE clinical trial; and one validation cohort with 364 patients from the SAVE trial including also 145 comparators. CRP, IP-10 and TRAIL were measured by the MeMed Key® platform in order to select the biomarker with the best prognostic performance for the early prediction of progression into SRF or death. RESULTS IP-10 had the best prognostic performance: baseline concentrations 2000 pg/ml or higher predicted equally well to suPAR (sensitivity 85.0 %; negative predictive value 96.6 %). Odds ratio for poor outcome among anakinra-treated participants of the SAVE-MORE trial was 0.35 compared to placebo when IP-10 was 2,000 pg/ml or more. IP-10 could divide different strata of severity for SRF/death by day 14 in the validation cohort. Anakinra treatment decreased this risk irrespective the IP-10 concentrations. CONCLUSIONS IP-10 concentrations of 2,000 pg/ml or higher are a valid alternative to suPAR for the early prediction of progression into SRF or death the first 14 days from hospital admission for COVID-19 and they may guide anakinra treatment. TRIAL REGISTRATION CLINICALTRIALS gov, NCT04680949 and NCT04357366.
Collapse
Affiliation(s)
- Charilaos Samaras
- 1(st) Department of Internal Medicine, Asklepieio General Hospital of Voula, Greece
| | - Evdoxia Kyriazopoulou
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Garyfallia Poulakou
- 3(rd) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Maria Kosmidou
- 1st Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Ioanna Karanika
- 1st Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Vasileios Petrakis
- 2(nd) Department of Internal Medicine, Democritus University of Thrace, Medical School, 681 00, Alexandroupolis, Greece
| | - George Adamis
- 1(st) Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Nikolaos K Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, 41110 Larissa, Greece
| | | | - Aggeliki Rapti
- 2(nd) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Eleonora Taddei
- Dipartimento Scienze di Laboratorio e Infettivologiche - Fondazione Policlinico Universitario Agostino Gemelli IRCCS - Roma, Italy
| | - Ioannis Kalomenidis
- 1(st) Department of Critical Care and Pulmonary Medicine, Medical School, National and Kapodistrian University of Athens, Evangelismos General Hospital, Athens, Greece
| | - George Chrysos
- 2(nd) Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Giulia Bertoli
- Department of Infectious - Tropical Diseases and Microbiology, IRCSS Sacro Cuore Hospital, Negrar, Verona, Italy
| | - Ilias Kainis
- 10(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases of Athens, Greece
| | - Zoi Alexiou
- 2(nd) Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Francesco Castelli
- Spedali Civili, Brescia ASST Spedali Civili Hospital, University of Brescia, Italy
| | | | - Petros Bakakos
- 1(st) Department of Chest Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Emanuele Nicastri
- Department of Internal Medicine, Spallanzani Institute of Rome, Italy
| | - Vassiliki Tzavara
- 1(st) Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Evangelos Kostis
- Department of Therapeutics, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS Ospedale San Raffaele & Vita-Salute San Raffaele University, Milan, Italy
| | - Sofia Koukidou
- 5(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Glykeria Tzatzagou
- 1(st) Department of Internal Medicine, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Maria Chini
- 3rd Dpt of Internal Medicine and Infectious Diseases Unit, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Matteo Bassetti
- Infectious Diseases Clinic, Ospedale Policlinico San Martino IRCCS and Department of Health Sciences, University of Genova, Genova, Italy
| | - Christina Trakatelli
- 3(rd) Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - George Tsoukalas
- 4(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele and IRCCS Humanitas Research Hospital, via Manzoni, 56, 20089 Rozzano, Milan, Italy
| | - Michael Samarkos
- 1(st) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Athina Pyrpasopoulou
- 2(nd) Department of Propedeutic Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Aikaterini Masgala
- 2(nd) Department of Internal Medicine, Konstantopouleio General Hospital, Athens, Greece
| | | | - Aikaterini Argyraki
- Department of Internal Medicine, Sotiria General Hospital of Chest Diseases, Greece
| | | | - Styliani Sympardi
- 1(st) Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Periklis Panagopoulos
- 2(nd) Department of Internal Medicine, Democritus University of Thrace, Medical School, 681 00, Alexandroupolis, Greece
| | - Haralampos Milionis
- 1st Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Simeon Metallidis
- 1st Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Konstantinos N Syrigos
- 3(rd) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, 41110 Larissa, Greece
| | - Mihai G Netea
- Department of Internal Medicine and Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Evangelos J Giamarellos-Bourboulis
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
3
|
Influenza B Virus (IBV) Immune-Mediated Disease in C57BL/6 Mice. Vaccines (Basel) 2022; 10:vaccines10091440. [PMID: 36146518 PMCID: PMC9504307 DOI: 10.3390/vaccines10091440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Influenza B viruses (IBV) primarily infect humans, causing seasonal epidemics. The absence of an animal reservoir limits pandemic concern, but IBV infections may cause severe respiratory disease, predominantly in young children and the elderly. The IBV disease burden is largely controlled by seasonal influenza vaccination; however, immunity due to vaccination is sometimes incomplete, a feature linked to antigenic mismatches. Thus, understanding the features that contribute to disease pathogenesis is important, particularly immune-mediated versus virus-mediated outcomes. Unexpectedly, C57BL/6 (B6) mice intranasally infected with a low multiplicity of infection of B/Florida/04/2006 developed substantial morbidity and mortality. To address the cause, B6 mice were treated daily with dexamethasone to dampen the immune and pro-inflammatory response to IBV infection, allowing the determination of whether the responses were immune- and/or virus-associated. As expected, dexamethasone (DEX)-treated mice had a lower pro-inflammatory response and reduced lung pathology despite the presence of high viral lung titers, but mortality was comparable to PBS-treated mice, indicating that mortality may be linked to lung virus replication. The results showed that the immune response to IBV is the major cause of morbidity, mortality, lung pathology, and viral clearance. Importantly, the results suggest that a robust lung CTL response and associated leukocyte influx contribute to disease.
Collapse
|
4
|
AbouAitah K, Swiderska-Sroda A, Kandeil A, Salman AMM, Wojnarowicz J, Ali MA, Opalinska A, Gierlotka S, Ciach T, Lojkowski W. Virucidal Action Against Avian Influenza H5N1 Virus and Immunomodulatory Effects of Nanoformulations Consisting of Mesoporous Silica Nanoparticles Loaded with Natural Prodrugs. Int J Nanomedicine 2020; 15:5181-5202. [PMID: 32801685 PMCID: PMC7398888 DOI: 10.2147/ijn.s247692] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background Combating infectious diseases caused by influenza virus is a major challenge due to its resistance to available drugs and vaccines, side effects, and cost of treatment. Nanomedicines are being developed to allow targeted delivery of drugs to attack specific cells or viruses. Materials and Methods In this study, mesoporous silica nanoparticles (MSNs) functionalized with amino groups and loaded with natural prodrugs of shikimic acid (SH), quercetin (QR) or both were explored as a novel antiviral nanoformulations targeting the highly pathogenic avian influenza H5N1 virus. Also, the immunomodulatory effects were investigated in vitro tests and anti-inflammatory activity was determined in vivo using the acute carrageenan-induced paw edema rat model. Results Prodrugs alone or the MSNs displayed weaker antiviral effects as evidenced by virus titers and plaque formation compared to nanoformulations. The MSNs-NH2-SH and MSNs-NH2-SH-QR2 nanoformulations displayed a strong virucidal by inactivating the H5N1 virus. They induced also strong immunomodulatory effects: they inhibited cytokines (TNF-α, IL-1β) and nitric oxide production by approximately 50% for MSNs-NH2-SH-QR2 (containing both SH and QR). Remarkable anti-inflammatory effects were observed during in vivo tests in an acute carrageenan-induced rat model. Conclusion Our preliminary findings show the potential of nanotechnology for the application of natural prodrug substances to produce a novel safe, effective, and affordable antiviral drug.
Collapse
Affiliation(s)
- Khaled AbouAitah
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland.,Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), P.C.12622, Dokki, Giza, Egypt
| | - Anna Swiderska-Sroda
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Division, National Research Centre (NRC) P.C.12622, Dokki, Giza, Egypt
| | - Asmaa M M Salman
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), P.C. 12622, Dokki, Giza, Egypt
| | - Jacek Wojnarowicz
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Division, National Research Centre (NRC) P.C.12622, Dokki, Giza, Egypt
| | - Agnieszka Opalinska
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Stanislaw Gierlotka
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Ciach
- Biomedical Engineering Laboratory, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Witold Lojkowski
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
5
|
Hayashi H, Okamatsu M, Ogasawara H, Tsugawa N, Isoda N, Matsuno K, Sakoda Y. Oral Supplementation of the Vitamin D Metabolite 25(OH)D 3 Against Influenza Virus Infection in Mice. Nutrients 2020; 12:E2000. [PMID: 32635656 PMCID: PMC7400405 DOI: 10.3390/nu12072000] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
Vitamin D is a fat-soluble vitamin that is metabolized by the liver into 25-hydroxyvitamin D [25(OH)D] and then by the kidney into 1,25-dihydroxyvitamin D [1,25(OH)2D], which activates the vitamin D receptor expressed in various cells, including immune cells, for an overall immunostimulatory effect. Here, to investigate whether oral supplementation of 25-hydroxyvitamin D3 [25(OH)D3], a major form of vitamin D metabolite 25(OH)D, has a prophylactic effect on influenza A virus infection, mice were fed a diet containing a high dose of 25(OH)D3 and were challenged with the influenza virus. In the lungs of 25(OH)D3-fed mice, the viral titers were significantly lower than in the lungs of standardly fed mice. Additionally, the proinflammatory cytokines IL-5 and IFN-γ were significantly downregulated after viral infection in 25(OH)D3-fed mice, while anti-inflammatory cytokines were not significantly upregulated. These results indicate that 25(OH)D3 suppresses the production of inflammatory cytokines and reduces virus replication and clinical manifestations of influenza virus infection in a mouse model.
Collapse
Affiliation(s)
- Hirotaka Hayashi
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan; (H.H.); (M.O.); (K.M.)
| | - Masatoshi Okamatsu
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan; (H.H.); (M.O.); (K.M.)
| | - Honami Ogasawara
- Department of Health and Nutrition, Faculty of Health and Nutrition, Osaka Shoin Woman’s University, Hishiyanishi 4-2-26, Higashiosaka 577-8550, Japan; (H.O.); (N.T.)
| | - Naoko Tsugawa
- Department of Health and Nutrition, Faculty of Health and Nutrition, Osaka Shoin Woman’s University, Hishiyanishi 4-2-26, Higashiosaka 577-8550, Japan; (H.O.); (N.T.)
| | - Norikazu Isoda
- Unit of Risk Analysis and Management, Research Center for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo 001-0020, Japan;
- Global Station of Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
| | - Keita Matsuno
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan; (H.H.); (M.O.); (K.M.)
- Global Station of Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan; (H.H.); (M.O.); (K.M.)
- Global Station of Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
| |
Collapse
|
6
|
Blum SI, Tse HM. Innate Viral Sensor MDA5 and Coxsackievirus Interplay in Type 1 Diabetes Development. Microorganisms 2020; 8:microorganisms8070993. [PMID: 32635205 PMCID: PMC7409145 DOI: 10.3390/microorganisms8070993] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is a polygenic autoimmune disease characterized by immune-mediated destruction of insulin-producing β-cells. The concordance rate for T1D in monozygotic twins is ≈30-50%, indicating that environmental factors also play a role in T1D development. Previous studies have demonstrated that enterovirus infections such as coxsackievirus type B (CVB) are associated with triggering T1D. Prior to autoantibody development in T1D, viral RNA and antibodies against CVB can be detected within the blood, stool, and pancreata. An innate pathogen recognition receptor, melanoma differentiation-associated protein 5 (MDA5), which is encoded by the IFIH1 gene, has been associated with T1D onset. It is unclear how single nucleotide polymorphisms in IFIH1 alter the structure and function of MDA5 that may lead to exacerbated antiviral responses contributing to increased T1D-susceptibility. Binding of viral dsRNA via MDA5 induces synthesis of antiviral proteins such as interferon-alpha and -beta (IFN-α/β). Viral infection and subsequent IFN-α/β synthesis can lead to ER stress within insulin-producing β-cells causing neo-epitope generation, activation of β-cell-specific autoreactive T cells, and β-cell destruction. Therefore, an interplay between genetics, enteroviral infections, and antiviral responses may be critical for T1D development.
Collapse
|
7
|
Kostinov MP, Akhmatova NK, Khromova EA, Kostinova AM. Cytokine Profile in Human Peripheral Blood Mononuclear Leukocytes Exposed to Immunoadjuvant and Adjuvant-Free Vaccines Against Influenza. Front Immunol 2020; 11:1351. [PMID: 32695114 PMCID: PMC7339108 DOI: 10.3389/fimmu.2020.01351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/27/2020] [Indexed: 12/22/2022] Open
Abstract
Background: In the last decade, adjuvant-containing vaccines, exerting different effects on the immune system, including the production of cytokines, which are one of the most important regulatory systems of the body, are introduced into practice. Objectives: An effect of the immunoadjuvant polymer-subunit and adjuvant-free vaccines against influenza on the cytokine profile of mononuclear leukocytes in 27 healthy women was studied. Methods: The study of cytokine profile in human peripheral blood mononuclear leukocytes exposed to vaccines against influenza virus was determined by flow cytometry method (Cytomix FC-500, Beckman Coulter, USA) using the Multiplex-13 test system (Bender MedSystems, Austria). Results: It was established that all the studied vaccines leaded to somewhat increased levels of Th1/Th2/Th17/Th9/Th22 cytokines in the culture fluid of peripheral blood mononuclear leukocytes (PBML), which indicates the activation of both humoral and cellular immunity. An immunoadjuvant vaccine has been shown to be superior in activating the synthesis of Th1 (IL-12, INF-g, IL-2, IL-6, IL-1β, TNF-α) cytokines, IL-9 and IL-22, while the subunit vaccine was superior in activating the synthesis of IL-4, and split vaccine-of IL-5. Conclusions: Immunoadjuvant vaccine is superior in terms of inducing cellular immune effectors to a greater extent compared to subunit and split vaccines.
Collapse
Affiliation(s)
- Mikhail Petrovich Kostinov
- Department of Immunology, I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
- Department of Immunology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nelli Kimovna Akhmatova
- Department of Immunology, I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | | | | |
Collapse
|
8
|
Chang FY, Chen HC, Chen PJ, Ho MS, Hsieh SL, Lin JC, Liu FT, Sytwu HK. Immunologic aspects of characteristics, diagnosis, and treatment of coronavirus disease 2019 (COVID-19). J Biomed Sci 2020; 27:72. [PMID: 32498686 PMCID: PMC7270518 DOI: 10.1186/s12929-020-00663-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022] Open
Abstract
On March 11, 2020, the World Health Organization declared the worldwide spread of the infectious disease COVID-19, caused by a new strain of coronavirus, SARS-CoV-2, as a pandemic. Like in all other infectious diseases, the host immune system plays a key role in our defense against SARS-CoV-2 infection. However, viruses are able to evade the immune attack and proliferate and, in susceptible individuals, cause severe inflammatory response known as cytokine storm, particularly in the lungs. The advancement in our understanding of the mechanisms underlying the host immune responses promises to facilitate the development of approaches for prevention or treatment of diseases. Components of immune system, such as antibodies, can also be used to develop sensitive and specific diagnostic methods as well as novel therapeutic agents. In this review, we summarize our knowledge about how the host mounts immune responses to infection by SARS-CoV-2. We also describe the diagnostic methods being used for COVID-19 identification and summarize the current status of various therapeutic strategies, including vaccination, being considered for treatment of the disease.
Collapse
Affiliation(s)
- Feng-Yee Chang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsiang-Cheng Chen
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Jer Chen
- Division of Gastroenterology, Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Shang Ho
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, Taiwan
| | | | - Jung-Chung Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, Taiwan.
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| |
Collapse
|
9
|
Labzin LI, Bottermann M, Rodriguez‐Silvestre P, Foss S, Andersen JT, Vaysburd M, Clift D, James LC. Antibody and DNA sensing pathways converge to activate the inflammasome during primary human macrophage infection. EMBO J 2019; 38:e101365. [PMID: 31468569 PMCID: PMC6826209 DOI: 10.15252/embj.2018101365] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 01/07/2023] Open
Abstract
Inflammasomes are potent innate immune signalling complexes that couple cytokine release with pro-inflammatory cell death. However, pathogens have evolved strategies to evade this cell autonomous system. Here, we show how antibodies combine with innate sensors in primary human macrophages to detect viral infection and activate the inflammasome. Our data demonstrate that antibody opsonisation of virions can activate macrophages in multiple ways. In the first, antibody binding of adenovirus causes lysosomal damage, activating NLRP3 to drive inflammasome formation and IL-1β release. Importantly, this mechanism enhances virion capture but not infection and is accompanied by cell death, denying the opportunity for viral replication. Unexpectedly, we also find that antibody-coated viruses, which successfully escape into the cytosol, trigger a second system of inflammasome activation. These viruses are intercepted by the cytosolic antibody receptor TRIM21 and the DNA sensor cGAS. Together, these sensors stimulate both NLRP3 inflammasome formation and NFκB activation, driving dose-dependent IL-1β and TNF secretion, without inducing cell death. Our data highlight the importance of cooperativity between multiple sensing networks to expose viruses to the inflammasome pathway, which is particularly important for how our innate immune system responds to infection in the presence of pre-existing immunity.
Collapse
Affiliation(s)
- Larisa I Labzin
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| | - Maria Bottermann
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| | - Pablo Rodriguez‐Silvestre
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| | - Stian Foss
- Centre for Immune Regulation (CIR)Department of BiosciencesUniversity of OsloOsloNorway
| | - Jan Terje Andersen
- CIR and Department of ImmunologyOslo University Hospital RikshospitaletOsloNorway
- Department of PharmacologyInstitute of Clinical MedicineOslo University HospitalUniversity of OsloOsloNorway
| | - Marina Vaysburd
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| | - Dean Clift
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| | - Leo C James
- Protein and Nucleic Acid Chemistry DivisionMedical Research CouncilLaboratory of Molecular BiologyCambridgeUK
| |
Collapse
|
10
|
Yan W, Zheng C, He J, Zhang W, Huang XA, Li X, Wang Y, Wang X. Eleutheroside B1 mediates its anti-influenza activity through POLR2A and N-glycosylation. Int J Mol Med 2018; 42:2776-2792. [PMID: 30226535 PMCID: PMC6192727 DOI: 10.3892/ijmm.2018.3863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/04/2018] [Indexed: 12/26/2022] Open
Abstract
Influenza viruses represent a serious threat to human health. Although our research group has previously demonstrated the antiviral and anti-inflammatory activities of eleutheroside B1, a detailed explanation of the mechanism by which it is effective against the influenza virus remains to be elucidated. In the present study, the transcriptomic responses of influenza A virus-infected lung epithelial cells (A549) treated with eleutheroside B1 were investigated using high-throughput RNA sequencing, and potential targets were identified using a molecular docking technique, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay, and DNA methylation analysis. The transcriptomic data revealed that there are 1,871 differentially expressed genes (DEGs) between the cells infected with the influenza virus strain variant PR8, and the cells infected with PR8 and treated with eleutheroside B1. Among the DEGs, RNA polymerase II subunit A (POLR2A; encoding the largest subunit of RNA polymerase II) and mannosidase α class II member 1 (MAN2A1) were selected from the molecular docking analysis with eleutheroside B1. The docking score of Drosophila melanogaster MAN2A1 (3BVT) was 11.3029, whereas that of POLR2A was 9.0133. The RT-qPCR results demonstrated that the expression levels of host genes (MAN2A2, POLR2A) and viral genes (PA, PB1, PB2, HA) were downregulated following eleutheroside B1 treatment. Bisulfite-sequencing PCR was performed to investigate whether eleutheroside B1 was able to modify the DNA methylation of POLR2A, and the results suggested that the average proportion of methylated CpGs (-222-72 bp) increased significantly following treatment with eleutheroside B1. Taken together, these findings suggested that eleutheroside B1 may affect N-glycan biosynthesis, the chemokine signaling pathway, cytokine-cytokine receptor interaction and, in particular, may target the POLR2A to inhibit the production of influenza virus genes.
Collapse
Affiliation(s)
- Wen Yan
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Chunge Zheng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Jiayang He
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Wenjie Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Xin-An Huang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Xiong Li
- Department of Integrated Chinese Medicine Immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yutao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Xinhua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
11
|
Jung YJ, Lee YT, Ngo VL, Cho YH, Ko EJ, Hong SM, Kim KH, Jang JH, Oh JS, Park MK, Kim CH, Sun J, Kang SM. Heat-killed Lactobacillus casei confers broad protection against influenza A virus primary infection and develops heterosubtypic immunity against future secondary infection. Sci Rep 2017; 7:17360. [PMID: 29234060 PMCID: PMC5727132 DOI: 10.1038/s41598-017-17487-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/23/2017] [Indexed: 02/08/2023] Open
Abstract
Lactic acid bacteria (LAB) are the common probiotics. Here, we investigated the antiviral protective effects of heat-killed LAB strain Lactobacillus casei DK128 (DK128) on influenza viruses. Intranasal treatment of mice with DK128 conferred protection against different subtypes of influenza viruses by lessening weight loss and lowering viral loads. Protection via heat-killed DK128 was correlated with an increase in alveolar macrophage cells in the lungs and airways, early induction of virus specific antibodies, reduced levels of pro-inflammatory cytokines and innate immune cells. Importantly, the mice that were protected against primary viral infection as a result of heat-killed DK128 pretreatment developed subsequent heterosubtypic immunity against secondary virus infection. For protection against influenza virus via heat-killed DK128 pretreatment, B cells and partially CD4 T cells but not CD8 T cells were required as inferred from studies using knockout mouse models. Our study provides insight into how hosts can be equipped with innate and adaptive immunity via heat-killed DK128 treatment to protect against influenza virus, supporting that heat-killed LAB may be developed as anti-virus probiotics.
Collapse
Affiliation(s)
- Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Vu Le Ngo
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Young-Hee Cho
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Sung-Moon Hong
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Ji-Hun Jang
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, 331-858, Korea
| | - Joon-Suk Oh
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, 331-858, Korea
| | - Min-Kyung Park
- Department of Human Nutrition and Food Science, Chungwoon University, Namjang-Ri, Hongsung-Eup, Hongsung-Kun, Chungnam, 350-701, Korea
| | - Cheol-Hyun Kim
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA.
| |
Collapse
|
12
|
Cline TD, Beck D, Bianchini E. Influenza virus replication in macrophages: balancing protection and pathogenesis. J Gen Virol 2017; 98:2401-2412. [PMID: 28884667 DOI: 10.1099/jgv.0.000922] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Macrophages are essential for protection against influenza A virus infection, but are also implicated in the morbidity and mortality associated with severe influenza disease, particularly during infection with highly pathogenic avian influenza (HPAI) H5N1 virus. While influenza virus infection of macrophages was once thought to be abortive, it is now clear that certain virus strains can replicate productively in macrophages. This may have important consequences for the antiviral functions of macrophages, the course of disease and the outcome of infection for the host. In this article, we review findings related to influenza virus replication in macrophages and the impact of productive replication on macrophage antiviral functions. A clear understanding of the interactions between influenza viruses and macrophages may lead to new antiviral therapies to relieve the burden of severe disease associated with influenza viruses.
Collapse
Affiliation(s)
- Troy D Cline
- Department of Biological Sciences, California State University, Chico, California, USA
| | - Donald Beck
- Department of Biological Sciences, California State University, Chico, California, USA
| | - Elizabeth Bianchini
- Department of Biological Sciences, California State University, Chico, California, USA
| |
Collapse
|
13
|
Taye B, Chen H, Myaing MZ, Tan BH, Maurer-Stroh S, Sugrue RJ. Systems-based approach to examine the cytokine responses in primary mouse lung macrophages infected with low pathogenic avian Influenza virus circulating in South East Asia. BMC Genomics 2017; 18:420. [PMID: 28558796 PMCID: PMC5450074 DOI: 10.1186/s12864-017-3803-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 05/17/2017] [Indexed: 02/08/2023] Open
Abstract
Background Influenza A virus (IAV) is a major public health concern, being responsible for the death of approximately half a million people each year. Zoonotic transmissions of the virus from swine and avian origin have occurred in the past, and can potentially lead to the emgergence of new IAV stains in future pandemics. Pulmonary macrophages have been implicated in disease severity in the lower airway, and understanding the host response of macrophages infected with avian influenza viruses should provide new therapeutic strategies. Results We used a systems-based approach to investigate the transcriptome response of primary murine lung macrophages (PMФ) infected with the mouse-adapted H1N1/WSN virus and low pathogenic avian influenza (LPAI) viruses H5N2 and H5N3. The results showed that the LPAI viruses H5N2 and H5N3 can infect PMФ with similar efficiency to the H1N1/WSN virus. While all viruses induced antiviral responses, the H5N3 virus infection resulted in higher expression levels of cytokines and chemokines associated with inflammatory responses. Conclusions The LPAI H5N2 and H5N3 viruses are able to infect murine lung macrophages. However, the H5N3 virus was associated with increased expression of pro-inflammatory mediators. Although the H5N3 virus it is capable of inducing high levels of cytokines that are associated with inflammation, this property is distinct from its inability to efficiently replicate in a mammalian host. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3803-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Biruhalem Taye
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01, Matrix, Singapore, 138671, Republic of Singapore.,School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.,Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, P.O.BOX 1176, Ethiopia
| | - Hui Chen
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.,Current address Genome Institute of Singapore, A*STAR, 60 Biopolis Street, #02-01, Genome, Singapore, 138672, Republic of Singapore
| | - Myint Zu Myaing
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, Defence Science Organisation National Laboratories, 27 Medical Drive, Singapore, 117510, Republic of Singapore.,LKC School of Medicine, Nanyang Technological University, 50 Nanyang Ave, Singapore, 639798, Republic of Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01, Matrix, Singapore, 138671, Republic of Singapore.,National Public Health Laboratory, Ministry of Health, Singapore, Republic of Singapore.,Department of Biological Sciences, National University of Singapore, 8 Medical Drive, Singapore, 117597, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.
| |
Collapse
|
14
|
Zhou B, Li J, Liang X, Yang Z, Jiang Z. Transcriptome profiling of influenza A virus-infected lung epithelial (A549) cells with lariciresinol-4-β-D-glucopyranoside treatment. PLoS One 2017; 12:e0173058. [PMID: 28273165 PMCID: PMC5342222 DOI: 10.1371/journal.pone.0173058] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
The influenza A virus is an acute contagious pathogen that affects the human respiratory system and can cause severe lung disease and even death. Lariciresinol-4-β-D-glucopyranoside is a lignan that is extracted from Isatis indigotica, which is a medicinal herb plant that was commonly applied to treat infections, the common cold, fever and inflammatory diseases. Our previous study demonstrated that lariciresinol-4-β-D-glucopyranoside possesses anti-viral and anti-inflammatory properties. However, the comprehensive and detailed mechanisms that underlie the effect of lariciresinol-4-β-D-glucopyranoside interventions against influenza virus infection remain to be elucidated. In this study, we employed high-throughput RNA sequencing (RNA-seq) to investigate the transcriptomic responses of influenza A virus-infected lung epithelial (A549) cells with lariciresinol-4-β-D-glucopyranoside treatment. The transcriptome data show that infection with influenza A virus prompted the activation of 368 genes involved in RIG-I signalling, the inflammatory response, interferon α/β signalling and gene expression that was not affected by lariciresinol-4-β-D-glucopyranoside treatment. Lariciresinol-4-β-D-glucopyranoside exerted its pharmacological actions on the immune system, signal transduction, cell cycle and metabolism, which may be an underlying defense mechanism against influenza virus infection. In addition, 166 differentially expressed genes (DEGs) were uniquely expressed in lariciresinol-4-β-D-glucopyranoside-treated cells, which were concentrated in the cell cycle, DNA repair, chromatin organization, gene expression and biosynthesis domains. Among them, six telomere-associated genes were up-regulated by lariciresinol-4-β-D-glucopyranoside treatment, which have been implicated in telomere regulation and stability. Collectively, we employed RNA-seq analysis to provide comprehensive insight into the mechanism of lariciresinol-4-β-D-glucopyranoside against influenza virus infection.
Collapse
Affiliation(s)
- Beixian Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Jing Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiaoli Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Zifeng Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
- * E-mail: (ZFY); (ZHJ)
| | - Zhihong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
- * E-mail: (ZFY); (ZHJ)
| |
Collapse
|
15
|
Influenza Virus Overcomes Cellular Blocks To Productively Replicate, Impacting Macrophage Function. J Virol 2017; 91:JVI.01417-16. [PMID: 27807237 DOI: 10.1128/jvi.01417-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/27/2016] [Indexed: 11/20/2022] Open
Abstract
Whether influenza virus replication in macrophages is productive or abortive has been a topic of debate. Utilizing a panel of 28 distinct human, avian, and swine influenza viruses, we found that only a small subset can overcome cellular blocks to productively replicate in murine and primary human macrophages. Murine macrophages have two cellular blocks. The first block is during viral entry, where virions with relatively acid-stable hemagglutinin (HA) proteins are rendered incapable of pH-induced triggering for membrane fusion, resulting in lysosomal degradation. The second block is downstream of viral replication but upstream of late protein synthesis. In contrast, primary human macrophages only have one cellular block that occurs after late protein synthesis. To determine the impact of abortive replication at different stages of the viral life cycle or productive replication on macrophage function, we assessed cytotoxicity, nitric oxide or reactive oxygen species production, and phagocytosis. Intriguingly, productive viral replication decreased phagocytosis of IgG-opsonized bioparticles and Fc receptor CD16 and CD32 surface levels, a function, to our knowledge, never before reported for an RNA virus. These data suggest that replication in macrophages affects cellular function and plays an important role in pathogenesis during infection in vivo IMPORTANCE: Macrophages are a critical first line of defense against respiratory pathogens. Thus, understanding how viruses evade or exploit macrophage function will provide greater insight into viral pathogenicity and antiviral responses. We previously showed that only a subset of highly pathogenic avian (HPAI) H5N1 influenza virus strains could productively replicate in murine macrophages through a hemagglutinin (HA)-mediated mechanism. These studies expand upon this work and demonstrate that productive replication is not specific to unique HPAI H5N1 viruses; an H1N1 strain (A/WSN/33) can also replicate in macrophages. Importantly, we identify two cellular blocks limiting replication that can be overcome by an avian-like pH of activation for nuclear entry and a yet-to-be-identified mechanism(s) to overcome a postnuclear entry block. Overcoming these blocks reduces the cell's ability to phagocytose IgG-opsonized bioparticles by decreasing Fc receptor surface levels, a mechanism previously thought to occur during bacterial and DNA viral infections.
Collapse
|
16
|
Infection of Mouse Macrophages by Seasonal Influenza Viruses Can Be Restricted at the Level of Virus Entry and at a Late Stage in the Virus Life Cycle. J Virol 2015; 89:12319-29. [PMID: 26423941 DOI: 10.1128/jvi.01455-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/22/2015] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Airway epithelial cells are susceptible to infection with seasonal influenza A viruses (IAV), resulting in productive virus replication and release. Macrophages (MΦ) are also permissive to IAV infection; however, virus replication is abortive. Currently, it is unclear how productive infection of MΦ is impaired or the extent to which seasonal IAV replicate in MΦ. Herein, we compared mouse MΦ and epithelial cells for their ability to support genomic replication and transcription, synthesis of viral proteins, assembly of virions, and release of infectious progeny following exposure to genetically defined IAV. We confirm that seasonal IAV differ in their ability to utilize cell surface receptors for infectious entry and that this represents one level of virus restriction. Following virus entry, we demonstrate synthesis of all eight segments of genomic viral RNA (vRNA) and mRNA, as well as seven distinct IAV proteins, in IAV-infected mouse MΦ. Although newly synthesized hemagglutinin (HA) and neuraminidase (NA) glycoproteins are incorporated into the plasma membrane and expressed at the cell surface, electron microscopy confirmed that virus assembly was defective in IAV-infected MΦ, defining a second level of restriction late in the virus life cycle. IMPORTANCE Seasonal influenza A viruses (IAV) and highly pathogenic avian influenza viruses (HPAI) infect macrophages, but only HPAI replicate productively in these cells. Herein, we demonstrate that impaired virus uptake into macrophages represents one level of restriction limiting infection by seasonal IAV. Following uptake, seasonal IAV do not complete productive replication in macrophages, representing a second level of restriction. Using murine macrophages, we demonstrate that productive infection is blocked late in the virus life cycle, such that virus assembly is defective and newly synthesized virions are not released. These studies represent an important step toward identifying host-encoded factors that block replication of seasonal IAV, but not HPAI, in macrophages.
Collapse
|
17
|
Jang H, Koo BS, Jeon EO, Lee HR, Lee SM, Mo IP. Altered pro-inflammatory cytokine mRNA levels in chickens infected with infectious bronchitis virus. Poult Sci 2013; 92:2290-8. [PMID: 23960111 PMCID: PMC7194964 DOI: 10.3382/ps.2013-03116] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Infectious bronchitis virus (IBV) replicates primarily in the respiratory tract and grows in various organs in chickens, with or without pathological effects. The diversity of this virus has been verified by sequence analysis of the S1 glycoprotein gene, but this method must be supplemented with further analysis for characterization of the agent. To increase our understanding of the pathogenesis of the disease caused by this virus, we investigated the response of chickens to 2 IBV with different genotypes, KIIa and ChVI. The clinical signs induced by the viruses were observed. In addition, the mRNA levels of the pro-inflammatory cytokines, IL-6, IL-1β, and lipopolysaccharide-induced tumor necrosis factor-α factor and the serum levels of α1-acid glycoprotein, which is a major acute phase protein, were measured. The KIIa genotype (Kr/ADL110002/2011) induced clinical signs accompanied by the excessive production of pro-inflammatory cytokines and a higher viral load. In chickens infected with this isolate, simultaneous peaks in the viral copy number and cytokine production were observed at 7 dpi in the trachea and 9 d postinoculation in the kidney. On the other hand, the chickens infected with the ChVI genotype (Kr/ADL120003/2012) did not show a response other than a mild upregulation of cytokines at 1 d postinoculation, which appears to indicate the invasion of the virus. In summary, we confirmed a differential innate response following infection with distinct IBV. We hypothesize that an excessive innate response contributes to the scale of the pathophysiologic effect in chickens.
Collapse
Affiliation(s)
- Hyesun Jang
- Preventive Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University 12, Gaeshin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea
| | | | | | | | | | | |
Collapse
|