51
|
Walaza S, Cohen C, Tempia S, Moyes J, Nguweneza A, Madhi SA, McMorrow M, Cohen AL. Influenza and tuberculosis co-infection: A systematic review. Influenza Other Respir Viruses 2019; 14:77-91. [PMID: 31568678 PMCID: PMC6928059 DOI: 10.1111/irv.12670] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Introduction There are limited data on risk of severe disease or outcomes in patients with influenza and pulmonary tuberculosis (PTB) co‐infection compared to those with single infection. Methods We conducted a systematic review of published literature on the interaction of influenza viruses and PTB. Studies were eligible for inclusion if they presented data on prevalence, disease association, presentation or severity of laboratory‐confirmed influenza among clinically diagnosed or laboratory‐confirmed PTB cases. We searched eight databases from inception until December 2018. Summary characteristics of each study were extracted, and a narrative summary was presented. Cohort or case‐control studies were assessed for potential bias using the Newcastle‐Ottawa scale. Results We assessed 5154 abstracts, reviewed 146 manuscripts and included 19 studies fulfilling selection criteria (13 human and six animal). Of seven studies reporting on the possible effect of the underlying PTB disease in patients with influenza, three of four analytical studies reported no association with disease severity of influenza infection in those with PTB, whilst one study reported PTB as a risk factor for influenza‐associated hospitalization. An association between influenza infection and PTB disease was found in three of five analytical studies; whereas the two other studies reported a high frequency of PTB disease progression and complications among patients with seasonal influenza co‐infection. Conclusion Human analytical studies of an association between co‐infection and severe influenza‐ or PTB‐associated disease or increased prevalence of influenza co‐infection in individuals' hospitalized for PTB were not conclusive. Data are limited from large, high‐quality, analytical epidemiological studies with laboratory‐confirmed endpoints.
Collapse
Affiliation(s)
- Sibongile Walaza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stefano Tempia
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa
| | - Jocelyn Moyes
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Athermon Nguweneza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Shabir A Madhi
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Meredith McMorrow
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa.,U.S. Public Health Service, Rockville, MD, USA
| | - Adam L Cohen
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa.,Global Immunization Monitoring and Surveillance, Expanded Programme on Immunization Department of Immunization, Vaccines and Biologicals World Health Organization, Geneva, Switzerland
| |
Collapse
|
52
|
Antibody Treatment against Angiopoietin-Like 4 Reduces Pulmonary Edema and Injury in Secondary Pneumococcal Pneumonia. mBio 2019; 10:mBio.02469-18. [PMID: 31164474 PMCID: PMC6550533 DOI: 10.1128/mbio.02469-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Secondary bacterial lung infection by Streptococcus pneumoniae (S. pneumoniae) poses a serious health concern, especially in developing countries. We posit that the emergence of multiantibiotic-resistant strains will jeopardize current treatments in these regions. Deaths arising from secondary infections are more often associated with acute lung injury, a common consequence of hypercytokinemia, than with the infection per se Given that secondary bacterial pneumonia often has a poor prognosis, newer approaches to improve treatment outcomes are urgently needed to reduce the high levels of morbidity and mortality. Using a sequential dual-infection mouse model of secondary bacterial lung infection, we show that host-directed therapy via immunoneutralization of the angiopoietin-like 4 c-isoform (cANGPTL4) reduced pulmonary edema and damage in infected mice. RNA sequencing analysis revealed that anti-cANGPTL4 treatment improved immune and coagulation functions and reduced internal bleeding and edema. Importantly, anti-cANGPTL4 antibody, when used concurrently with either conventional antibiotics or antipneumolysin antibody, prolonged the median survival of mice compared to monotherapy. Anti-cANGPTL4 treatment enhanced immune cell phagocytosis of bacteria while restricting excessive inflammation. This modification of immune responses improved the disease outcomes of secondary pneumococcal pneumonia. Taken together, our study emphasizes that host-directed therapeutic strategies are viable adjuncts to standard antimicrobial treatments.IMPORTANCE Despite extensive global efforts, secondary bacterial pneumonia still represents a major cause of death in developing countries and is an important cause of long-term functional disability arising from lung tissue damage. Newer approaches to improving treatment outcomes are needed to reduce the significant morbidity and mortality caused by infectious diseases. Our study, using an experimental mouse model of secondary S. pneumoniae infection, shows that a multimodal treatment that concurrently targets host and pathogen factors improved lung tissue integrity and extended the median survival time of infected mice. The immunoneutralization of host protein cANGPTL4 reduced the severity of pulmonary edema and damage. We show that host-directed therapeutic strategies as well as neutralizing antibodies against pathogen virulence factors are viable adjuncts to standard antimicrobial treatments such as antibiotics. In view of their different modes of action compared to antibiotics, concurrent immunotherapies using antibodies are potentially efficacious against secondary pneumococcal pneumonia caused by antibiotic-resistant pathogens.
Collapse
|
53
|
Peroxisome Proliferator-Activated Receptor Gamma (PPAR) Suppresses Inflammation and Bacterial Clearance during Influenza-Bacterial Super-Infection. Viruses 2019; 11:v11060505. [PMID: 31159430 PMCID: PMC6630660 DOI: 10.3390/v11060505] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 01/21/2023] Open
Abstract
Influenza virus is among the most common causes of respiratory illness worldwide and can be complicated by secondary bacterial pneumonia, a frequent cause of mortality. When influenza virus infects the lung, the innate immune response is activated, and interferons and inflammatory mediators are released. This "cytokine storm" is thought to play a role in influenza-induced lung pathogenesis. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear hormone receptor super-family. PPARγ has numerous functions including enhancing lipid and glucose metabolism and cellular differentiation and suppressing inflammation. Synthetic PPARγagonists (thiazolidinediones or glitazones) have been used clinically in the treatment of type II diabetes. Using data from the National Health and Nutrition Examination Survey (NHANES), diabetic participants taking rosiglitazone had an increased risk of mortality from influenza/pneumonia compared to those not taking the drug. We examined the effect of rosiglitazone treatment during influenza and secondary bacterial (Methicillin resistant Staphylococcus aureus) pneumonia in mice. We found decreased influenza viral burden, decreased numbers of neutrophils and macrophages in bronchoalveolar lavage, and decreased production of cytokines and chemokines in influenza infected, rosiglitazone-treated mice when compared to controls. However, rosiglitazone treatment compromised bacterial clearance during influenza-bacterial super-infection. Both human and mouse data suggest that rosiglitazone treatment worsens the outcome of influenza-associated pneumonia.
Collapse
|
54
|
Paget C, Trottein F. Mechanisms of Bacterial Superinfection Post-influenza: A Role for Unconventional T Cells. Front Immunol 2019; 10:336. [PMID: 30881357 PMCID: PMC6405625 DOI: 10.3389/fimmu.2019.00336] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Despite the widespread application of vaccination programs and antiviral drug treatments, influenza viruses are still among the most harmful human pathogens. Indeed, influenza results in significant seasonal and pandemic morbidity and mortality. Furthermore, severe bacterial infections can occur in the aftermath of influenza virus infection, and contribute substantially to the excess morbidity and mortality associated with influenza. Here, we review the main features of influenza viruses and current knowledge about the mechanical and immune mechanisms that underlie post-influenza secondary bacterial infections. We present the emerging literature describing the role of "innate-like" unconventional T cells in post-influenza bacterial superinfection. Unconventional T cell populations span the border between the innate and adaptive arms of the immune system, and are prevalent in mucosal tissues (including the airways). They mainly comprise Natural Killer T cells, mucosal-associated invariant T cells and γδ T cells. We provide an overview of the principal functions that these cells play in pulmonary barrier functions and immunity, highlighting their unique ability to sense environmental factors and promote protection against respiratory bacterial infections. We focus on two major opportunistic pathogens involved in superinfections, namely Streptococcus pneumoniae and Staphylococcus aureus. We discuss mechanisms through which influenza viruses alter the antibacterial activity of unconventional T cells. Lastly, we discuss recent fundamental advances and possible therapeutic approaches in which unconventional T cells would be targeted to prevent post-influenza bacterial superinfections.
Collapse
Affiliation(s)
- Christophe Paget
- Centre d'Etude des Pathologies Respiratoires, Institut National de la Santé et de la Recherche Médicale U1100, Tours, France.,Faculty of Medicine, Université de Tours, Tours, France
| | - François Trottein
- U1019-UMR 8204-CIIL-Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Centre Hospitalier, Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
55
|
Fischer KJ, Yajjala VK, Bansal S, Bauer C, Chen R, Sun K. Monocytes Represent One Source of Bacterial Shielding from Antibiotics following Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:2027-2034. [PMID: 30745458 DOI: 10.4049/jimmunol.1801471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/26/2019] [Indexed: 12/24/2022]
Abstract
Methicillin-resistant Staphylococcus aureus has emerged as a significant contributor to morbidity and mortality associated with influenza infection. In this study, we show in a mouse model that preceding influenza infection promotes S. aureus resistance to killing by antibiotics. This resistance coincides with influenza-induced accumulation of inflammatory monocytes in the lung. CCR type 2 (CCR2) is responsible for pulmonary monocyte recruitment after influenza infection. We found that antibiotic-treated Ccr2-deficient (Ccr2-/-) mice exhibit significantly improved bacterial control and survival from influenza and methicillin-resistant S. aureus coinfection, despite a delay in viral clearance. Mechanistically, our results from in vivo studies indicate that influenza-induced monocytes serve as reservoirs for intracellular S. aureus survival, thereby promoting bacterial resistance to antibiotic treatment. Blocking CCR2 with a small molecular inhibitor (PF-04178903), in conjunction with antibiotic treatment, enhanced lung bacterial clearance and significantly improved animal survival. Collectively, our study demonstrates that inflammatory monocytes constitute an important and hitherto underappreciated mechanism of the conflicting immune requirements for viral and bacterial clearance by hosts, which subsequently leads to exacerbated outcomes of influenza and S. aureus coinfection.
Collapse
Affiliation(s)
- Karl J Fischer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Vijaya Kumar Yajjala
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Shruti Bansal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Christopher Bauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Ruiling Chen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Keer Sun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| |
Collapse
|
56
|
Mazel-Sanchez B, Yildiz S, Schmolke M. Ménage à trois: Virus, Host, and Microbiota in Experimental Infection Models. Trends Microbiol 2019; 27:440-452. [PMID: 30638775 DOI: 10.1016/j.tim.2018.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 12/22/2022]
Abstract
Infections of mammals with pathogenic viruses occur mostly in the polymicrobial environment of mucosal surfaces or the skin. In recent years our understanding of immune modulation by the commensal microbiota has increased dramatically. The microbiota is today accepted as the prime educator and maintainer of innate and adaptive immune functions. It became further apparent that some viral pathogens profit from the presence of commensal bacteria and their metabolites, especially in the intestinal tract. We further learned that the composition and abundance of the microbiota can change as a consequence of acute and chronic viral infections. Here we discuss recent developments in our understanding of the triangular relationship of virus, host, and microbiota under experimental infection settings.
Collapse
Affiliation(s)
- Beryl Mazel-Sanchez
- Department of Microbiology and Molecular Medicine, University of Geneva, Switzerland
| | - Soner Yildiz
- Department of Microbiology and Molecular Medicine, University of Geneva, Switzerland
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine, University of Geneva, Switzerland.
| |
Collapse
|
57
|
Ayegbusi OT, Ajagbe OA, Afowowe TO, Aransi AT, Olusola BA, Awogbindin IO, Ogunsemowo OO, Faneye AO, Odaibo GN, Olaleye DO. Virus genes and host correlates of pathology are markedly reduced during respiratory syncytial and influenza virus co-infection in BALB/c mice. Heliyon 2019; 5:e01094. [PMID: 30623128 PMCID: PMC6319304 DOI: 10.1016/j.heliyon.2018.e01094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/14/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022] Open
Abstract
Globally, influenza A virus (IAV) and respiratory syncytial virus (RSV) infection remain very high. There is also a high burden of IAV and RSV co-infection in developing countries. To develop universally protective vaccines against these infections, it is imperative that viral genes and immune correlates of pathology are elucidated. As such, we profiled virus genes expressions, histopathology and immunological responses of BALB/c mice infected with RSV and/or IAV in this study. RSV A2 and/or influenza A/H3N2/Perth/16/09 (Pr/H3N2) were induced over a seven-day period in BALB/c mice. Anaesthetized BALB/c mice (12-14 g) were divided into six groups (15-20 mice per group), inoculated with 32 μl each of 3LD50 Pr/H3N2 and/or 100 TCID50 RSV. Two groups (R or I) received RSV or Pr/H3N2 intranasally. Prior infection with either RSV or Pr/H3N2 was followed with a second challenge of the other virus 24 hours post inoculation in RI and IR groups. Another set was exposed to the two viruses simultaneously (I + R group) while the last group served as healthy controls. Five to seven mice per group were euthanized at days 2, 4 and 7. Lung and spleen organs were harvested for virus genes quantitation and immune cells phenotyping respectively. I + R group showed progressive downregulation of RSV F, G, NS1 and NS2 genes. IAV PB2 and M genes had high fold increase on day 2 and 4 post infections. However, by day 7 post infection, M and PB2 fold increase was lower. Also, increased proportions of NKT and T cell subsets were observed throughout the period in I + R group. Conversely, I group was characterized by reduced NKT cell counts and enhanced CD8 T cells levels while R group only showed an increased proportion of CD8 T cells towards the peak of infection. This study shows that RSV and IAV co-infection lead to reduced virulence and pathology compared to single infections. This information is very useful in combinatorial RSV/IAV vaccine design and development.
Collapse
Affiliation(s)
- Olaitan T. Ayegbusi
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwaseyi A. Ajagbe
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Tosin O. Afowowe
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abideen T. Aransi
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Babatunde A. Olusola
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | | - Adedayo O. Faneye
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Georgina N. Odaibo
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - David O. Olaleye
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
58
|
Morgan DJ, Casulli J, Chew C, Connolly E, Lui S, Brand OJ, Rahman R, Jagger C, Hussell T. Innate Immune Cell Suppression and the Link With Secondary Lung Bacterial Pneumonia. Front Immunol 2018; 9:2943. [PMID: 30619303 PMCID: PMC6302086 DOI: 10.3389/fimmu.2018.02943] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Secondary infections arise as a consequence of previous or concurrent conditions and occur in the community or in the hospital setting. The events allowing secondary infections to gain a foothold have been studied for many years and include poor nutrition, anxiety, mental health issues, underlying chronic diseases, resolution of acute inflammation, primary immune deficiencies, and immune suppression by infection or medication. Children, the elderly and the ill are particularly susceptible. This review is concerned with secondary bacterial infections of the lung that occur following viral infection. Using influenza virus infection as an example, with comparisons to rhinovirus and respiratory syncytial virus infection, we will update and review defective bacterial innate immunity and also highlight areas for potential new investigation. It is currently estimated that one in 16 National Health Service (NHS) hospital patients develop an infection, the most common being pneumonia, lower respiratory tract infections, urinary tract infections and infection of surgical sites. The continued drive to understand the mechanisms of why secondary infections arise is therefore of key importance.
Collapse
Affiliation(s)
- David J Morgan
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christine Chew
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma Connolly
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Sylvia Lui
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Oliver J Brand
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Rizwana Rahman
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher Jagger
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
59
|
Abstract
Pneumonia is a type of acute lower respiratory infection that is common and severe. The outcome of lower respiratory infection is determined by the degrees to which immunity is protective and inflammation is damaging. Intercellular and interorgan signaling networks coordinate these actions to fight infection and protect the tissue. Cells residing in the lung initiate and steer these responses, with additional immunity effectors recruited from the bloodstream. Responses of extrapulmonary tissues, including the liver, bone marrow, and others, are essential to resistance and resilience. Responses in the lung and extrapulmonary organs can also be counterproductive and drive acute and chronic comorbidities after respiratory infection. This review discusses cell-specific and organ-specific roles in the integrated physiological response to acute lung infection, and the mechanisms by which intercellular and interorgan signaling contribute to host defense and healthy respiratory physiology or to acute lung injury, chronic pulmonary disease, and adverse extrapulmonary sequelae. Pneumonia should no longer be perceived as simply an acute infection of the lung. Pneumonia susceptibility reflects ongoing and poorly understood chronic conditions, and pneumonia results in diverse and often persistent deleterious consequences for multiple physiological systems.
Collapse
Affiliation(s)
- Lee J Quinton
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Allan J Walkey
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
60
|
Bouras M, Asehnoune K, Roquilly A. Contribution of Dendritic Cell Responses to Sepsis-Induced Immunosuppression and to Susceptibility to Secondary Pneumonia. Front Immunol 2018; 9:2590. [PMID: 30483258 PMCID: PMC6243084 DOI: 10.3389/fimmu.2018.02590] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/22/2018] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are bone marrow derived cells which continuously seed in peripheral tissue. During infection, DCs play an essential interface between innate and adaptive immunity. Pneumonia is a lung inflammation triggered by pathogens and is characterized by excessive release of inflammatory cytokines that activate innate and acquired immunity. Pneumonia induces a rapid and protracted state of susceptibility to secondary infection, a state so-called sepsis-induced immunosuppression. In this review, we focus on the role of DCs in the development of this state of immunosuppression. Early during inflammation, activated DCs are characterized by decreased capacity of antigen (cross)- presentation of newly encountered antigens and decreased production of immunogenic cytokines, and sepsis-induced immunosuppression is mainly explained by a depletion of immature DCs which had all become mature. At a later stage, newly formed respiratory immature DCs are locally programmed by an immunological scare left-over by inflammation to induce tolerance. Tolerogenic Blimp1+ DCs produce suppressive cytokines such as tumor growth factor-B and participate to the maintenance of a local tolerogenic environment notably characterized by accumulation of Treg cells. In mice, the restoration of the immunogenic functions of DCs restores the mucosal immune response to pathogens. In humans, the modulation of inflammation by glucocorticoid during sepsis or trauma preserves DC immunogenic functions and is associated with resistance to secondary pneumonia. Finally, we propose that the alterations of DCs during and after inflammation can be used as biomarkers of susceptibility to secondary pneumonia and are promising therapeutic targets to enhance outcomes of patients with secondary pneumonia.
Collapse
Affiliation(s)
- Marwan Bouras
- Surgical Intensive Care Unit, Hotel Dieu, University Hospital of Nantes, Nantes, France.,EA3826 Thérapeutiques Anti-Infectieuses, Institut de Recherche en Santé 2 Nantes Biotech, Medical University of Nantes, Nantes, France
| | - Karim Asehnoune
- Surgical Intensive Care Unit, Hotel Dieu, University Hospital of Nantes, Nantes, France.,EA3826 Thérapeutiques Anti-Infectieuses, Institut de Recherche en Santé 2 Nantes Biotech, Medical University of Nantes, Nantes, France
| | - Antoine Roquilly
- Surgical Intensive Care Unit, Hotel Dieu, University Hospital of Nantes, Nantes, France.,EA3826 Thérapeutiques Anti-Infectieuses, Institut de Recherche en Santé 2 Nantes Biotech, Medical University of Nantes, Nantes, France
| |
Collapse
|
61
|
Kang SS, Kim AR, Yun CH, Han SH. Staphylococcus aureus lipoproteins augment inflammatory responses in poly I:C-primed macrophages. Cytokine 2018; 111:154-161. [PMID: 30153621 DOI: 10.1016/j.cyto.2018.08.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 01/22/2023]
Abstract
Secondary bacterial infection contributes to severe inflammation following viral infection. Among foodborne pathogenic bacteria, Staphylococcus aureus is known to exacerbate severe inflammatory responses after infection with single-stranded RNA viruses such as influenza viruses. However, it has not been determined if S. aureus infection enhances inflammatory responses after infection with RNA enteric viruses, including rotavirus, which is a double-stranded RNA virus. We therefore investigated the molecular mechanisms by which a cell wall component of S. aureus enhanced inflammatory responses during enteric viral infection using poly I:C-primed macrophages, which is a well-established model for double-stranded RNA virus infection. S. aureus lipoproteins enhanced IL-6 as well as TNF-α production in poly I:C-primed macrophages. Pam2CSK4, a mimic of Gram-positive bacterial lipoproteins and S. aureus lipoproteins, also significantly enhanced IL-6 production in poly I:C-primed macrophages. While IFN-β expression was increased in poly I:C-primed macrophages treated with Pam2CSK4 or S. aureus lipoproteins, the level of IL-6 enhancement in poly I:C-primed macrophages was decreased in the presence of anti-IFN-α/β receptor antibody, suggesting that IFN-β plays an important role in enhanced IL-6 production. Phosphatidylinositol-3-kinase, Akt, ERK and NF-κB were also involved in the enhanced IL-6 production. Collectively, these results suggest that S. aureus lipoproteins induce excessive inflammatory responses in the presence of poly I:C.
Collapse
Affiliation(s)
- Seok-Seong Kang
- Department of Food Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - A Reum Kim
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
62
|
Koch RM, Diavatopoulos DA, Ferwerda G, Pickkers P, de Jonge MI, Kox M. The endotoxin-induced pulmonary inflammatory response is enhanced during the acute phase of influenza infection. Intensive Care Med Exp 2018; 6:15. [PMID: 29978355 PMCID: PMC6033844 DOI: 10.1186/s40635-018-0182-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/22/2018] [Indexed: 12/19/2022] Open
Abstract
Background Influenza infections are often complicated by secondary infections, which are associated with high morbidity and mortality, suggesting that influenza profoundly influences the immune response towards a subsequent pathogenic challenge. However, data on the immunological interplay between influenza and secondary infections are equivocal, with some studies reporting influenza-induced augmentation of the immune response, whereas others demonstrate that influenza suppresses the immune response towards a subsequent challenge. These contrasting results may be due to the use of various types of live bacteria as secondary challenges, which impedes clear interpretation of causal relations, and to differences in timing of the secondary challenge relative to influenza infection. Herein, we investigated whether influenza infection results in an enhanced or suppressed innate immune response upon a secondary challenge with bacterial lipopolysaccharide (LPS) in either the acute or the recovery phase of infection. Methods Male C57BL/6J mice were intranasally inoculated with 5 × 103 PFU influenza virus (pH1N1, strain A/Netherlands/602/2009) or mock treated. After 4 (acute phase) or 10 (recovery phase) days, 5 mg/kg LPS or saline was administered intravenously, and mice were sacrificed 90 min later. Cytokine levels in plasma and lung tissue, and lung myeloperoxidase (MPO) content were determined. Results LPS administration 4 days after influenza infection resulted in a synergistic increase in TNF-α, IL-1β, and IL-6 concentrations in lung tissue, but not in plasma. This effect was also observed 10 days after influenza infection, albeit to a lesser extent. LPS-induced plasma levels of the anti-inflammatory cytokine IL-10 were enhanced 4 days after influenza infection, whereas a trend towards increased pulmonary IL-10 concentrations was found. LPS-induced increases in pulmonary MPO content tended to be enhanced as well, but only at 4 days post-infection. Conclusions An LPS challenge in the acute phase of influenza infection results in an enhanced pulmonary pro-inflammatory innate immune response. These data increase our insight on influenza-bacterial interplay. Combing data of the present study with previous findings, it appears that this enhanced response is not beneficial in terms of protection against secondary infections, but rather damaging by increasing immunopathology.
Collapse
Affiliation(s)
- R M Koch
- Department of Intensive Care Medicine, Radboud university medical centre, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands
| | - D A Diavatopoulos
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical centre, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands
| | - G Ferwerda
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical centre, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands
| | - P Pickkers
- Department of Intensive Care Medicine, Radboud university medical centre, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands
| | - M I de Jonge
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical centre, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands
| | - M Kox
- Department of Intensive Care Medicine, Radboud university medical centre, Nijmegen, The Netherlands. .,Radboud Center for Infectious Diseases (RCI), Radboud university medical centre, Nijmegen, The Netherlands.
| |
Collapse
|
63
|
Borges LGDA, Giongo A, Pereira LDM, Trindade FJ, Gregianini TS, Campos FS, Ghedin E, da Veiga ABG. Comparison of the nasopharynx microbiome between influenza and non-influenza cases of severe acute respiratory infections: A pilot study. Health Sci Rep 2018; 1:e47. [PMID: 30623080 PMCID: PMC6266421 DOI: 10.1002/hsr2.47] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/21/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022] Open
Abstract
AIMS Influenza A virus (IAV) can cause severe acute respiratory infection (SARI), and disease outcome may be associated with changes in the microbiome of the nasopharynx. This is a pilot study to characterize the microbiome of the nasopharynx in patients hospitalized with SARI, infected and not infected by IAV. METHODS AND RESULTS Using target sequencing of the 16S rRNA gene, we assessed the bacterial community of nasopharyngeal aspirate samples and compared the microbiome of patients infected with IAV with the microbiome of patients who were negative for IAV. We observed differences in the relative abundance of Proteobacteria and Firmicutes between SARI patients, with Streptococcus being enriched and Pseudomonas underrepresented in IAV patients compared with patients who were not infected with IAV. CONCLUSION Pseudomonas taxon seems to be in high frequency on the nasopharynx of SARI patients with non-IAV infection and might present a negative association with Streptococcus taxon. Microbial profile appears to be different between SARI patients infected or not infected with IAV.
Collapse
Affiliation(s)
- Luiz Gustavo dos Anjos Borges
- Laboratório de Biologia Molecular, Programa de Pós‐Graduação em PatologiaUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreRSBrazil
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Adriana Giongo
- Instituto do Petróleo e dos Recursos Naturais (IPR)Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Leandro de Mattos Pereira
- Faculdade de BiociênciasPontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Fernanda J. Trindade
- Faculdade de BiociênciasPontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Tatiana Schäffer Gregianini
- Laboratório Central de Saúde Pública da Secretaria de Saúde do Estado do Rio Grande do Sul (LACEN/SES‐RS)Porto AlegreRSBrazil
| | - Fabrício Souza Campos
- College of Veterinary Medicine and AgronomyUniversity of Brasília, Darcy Ribeiro University Campus, ICCAsa Norte, CEP 70.910-970 BrasíliaDFBrazil
| | - Elodie Ghedin
- Center for Genomics and Systems Biology, Department of BiologyNew York UniversityNew YorkNYUSA
- Department of Epidemiology, College of Global Public HealthNew York UniversityNew YorkNYUSA
| | - Ana Beatriz Gorini da Veiga
- Laboratório de Biologia Molecular, Programa de Pós‐Graduação em PatologiaUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreRSBrazil
| |
Collapse
|
64
|
Rasid O, Cavaillon JM. Compartment diversity in innate immune reprogramming. Microbes Infect 2018; 20:156-165. [DOI: 10.1016/j.micinf.2017.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
|
65
|
Abstract
Immunosuppression renders the host increased susceptible for secondary infections. It is becoming increasingly clear that not only bacterial sepsis, but also respiratory viruses with both severe and mild disease courses such as influenza, respiratory syncytial virus, and the human rhinovirus may induce immunosuppression. In this review, the current knowledge on (mechanisms of) bacterial- and virus-induced immunosuppression and the accompanying susceptibility toward various secondary infections is described. In addition, the frequently encountered secondary pathogens and their preferred localizations are presented. Finally, future perspectives in the context of the development of diagnostic markers and possibilities for personalized therapy to improve the diagnosis and treatment of immunocompromised patients are discussed.
Collapse
|
66
|
Bansal S, Yajjala VK, Bauer C, Sun K. IL-1 Signaling Prevents Alveolar Macrophage Depletion during Influenza and Streptococcus pneumoniae Coinfection. THE JOURNAL OF IMMUNOLOGY 2018; 200:1425-1433. [PMID: 29311363 DOI: 10.4049/jimmunol.1700210] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 12/11/2017] [Indexed: 01/17/2023]
Abstract
Influenza and bacterial coinfection is a significant cause of hospitalization and death in humans during influenza epidemics and pandemics. However, the fundamental protective and pathogenic mechanisms involved in this complex virus-host-bacterium interaction remain incompletely understood. In this study, we have developed mild to lethal influenza and Streptococcus pneumoniae coinfection models for comparative analyses of disease pathogenesis. Specifically, wild-type and IL-1R type 1-deficient (Il1r1-/- ) mice were infected with influenza virus and then superchallenged with noninvasive S. pneumoniae serotype 14 (Spn14) or S. pneumoniae serotype 19A (Spn19A). The coinfections were followed by comparative analyses of inflammatory responses and animal protection. We found that resident alveolar macrophages are efficient in the clearance of both pneumococcal serotypes in the absence of influenza infection; in contrast, they are essential for airway control of Spn14 infection but not Spn19A infection. In agreement, TNF-α and neutrophils play a compensatory protective role in secondary bacterial infection associated with Spn19A; however, the essential requirement for alveolar macrophage-mediated clearance significantly enhances the virulence of Spn14 during postinfluenza pneumococcal infection. Furthermore, we show that, although IL-1 signaling is not required for host defense against pneumococcal infection alone, it is essential for sustaining antibacterial immunity during postinfluenza pneumococcal infection, as evidenced by significantly aggravated bacterial burden and animal mortality in Il1r1-/- mice. Mechanistically, we show that through preventing alveolar macrophage depletion, inflammatory cytokine IL-1 signaling is critically involved in host resistance to influenza and pneumococcal coinfection.
Collapse
Affiliation(s)
- Shruti Bansal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Vijaya Kumar Yajjala
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Christopher Bauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| | - Keer Sun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900
| |
Collapse
|
67
|
Novel protective mechanism for interleukin-33 at the mucosal barrier during influenza-associated bacterial superinfection. Mucosal Immunol 2018; 11:199-208. [PMID: 28401938 PMCID: PMC5638662 DOI: 10.1038/mi.2017.32] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/01/2017] [Indexed: 02/04/2023]
Abstract
Influenza A is a highly contagious respiratory virus that causes seasonal epidemics and occasional worldwide pandemics. The primary cause of influenza-related mortality is bacterial superinfection. There are numerous mechanisms by which preceding influenza infection attenuates host defense, allowing for increased susceptibility to bacterial pneumonia. Herein, we demonstrate that influenza inhibits Staphylococcus aureus-induced production of interleukin-33 (IL-33). Restoration of IL-33 during influenza A and methicillin-resistant S. aureus superinfection enhanced bacterial clearance and improved mortality. Innate lymphoid Type 2 cells and alternatively activated macrophages are not required for IL-33-mediated protection during superinfection. We show that IL-33 treatment resulted in neutrophil recruitment to the lung, associated with improved bacterial clearance. These findings identify a novel role for IL-33 in antibacterial host defense at the mucosal barrier.
Collapse
|
68
|
Siemens N, Oehmcke-Hecht S, Mettenleiter TC, Kreikemeyer B, Valentin-Weigand P, Hammerschmidt S. Port d'Entrée for Respiratory Infections - Does the Influenza A Virus Pave the Way for Bacteria? Front Microbiol 2017; 8:2602. [PMID: 29312268 PMCID: PMC5742597 DOI: 10.3389/fmicb.2017.02602] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022] Open
Abstract
Bacterial and viral co-infections of the respiratory tract are life-threatening and present a global burden to the global community. Staphylococcus aureus, Streptococcus pneumoniae, and Streptococcus pyogenes are frequent colonizers of the upper respiratory tract. Imbalances through acquisition of seasonal viruses, e.g., Influenza A virus, can lead to bacterial dissemination to the lower respiratory tract, which in turn can result in severe pneumonia. In this review, we summarize the current knowledge about bacterial and viral co-infections of the respiratory tract and focus on potential experimental models suitable for mimicking this disease. Transmission of IAV and pneumonia is mainly modeled by mouse infection. Few studies utilizing ferrets, rats, guinea pigs, rabbits, and non-human primates are also available. The knowledge gained from these studies led to important discoveries and advances in understanding these infectious diseases. Nevertheless, mouse and other infection models have limitations, especially in translation of the discoveries to humans. Here, we suggest the use of human engineered lung tissue, human ex vivo lung tissue, and porcine models to study respiratory co-infections, which might contribute to a greater translation of the results to humans and improve both, animal and human health.
Collapse
Affiliation(s)
- Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Peter Valentin-Weigand
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
69
|
Koch RM, Kox M, Thijs EJM, Rahamat-Langendoen JC, van de Veerdonk FL, Gerretsen J, Schloesser J, Diavatopoulos D, Rimmelzwaan GF, Netea MG, van der Hoeven JG, de Jonge MI, Pickkers P. Development of Endotoxin Tolerance Does Not Influence the Response to a Challenge with the Mucosal Live-Attenuated Influenza Vaccine in Humans In Vivo. Front Immunol 2017; 8:1600. [PMID: 29312282 PMCID: PMC5732479 DOI: 10.3389/fimmu.2017.01600] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/06/2017] [Indexed: 01/11/2023] Open
Abstract
Introduction The effects of bacterial infections on the response to subsequent viral infections are largely unknown. This is important to elucidate to increase insight into the pathophysiology of bacterial and viral co-infections, and to assess whether bacterial infections may influence the course of viral infections. Methods Healthy male subjects received either bacterial endotoxin [Escherichia coli-derived lipopolysaccharide (LPS), 2 ng/kg, n = 15] or placebo (n = 15) intravenously, followed by intranasal Fluenz (live-attenuated influenza vaccine) 1 week later. Results LPS administration resulted in increased plasma cytokine levels and development of endotoxin tolerance in vivo and ex vivo, illustrated by attenuated cytokine production upon rechallenge with LPS. Following Fluenz administration, infectivity for the Fluenz A/B strains was similar between the LPS-Fluenz and placebo-Fluenz groups (13/15 subjects in both groups). Also, the Fluenz-induced increase in temperature and IL-6, G-CSF and IP-10 concentrations in nasal wash were similar between both groups. Conclusion While endotoxemia profoundly attenuates the immune response upon a second LPS challenge, it does not influence the Fluenz-induced immune response. These results suggest immune suppression after bacterial infection does not alter the response to a subsequent viral infection.
Collapse
Affiliation(s)
- Rebecca M Koch
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands
| | - Eleonora J M Thijs
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janette C Rahamat-Langendoen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank L van de Veerdonk
- Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands.,Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands
| | | | - Dimitri Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Mihai G Netea
- Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands.,Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands
| | - Marien I de Jonge
- Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands.,Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Nijmegen, Netherlands
| |
Collapse
|
70
|
The nasopharyngeal microbiome. Emerg Top Life Sci 2017; 1:297-312. [PMID: 33525776 DOI: 10.1042/etls20170041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023]
Abstract
Human microbiomes have received increasing attention over the last 10 years, leading to a pervasiveness of hypotheses relating dysbiosis to health and disease. The respiratory tract has received much less attention in this respect than that of, for example, the human gut. Nevertheless, progress has been made in elucidating the immunological, ecological and environmental drivers that govern these microbial consortia and the potential consequences of aberrant microbiomes. In this review, we consider the microbiome of the nasopharynx, a specific niche of the upper respiratory tract. The nasopharynx is an important site, anatomically with respect to its gateway position between upper and lower airways, and for pathogenic bacterial colonisation. The dynamics of the latter are important for long-term respiratory morbidity, acute infections of both invasive and non-invasive disease and associations with chronic airway disease exacerbations. Here, we review the development of the nasopharyngeal (NP) microbiome over the life course, examining it from the early establishment of resilient profiles in neonates through to perturbations associated with pneumonia risk in the elderly. We focus specifically on the commensal, opportunistically pathogenic members of the NP microbiome that includes Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae and Moraxella catarrhalis. In addition, we consider the role of relatively harmless genera such as Dolosigranulum and Corynebacterium. Understanding that the NP microbiome plays such a key, beneficial role in maintaining equilibrium of commensal species, prevention of pathogen outgrowth and host immunity enables future research to be directed appropriately.
Collapse
|
71
|
Smith AM, Huber VC. The Unexpected Impact of Vaccines on Secondary Bacterial Infections Following Influenza. Viral Immunol 2017; 31:159-173. [PMID: 29148920 DOI: 10.1089/vim.2017.0138] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Influenza virus infections remain a significant health burden worldwide, despite available vaccines. Factors that contribute to this include a lack of broad coverage by current vaccines and continual emergence of novel virus strains. Further complicating matters, when influenza viruses infect a host, severe infections can develop when bacterial pathogens invade. Secondary bacterial infections (SBIs) contribute to a significant proportion of influenza-related mortality, with Streptococcus pneumoniae, Staphylococcus aureus, Streptococcus pyogenes, and Haemophilus influenzae as major coinfecting pathogens. Vaccines against bacterial pathogens can reduce coinfection incidence and severity, but few vaccines are available and those that are, may have decreased efficacy in influenza virus-infected hosts. While some studies indicate a benefit of vaccine-induced immunity in providing protection against SBIs, a comprehensive understanding is lacking. In this review, we discuss the current knowledge of viral and bacterial vaccine availability, the generation of protective immunity from these vaccines, and the effectiveness in limiting influenza-associated bacterial infections.
Collapse
Affiliation(s)
- Amber M Smith
- 1 Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Victor C Huber
- 2 Division of Basic Biomedical Sciences, University of South Dakota , Vermillion, South Dakota
| |
Collapse
|
72
|
Coch C, Stümpel JP, Lilien-Waldau V, Wohlleber D, Kümmerer BM, Bekeredjian-Ding I, Kochs G, Garbi N, Herberhold S, Schuberth-Wagner C, Ludwig J, Barchet W, Schlee M, Hoerauf A, Bootz F, Staeheli P, Hartmann G, Hartmann E. RIG-I Activation Protects and Rescues from Lethal Influenza Virus Infection and Bacterial Superinfection. Mol Ther 2017; 25:2093-2103. [PMID: 28760668 DOI: 10.1016/j.ymthe.2017.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/02/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022] Open
Abstract
Influenza A virus infection causes substantial morbidity and mortality in seasonal epidemic outbreaks, and more efficient treatments are urgently needed. Innate immune sensing of viral nucleic acids stimulates antiviral immunity, including cell-autonomous antiviral defense mechanisms that restrict viral replication. RNA oligonucleotide ligands that potently activate the cytoplasmic helicase retinoic-acid-inducible gene I (RIG-I) are promising candidates for the development of new antiviral therapies. Here, we demonstrate in an Mx1-expressing mouse model of influenza A virus infection that a single intravenous injection of low-dose RIG-I ligand 5'-triphosphate RNA (3pRNA) completely protected mice from a lethal challenge with influenza A virus for at least 7 days. Furthermore, systemic administration of 3pRNA rescued mice with pre-established fulminant influenza infection and prevented the fatal effects of a streptococcal superinfection. Type I interferon, but not interferon-λ, was required for the therapeutic effect. Our results suggest that the use of RIG-I activating oligonucleotide ligands has the clinical potential to confine influenza epidemics when a strain-specific vaccine is not yet available and to reduce lethality of influenza in severely infected patients.
Collapse
Affiliation(s)
- Christoph Coch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany.
| | - Jan Phillip Stümpel
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Vanessa Lilien-Waldau
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology and Experimental Oncology, TU Munich, 81675 Munich, Germany
| | - Beate M Kümmerer
- Institute of Virology, University Hospital Bonn, 53105 Bonn, Germany
| | - Isabelle Bekeredjian-Ding
- Division of Microbiology, Paul-Ehrlich Institute, 63225 Langen, Germany; Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Georg Kochs
- Institute of Virology, Medical Center Freiburg, 79104 Freiburg, Germany
| | - Natalio Garbi
- Institute of Experimental Immunology, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephan Herberhold
- Department of Otolaryngology, University Hospital Bonn, 53127 Bonn, Germany
| | - Christine Schuberth-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Janos Ludwig
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Winfried Barchet
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Friedrich Bootz
- Department of Otolaryngology, University Hospital Bonn, 53127 Bonn, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center Freiburg, 79104 Freiburg, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Evelyn Hartmann
- Department of Otolaryngology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
73
|
Lemire P, Galbas T, Thibodeau J, Segura M. Natural Killer Cell Functions during the Innate Immune Response to Pathogenic Streptococci. Front Microbiol 2017; 8:1196. [PMID: 28706510 PMCID: PMC5489694 DOI: 10.3389/fmicb.2017.01196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023] Open
Abstract
Dendritic cells (DCs) and NK cells play a crucial role in the first phase of host defense against infections. Group B Streptococcus (GBS) and Streptococcus suis are encapsulated streptococci causing severe systemic inflammation, leading to septicemia and meningitis. Yet, the involvement of NK cells in the innate immune response to encapsulated bacterial infection is poorly characterized. Here, it was observed that these two streptococcal species rapidly induce the release of IFN-γ and that NK cells are the major cell type responsible for this production during the acute phase of the infection. Albeit S. suis capacity to activate NK cells was lower than that of GBS, these cells partially contribute to S. suis systemic infection; mainly through amplification of the inflammatory loop. In contrast, such a role was not observed during GBS systemic infection. IFN-γ release by NK cells required the presence of DCs, which in turn had a synergistic effect on DC cytokine production. These responses were mainly mediated by direct DC-NK cell contact and partially dependent on soluble factors. Though IL-12 and LFA-1 were shown to be critical in S. suis-mediated activation of the DC-NK cell crosstalk, different or redundant molecular pathways modulate DC-NK interactions during GBS infection. The bacterial capsular polysaccharides also differently modulated NK cell activation. Together, these results demonstrated a role of NK cells in the innate immune response against encapsulated streptococcal infections; yet the molecular pathways governing NK activation seem to differ upon the pathogen and should not be generalized when studying bacterial infections.
Collapse
Affiliation(s)
- Paul Lemire
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| | - Tristan Galbas
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Jacques Thibodeau
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Mariela Segura
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| |
Collapse
|
74
|
Ordija CM, Chiou TTY, Yang Z, Deloid GM, de Oliveira Valdo M, Wang Z, Bedugnis A, Noah TL, Jones S, Koziel H, Kobzik L. Free actin impairs macrophage bacterial defenses via scavenger receptor MARCO interaction with reversal by plasma gelsolin. Am J Physiol Lung Cell Mol Physiol 2017; 312:L1018-L1028. [PMID: 28385809 PMCID: PMC5495953 DOI: 10.1152/ajplung.00067.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 12/20/2022] Open
Abstract
Lung injury can release intracellular actin into the alveolar milieu and is also associated with increased susceptibility to secondary infections. We investigated the effect of free (extracellular) actin on lung macrophage host defense functions. Western blot analysis demonstrated free actin release into the lung lavage fluids of mouse models of ozone injury, influenza infection, and secondary pneumococcal pneumonia and in samples from patients following burn and inhalation injury. Using levels comparable with those observed in lung injury, we found that free actin markedly inhibited murine lung macrophage binding and uptake in vitro of S. pneumoniae, S. aureus, and E. coli, (e.g., S. pneumoniae, mean %inhibition, actin vs. vehicle: 85 ± 0.3 (SD); n = 22, P < .001). Similar effects were observed on the ability of primary human macrophages to bind and ingest fluorescent Saureus (~75% inhibition). Plasma gelsolin (pGSN), a protein that functions to bind and cleave actin, restored bacterial binding and uptake by both murine and human macrophages. Scavenger receptor inhibitors reduced binding of fluorescent actin by murine macrophages [fluorescence index (×10-3) after incubation with vehicle, actin, or actin + polyinosinic acid, respectively: 0.8 ± 0.7, 101.7 ± 50.7, or 52.7 ± 16.9; n = 5-6, P < 0.05]. In addition, actin binding was reduced in a MARCO/SR-AI/II-deficient cell line and by normal AMs obtained from MARCO-/- mice. After release from injured cells during lung injury, free actin likely contributes to impaired host defense by blocking scavenger receptor binding of bacteria. This mechanism for increased risk of secondary infections after lung injury or inflammation may represent another target for therapeutic intervention with pGSN.
Collapse
Affiliation(s)
- Christine M Ordija
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Terry Ting-Yu Chiou
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts.,Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang-Gung University College of Medicine, Kaohsiung, Taiwan
| | - Zhiping Yang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Glen M Deloid
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Melina de Oliveira Valdo
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Zhi Wang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Alice Bedugnis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Terry L Noah
- Department of Pediatrics, Pulmonology Division, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Samuel Jones
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Henry Koziel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Lester Kobzik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts;
| |
Collapse
|
75
|
Qi X, Liu C, Li R, Zhang H, Xu X, Wang J. Modulation of the innate immune-related genes expression in H9N2 avian influenza virus-infected chicken macrophage-like cells (HD11) in response to Escherichia coli LPS stimulation. Res Vet Sci 2017; 111:36-42. [DOI: 10.1016/j.rvsc.2016.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 11/07/2016] [Accepted: 11/16/2016] [Indexed: 01/08/2023]
|
76
|
Influenza A virus-induced release of interleukin-10 inhibits the anti-microbial activities of invariant natural killer T cells during invasive pneumococcal superinfection. Mucosal Immunol 2017; 10:460-469. [PMID: 27220813 DOI: 10.1038/mi.2016.49] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/07/2016] [Indexed: 02/04/2023]
Abstract
During influenza A virus (IAV) infection, changes in the lung's physical and immunological defenses predispose the host to bacterial superinfections. Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that have beneficial or harmful functions during infection. We investigated the iNKT cells' role in a model of invasive pneumococcal superinfection. The use of Jα18-/- mice indicated that iNKT cells limited susceptibility to influenza-pneumococcal infection and reduced the lethal synergism. This role did not depend on immune-based anti-bacterial mechanisms. At the time of bacterial exposure, iNKT cells from IAV-experienced mice failed to produce antipneumococcal interferon-γ and adoptive transfer of fresh iNKT cells before Streptococcus pneumoniae challenge did not restore anti-bacterial host defenses. Impaired iNKT cell activation in superinfected animals was related to the IAV-induced immunosuppressive cytokine interleukin-10 (IL-10), rather than to an intrinsic functional defect. IL-10 dampened the activation of iNKT cells in response to pneumococci by inhibiting the production of IL-12 by pulmonary monocyte-derived dendritic cells. Neutralization of IL-10 restored iNKT cell activation and tends to increase resistance to secondary bacterial infection. Overall, iNKT cells have a beneficial role (upstream of bacterial colonization) in controlling influenza-pneumococcal superinfection, although they represent novel targets of immunosuppression at the time of bacterial challenge.
Collapse
|
77
|
A Critical, Nonlinear Threshold Dictates Bacterial Invasion and Initial Kinetics During Influenza. Sci Rep 2016; 6:38703. [PMID: 27974820 PMCID: PMC5156930 DOI: 10.1038/srep38703] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022] Open
Abstract
Secondary bacterial infections increase morbidity and mortality of influenza A virus (IAV) infections. Bacteria are able to invade due to virus-induced depletion of alveolar macrophages (AMs), but this is not the only contributing factor. By analyzing a kinetic model, we uncovered a nonlinear initial dose threshold that is dependent on the amount of virus-induced AM depletion. The threshold separates the growth and clearance phenotypes such that bacteria decline for dose-AM depletion combinations below the threshold, stay constant near the threshold, and increase above the threshold. In addition, the distance from the threshold correlates to the growth rate. Because AM depletion changes throughout an IAV infection, the dose requirement for bacterial invasion also changes accordingly. Using the threshold, we found that the dose requirement drops dramatically during the first 7d of IAV infection. We then validated these analytical predictions by infecting mice with doses below or above the predicted threshold over the course of IAV infection. These results identify the nonlinear way in which two independent factors work together to support successful post-influenza bacterial invasion. They provide insight into coinfection timing, the heterogeneity in outcome, the probability of acquiring a coinfection, and the use of new therapeutic strategies to combat viral-bacterial coinfections.
Collapse
|
78
|
Sviriaeva EN, Korneev KV, Drutskaya MS, Kuprash DV. Mechanisms of Changes in Immune Response during Bacterial Coinfections of the Respiratory Tract. BIOCHEMISTRY (MOSCOW) 2016; 81:1340-1349. [PMID: 27914459 DOI: 10.1134/s0006297916110110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Acute diseases of the respiratory tract are often caused by viral pathogens and accompanying secondary bacterial infections. It is known that the development of such bacterial complications is caused mainly by a decreased infiltration with immune system cells and by suppressed inflammation in the lungs. There are significant advances in understanding the mechanisms of secondary infections, although many details remain unclear. This review summarizes current knowledge of the molecular and cellular changes in the host organism that can influence the course of bacterial coinfections in the respiratory tract.
Collapse
Affiliation(s)
- E N Sviriaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | | | | | | |
Collapse
|
79
|
Influenza virus infection causes neutrophil dysfunction through reduced G-CSF production and an increased risk of secondary bacteria infection in the lung. Virology 2016; 499:23-29. [DOI: 10.1016/j.virol.2016.08.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 11/24/2022]
|
80
|
Sun K, He SB, Qu JG, Dang SC, Chen JX, Gong AH, Xie R, Zhang JX. IRF5 regulates lung macrophages M2 polarization during severe acute pancreatitis in vitro. World J Gastroenterol 2016; 22:9368-9377. [PMID: 27895424 PMCID: PMC5107700 DOI: 10.3748/wjg.v22.i42.9368] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/13/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the role of interferon regulatory factor 5 (IRF5) in reversing polarization of lung macrophages during severe acute pancreatitis (SAP) in vitro.
METHODS A mouse SAP model was established by intraperitoneal (ip) injections of 20 μg/kg body weight caerulein. Pathological changes in the lung were observed by hematoxylin and eosin staining. Lung macrophages were isolated from bronchoalveolar lavage fluid. The quantity and purity of lung macrophages were detected by fluorescence-activated cell sorting and evaluated by real-time polymerase chain reaction (RT-PCR). They were treated with IL-4/IRF5 specific siRNA (IRF5 siRNA) to reverse their polarization and were evaluated by detecting markers expression of M1/M2 using RT-PCR.
RESULTS SAP associated acute lung injury (ALI) was induced successfully by ip injections of caerulein, which was confirmed by histopathology. Lung macrophages expressed high levels of IRF5 as M1 phenotype during the early acute pancreatitis stages. Reduction of IRF5 expression by IRF5 siRNA reversed the action of macrophages from M1 to M2 phenotype in vitro. The expressions of M1 markers, including IRF5 (S + IRF5 siRNA vs S + PBS, 0.013 ± 0.01 vs 0.054 ± 0.047, P < 0.01), TNF-α (S + IRF5 siRNA vs S + PBS, 0.0003 ± 0.0002 vs 0.019 ± 0.018, P < 0.001), iNOS (S + IRF5 siRNA vs S + PBS, 0.0003 ± 0.0002 vs 0.026 ± 0.018, P < 0.001) and IL-12 (S + IRF5 siRNA vs S + PBS, 0.000005 ± 0.00004 vs 0.024 ± 0.016, P < 0.001), were decreased. In contrast, the expressions of M2 markers, including IL-10 (S + IRF5 siRNA vs S + PBS, 0.060 ± 0.055 vs 0.0230 ± 0.018, P < 0.01) and Arg-1 (S + IRF5 siRNA vs S + PBS, 0.910 ± 0.788 vs 0.0036 ± 0.0025, P < 0.001), were increased. IRF5 siRNA could reverse the lung macrophage polarization more effectively than IL-4.
CONCLUSION Treatment with IRF5 siRNA can reverse the pancreatitis-induced activation of lung macrophages from M1 phenotype to M2 phenotype in SAP associated with ALI.
Collapse
|
81
|
Bellinghausen C, Rohde GGU, Savelkoul PHM, Wouters EFM, Stassen FRM. Viral-bacterial interactions in the respiratory tract. J Gen Virol 2016; 97:3089-3102. [PMID: 27902340 DOI: 10.1099/jgv.0.000627] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the respiratory tract, viruses and bacteria can interact on multiple levels. It is well known that respiratory viruses, particularly influenza viruses, increase the susceptibility to secondary bacterial infections. Numerous mechanisms, including compromised physical and immunological barriers, and changes in the microenvironment have hereby been shown to contribute to the development of secondary bacterial infections. In contrast, our understanding of how bacteria shape a response to subsequent viral infection is still limited. There is emerging evidence that persistent infection (or colonization) of the lower respiratory tract (LRT) with potential pathogenic bacteria, as observed in diseases like chronic obstructive pulmonary disease or cystic fibrosis, modulates subsequent viral infections by increasing viral entry receptors and modulating the inflammatory response. Moreover, recent studies suggest that even healthy lungs are not, as had long been assumed, sterile. The composition of the lung microbiome may thus modulate responses to viral infections. Here we summarize the current knowledge on the co-pathogenesis between viruses and bacteria in LRT infections.
Collapse
Affiliation(s)
- Carla Bellinghausen
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gernot G U Rohde
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paul H M Savelkoul
- Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frank R M Stassen
- Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
82
|
Gál O, Pletz MW, Ianoși ES. Severe pneumococcal pneumonia following Measles- Mumps- Rubella vaccination / Pneumonie pneumococică severă dezvoltată ca urmare a vaccinării Rujeolă-Oreion-Rubeolă. REV ROMANA MED LAB 2016. [DOI: 10.1515/rrlm-2016-0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Hereby, we report a case of severe multi-lobular pneumococcal pneumonia with gangrene requiring pneumonectomy and accompanying septic shock that developed 1 day after a live-attenuated measles-mumps-rubella vaccination in a 58-year old kindergarten teacher.
Collapse
Affiliation(s)
- Orsolya Gál
- University of Medicine and Pharmacy of Tîrgu Mureș, Romania
| | - Mathias W. Pletz
- Center for Infectious Diseases and Infection Control Jena University Hospital, Jena, Germany
| | - Edith Simona Ianoși
- University of Medicine and Pharmacy of Tîrgu Mureș, Str. Gh. Marinescu, Nr. 5 Tîrgu Mureș, Romania
| |
Collapse
|
83
|
Lan F, Zhang N, Gevaert E, Zhang L, Bachert C. Viruses and bacteria in Th2-biased allergic airway disease. Allergy 2016; 71:1381-92. [PMID: 27188632 DOI: 10.1111/all.12934] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 01/24/2023]
Abstract
Allergic airway diseases are typically characterized by a type 2-biased inflammation. Multiple distinct viruses and bacteria have been detected in the airways. Recently, it has been confirmed that the microbiome of allergic individuals differs from that of healthy subjects, showing a close relationship with the type 2 response in allergic airway disease. In this review, we summarize the recent findings on the prevalence of viruses and bacteria in type 2-biased airway diseases and on the mechanisms employed by viruses and bacteria in propagating type 2 responses. The understanding of the microbial composition and postinfectious immune programming is critical for the reconstruction of the normal microflora and immune status in allergic airway diseases.
Collapse
Affiliation(s)
- F. Lan
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - N. Zhang
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - E. Gevaert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - L. Zhang
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - C. Bachert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Division of ENT Diseases; Clintec; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
84
|
Smith AM. Quantifying the therapeutic requirements and potential for combination therapy to prevent bacterial coinfection during influenza. J Pharmacokinet Pharmacodyn 2016; 44:81-93. [PMID: 27679506 PMCID: PMC5376398 DOI: 10.1007/s10928-016-9494-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/17/2016] [Indexed: 01/17/2023]
Abstract
Secondary bacterial infections (SBIs) exacerbate influenza-associated disease and mortality. Antimicrobial agents can reduce the severity of SBIs, but many have limited efficacy or cause adverse effects. Thus, new treatment strategies are needed. Kinetic models describing the infection process can help determine optimal therapeutic targets, the time scale on which a drug will be most effective, and how infection dynamics will change under therapy. To understand how different therapies perturb the dynamics of influenza infection and bacterial coinfection and to quantify the benefit of increasing a drug's efficacy or targeting a different infection process, I analyzed data from mice treated with an antiviral, an antibiotic, or an immune modulatory agent with kinetic models. The results suggest that antivirals targeting the viral life cycle are most efficacious in the first 2 days of infection, potentially because of an improved immune response, and that increasing the clearance of infected cells is important for treatment later in the infection. For a coinfection, immunotherapy could control low bacterial loads with as little as 20 % efficacy, but more effective drugs would be necessary for high bacterial loads. Antibiotics targeting bacterial replication and administered 10 h after infection would require 100 % efficacy, which could be reduced to 40 % with prophylaxis. Combining immunotherapy with antibiotics could substantially increase treatment success. Taken together, the results suggest when and why some therapies fail, determine the efficacy needed for successful treatment, identify potential immune effects, and show how the regulation of underlying mechanisms can be used to design new therapeutic strategies.
Collapse
Affiliation(s)
- Amber M Smith
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
85
|
Sun K, Yajjala VK, Bauer C, Talmon GA, Fischer KJ, Kielian T, Metzger DW. Nox2-derived oxidative stress results in inefficacy of antibiotics against post-influenza S. aureus pneumonia. J Exp Med 2016; 213:1851-64. [PMID: 27526712 PMCID: PMC4995072 DOI: 10.1084/jem.20150514] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/30/2016] [Indexed: 01/21/2023] Open
Abstract
Phagocyte oxidative burst is the primary source of lethal lung injury during influenza and MRSA coinfection. Clinical post-influenza Staphylococcus aureus pneumonia is characterized by extensive lung inflammation associated with severe morbidity and mortality even after appropriate antibiotic treatment. In this study, we show that antibiotics rescue nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2)–deficient mice but fail to fully protect WT animals from influenza and S. aureus coinfection. Further experiments indicate that the inefficacy of antibiotics against coinfection is attributable to oxidative stress–associated inflammatory lung injury. However, Nox2-induced lung damage during coinfection was not associated with aggravated inflammatory cytokine response or cell infiltration but rather caused by reduced survival of myeloid cells. Specifically, oxidative stress increased necrotic death of inflammatory cells, thereby resulting in lethal damage to surrounding tissue. Collectively, our results demonstrate that influenza infection disrupts the delicate balance between Nox2-dependent antibacterial immunity and inflammation. This disruption leads to not only increased susceptibility to S. aureus infection, but also extensive lung damage. Importantly, we show that combination treatment of antibiotic and NADPH oxidase inhibitor significantly improved animal survival from coinfection. These findings suggest that treatment strategies that target both bacteria and oxidative stress will significantly benefit patients with influenza-complicated S. aureus pneumonia.
Collapse
Affiliation(s)
- Keer Sun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198 Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Vijaya Kumar Yajjala
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Christopher Bauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Karl J Fischer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Dennis W Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| |
Collapse
|
86
|
Crum-Cianflone NF. Invasive Aspergillosis Associated With Severe Influenza Infections. Open Forum Infect Dis 2016; 3:ofw171. [PMID: 27704024 PMCID: PMC5047415 DOI: 10.1093/ofid/ofw171] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/05/2016] [Indexed: 11/21/2022] Open
Abstract
Background. Invasive aspergillosis may occur in the setting of severe influenza infections due to viral-induced respiratory epithelium disruption and impaired immune effects, but data are limited. Methods. A retrospective study was conducted among severe influenza cases requiring medical intensive care unit (ICU) admission at an academic center during the 2015–2016 season. Data collected included respiratory cultures, medical conditions and immunosuppressants, laboratory and radiographic data, and outcomes. A systematic literature review of published cases in the English language of aspergillosis complicating influenza was conducted. Results. Six (75%) of 8 ICU influenza cases had Aspergillus isolated; 5 were classified as invasive disease. No ICU patient testing negative for influenza infection developed aspergillosis during the study period. Among cases with invasive aspergillosis, influenza infection was type A (H1N1) (n = 2) and influenza B (n = 3). Published and current cases yielded n = 57 (European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group criteria: 37% proven, 25% probable, and 39% possible cases). An increasing number of cases were reported since 2010. Sixty-five percent of cases lacked classic underlying conditions at admission for aspergillosis, 86% had lymphopenia, and 46% died. Conclusions. Aspergillosis may occur in the setting of severe influenza infections even among immunocompetent hosts. Risks may include influenza A (H1N1) or B infections and viral-induced lymphopenia, although further studies are needed. Prompt diagnosis and antifungal therapy are recommended given high mortality rates.
Collapse
|
87
|
Boianelli A, Sharma-Chawla N, Bruder D, Hernandez-Vargas EA. Oseltamivir PK/PD Modeling and Simulation to Evaluate Treatment Strategies against Influenza-Pneumococcus Coinfection. Front Cell Infect Microbiol 2016; 6:60. [PMID: 27379214 PMCID: PMC4906052 DOI: 10.3389/fcimb.2016.00060] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
Influenza pandemics and seasonal outbreaks have shown the potential of Influenza A virus (IAV) to enhance susceptibility to a secondary infection with the bacterial pathogen Streptococcus pneumoniae (Sp). The high morbidity and mortality rate revealed the poor efficacy of antiviral drugs and vaccines to fight IAV infections. Currently, the most effective treatment for IAV is by antiviral neuraminidase inhibitors. Among them, the most frequently stockpiled is Oseltamivir which reduces viral release and transmission. However, effectiveness of Oseltamivir is compromised by the emergence of resistant IAV strains and secondary bacterial infections. To date, little attention has been given to evaluate how Oseltamivir treatment strategies alter Influenza viral infection in presence of Sp coinfection and a resistant IAV strain emergence. In this paper we investigate the efficacy of current approved Oseltamivir treatment regimens using a computational approach. Our numerical results suggest that the curative regimen (75 mg) may yield 47% of antiviral efficacy and 9% of antibacterial efficacy. An increment in dose to 150 mg (pandemic regimen) may increase the antiviral efficacy to 49% and the antibacterial efficacy to 16%. The choice to decrease the intake frequency to once per day is not recommended due to a significant reduction in both antiviral and antibacterial efficacy. We also observe that the treatment duration of 10 days may not provide a clear improvement on the antiviral and antibacterial efficacy compared to 5 days. All together, our in silico study reveals the success and pitfalls of Oseltamivir treatment strategies within IAV-Sp coinfection and calls for testing the validity in clinical trials.
Collapse
Affiliation(s)
- Alessandro Boianelli
- Systems Medicine of Infectious Diseases, Department of Systems Immunology and Braunschweig Integrated Centre for Infection Research, Helmholtz Centre for Infection Research Braunschweig, Germany
| | - Niharika Sharma-Chawla
- Immune Regulation, Helmholtz Centre for Infection ResearchBraunschweig, Germany; Infection Immunology, Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke-UniversityMagdeburg, Germany
| | - Dunja Bruder
- Immune Regulation, Helmholtz Centre for Infection ResearchBraunschweig, Germany; Infection Immunology, Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke-UniversityMagdeburg, Germany
| | - Esteban A Hernandez-Vargas
- Systems Medicine of Infectious Diseases, Department of Systems Immunology and Braunschweig Integrated Centre for Infection Research, Helmholtz Centre for Infection Research Braunschweig, Germany
| |
Collapse
|
88
|
Chung DR, Huh K. Novel pandemic influenza A (H1N1) and community-associated methicillin-resistant Staphylococcus aureus pneumonia. Expert Rev Anti Infect Ther 2016; 13:197-207. [PMID: 25578884 DOI: 10.1586/14787210.2015.999668] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Postinfluenza bacterial pneumonia is a leading cause of influenza-associated death, and Staphylococcus aureus and Streptococcus pneumoniae have been important pathogens that have caused pneumonia since the influenza pandemic in 1919. Emergence of novel influenza A (H1N1) pdm09 and the concomitant global spread of community-associated methicillin-resistant S. aureus (CA-MRSA) have led to increasing prevalence of CA-MRSA pneumonia following influenza infection. Such an epidemiologic change poses a therapeutic challenge due to a high risk of inappropriate empiric antimicrobial therapy and poor clinical outcomes. Early diagnosis and initiation of appropriate antimicrobial therapy for post-influenza bacterial pneumonia have become even more important in the era of CA-MRSA. Therefore, novel molecular diagnostic techniques should be applied to more readily diagnose MRSA pneumonia.
Collapse
Affiliation(s)
- Doo Ryeon Chung
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-ro 81, Gangnam-gu, Seoul 135-710, Republic of Korea
| | | |
Collapse
|
89
|
de Steenhuijsen Piters WAA, Sanders EAM, Bogaert D. The role of the local microbial ecosystem in respiratory health and disease. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0294. [PMID: 26150660 DOI: 10.1098/rstb.2014.0294] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Respiratory tract infections are a major global health concern, accounting for high morbidity and mortality, especially in young children and elderly individuals. Traditionally, highly common bacterial respiratory tract infections, including otitis media and pneumonia, were thought to be caused by a limited number of pathogens including Streptococcus pneumoniae and Haemophilus influenzae. However, these pathogens are also frequently observed commensal residents of the upper respiratory tract (URT) and form-together with harmless commensal bacteria, viruses and fungi-intricate ecological networks, collectively known as the 'microbiome'. Analogous to the gut microbiome, the respiratory microbiome at equilibrium is thought to be beneficial to the host by priming the immune system and providing colonization resistance, while an imbalanced ecosystem might predispose to bacterial overgrowth and development of respiratory infections. We postulate that specific ecological perturbations of the bacterial communities in the URT can occur in response to various lifestyle or environmental effectors, leading to diminished colonization resistance, loss of containment of newly acquired or resident pathogens, preluding bacterial overgrowth, ultimately resulting in local or systemic bacterial infections. Here, we review the current body of literature regarding niche-specific upper respiratory microbiota profiles within human hosts and the changes occurring within these profiles that are associated with respiratory infections.
Collapse
Affiliation(s)
- Wouter A A de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Elisabeth A M Sanders
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
90
|
D’Anna SE, Balbi B, Cappello F, Carone M, Di Stefano A. Bacterial-viral load and the immune response in stable and exacerbated COPD: significance and therapeutic prospects. Int J Chron Obstruct Pulmon Dis 2016; 11:445-53. [PMID: 27042037 PMCID: PMC4780195 DOI: 10.2147/copd.s93398] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation and an abnormal inflammatory response of the lung. Bacteria and viruses are a major cause of COPD exacerbations and may contribute to COPD progression by perpetuating the inflammatory response in the airways. Bacterial variety diminishes with increasing COPD severity. Respiratory viruses can colonize the lower respiratory tract in stable COPD, altering the respiratory microbiome and facilitating secondary bacterial infections. In this review, we present the most updated information about the role of bacteria and viruses in stable and exacerbated COPD. In our opinion, to optimize therapeutic strategies, the dynamic events involving bacterial-viral infections and related immune response in COPD phenotypes need to be better clarified. Our paper would address these points that we consider of great importance for the clinical management of COPD.
Collapse
Affiliation(s)
- Silvestro Ennio D’Anna
- Department of Rehabilitation, Cardiorespiratory Unit, Fondazione Istituto G. Giglio di Cefalù, University of Palermo, Palermo, Italy
| | - Bruno Balbi
- Pneumology Unit and Laboratory of Cytoimmunopathology of Heart and Lung, Fondazione Salvatore Maugeri, IRCCS, Veruno (NO) and Cassano delle Murge (BA), University of Palermo, Palermo, Italy
| | - Francesco Cappello
- Human Anatomy Section, Department of Experimental Biomedicine and Clinical Neuroscience, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Mauro Carone
- Pneumology Unit and Laboratory of Cytoimmunopathology of Heart and Lung, Fondazione Salvatore Maugeri, IRCCS, Veruno (NO) and Cassano delle Murge (BA), University of Palermo, Palermo, Italy
| | - Antonino Di Stefano
- Pneumology Unit and Laboratory of Cytoimmunopathology of Heart and Lung, Fondazione Salvatore Maugeri, IRCCS, Veruno (NO) and Cassano delle Murge (BA), University of Palermo, Palermo, Italy
| |
Collapse
|
91
|
Parker D, Ahn D, Cohen T, Prince A. Innate Immune Signaling Activated by MDR Bacteria in the Airway. Physiol Rev 2016; 96:19-53. [PMID: 26582515 DOI: 10.1152/physrev.00009.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Health care-associated bacterial pneumonias due to multiple-drug resistant (MDR) pathogens are an important public health problem and are major causes of morbidity and mortality worldwide. In addition to antimicrobial resistance, these organisms have adapted to the milieu of the human airway and have acquired resistance to the innate immune clearance mechanisms that normally prevent pneumonia. Given the limited efficacy of antibiotics, bacterial clearance from the airway requires an effective immune response. Understanding how specific airway pathogens initiate and regulate innate immune signaling, and whether this response is excessive, leading to host-induced pathology may guide future immunomodulatory therapy. We will focus on three of the most important causes of health care-associated pneumonia, Staphylococcus aureus, Pseudomonas aeruginosa, and Klebsiella pneumoniae, and review the mechanisms through which an inappropriate or damaging innate immune response is stimulated, as well as describe how airway pathogens cause persistent infection by evading immune activation.
Collapse
Affiliation(s)
- Dane Parker
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Danielle Ahn
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Taylor Cohen
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Alice Prince
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| |
Collapse
|
92
|
Lee KH, Gordon A, Foxman B. The role of respiratory viruses in the etiology of bacterial pneumonia: An ecological perspective. Evol Med Public Health 2016; 2016:95-109. [PMID: 26884414 PMCID: PMC4801059 DOI: 10.1093/emph/eow007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/29/2016] [Indexed: 12/18/2022] Open
Abstract
Pneumonia is the leading cause of death among children less than 5 years old worldwide. A wide range of viral, bacterial and fungal agents can cause pneumonia: although viruses are the most common etiologic agent, the severity of clinical symptoms associated with bacterial pneumonia and increasing antibiotic resistance makes bacterial pneumonia a major public health concern. Bacterial pneumonia can follow upper respiratory viral infection and complicate lower respiratory viral infection. Secondary bacterial pneumonia is a major cause of influenza-related deaths. In this review, we evaluate the following hypotheses: (i) respiratory viruses influence the etiology of pneumonia by altering bacterial community structure in the upper respiratory tract (URT) and (ii) respiratory viruses promote or inhibit colonization of the lower respiratory tract (LRT) by certain bacterial species residing in the URT. We conducted a systematic review of the literature to examine temporal associations between respiratory viruses and bacteria and a targeted review to identify potential mechanisms of interactions. We conclude that viruses both alter the bacterial community in the URT and promote bacterial colonization of the LRT. However, it is uncertain whether changes in the URT bacterial community play a substantial role in pneumonia etiology. The exception is Streptococcus pneumoniae where a strong link between viral co-infection, increased carriage and pneumococcal pneumonia has been established.
Collapse
Affiliation(s)
- Kyu Han Lee
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Betsy Foxman
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| |
Collapse
|
93
|
Zurli V, Gallotta M, Taccone M, Chiarot E, Brazzoli M, Corrente F, Bonci A, Casini D, De Gregorio E, Baudner BC, Bertholet S, Seubert A. Positive Contribution of Adjuvanted Influenza Vaccines to the Resolution of Bacterial Superinfections. J Infect Dis 2016; 213:1876-85. [PMID: 26908732 DOI: 10.1093/infdis/jiw048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/27/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Most preclinical studies assess vaccine effectiveness in single-pathogen infection models. This is unrealistic given that humans are continuously exposed to different commensals and pathogens in sequential and mixed infections. Accordingly, complications from secondary bacterial infection are a leading cause of influenza-associated morbidity and mortality. New vaccination strategies are needed to control infections on simultaneous fronts. METHODS We compared different anti-influenza vaccines for their protective potential in a model of viral infection with bacterial superinfection. Mice were immunized with H1N1/A/California/7/2009 subunit vaccines, formulated with different adjuvants inducing either T-helper type 1 (Th1) (MF59 plus CpG)-, Th1/2 (MF59)-, or Th17 (LTK63)-prone immune responses and were sequentially challenged with mouse-adapted influenza virus H1N1/A/Puerto Rico/8/1934 and Staphylococcus aureus USA300, a clonotype emerging as a leading contributor in postinfluenza pneumonia in humans. RESULTS Unadjuvanted vaccine controlled single viral infection, yet mice had considerable morbidity from viral disease and bacterial superinfection. In contrast, all adjuvanted vaccines efficiently protected mice in both conditions. Interestingly, the Th1-inducing formulation was superior to Th1/2 or Th17 inducers. CONCLUSIONS Our studies should help us better understand how differential immunity to influenza skews immune responses toward coinfecting bacteria and discover novel modes to prevent bacterial superinfections in the lungs of persons with influenza.
Collapse
Affiliation(s)
- Vanessa Zurli
- GSK Vaccines S.r.l., Vaccines Research Center, Siena Department of Biology, University of Padua, Italy
| | - Marilena Gallotta
- GSK Vaccines S.r.l., Vaccines Research Center, Siena Dynavax Technologies, Berkeley, California
| | | | | | | | | | | | | | | | | | | | - Anja Seubert
- GSK Vaccines S.r.l., Vaccines Research Center, Siena
| |
Collapse
|
94
|
Pichichero ME, Khan MN, Xu Q. Next generation protein based Streptococcus pneumoniae vaccines. Hum Vaccin Immunother 2016; 12:194-205. [PMID: 26539741 PMCID: PMC4962723 DOI: 10.1080/21645515.2015.1052198] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 04/27/2015] [Accepted: 05/12/2015] [Indexed: 12/22/2022] Open
Abstract
All currently available Streptococcus pneumoniae (Spn) vaccines have limitations due to their capsular serotype composition. Both the 23-valent Spn polysaccharide vaccine (PPV) and 7, 10, or 13-valent Spn conjugate vaccines (PCV-7, 10, -13) are serotype-based vaccines and therefore they elicit only serotype-specific immunity. Emergence of replacement Spn strains expressing other serotypes has consistently occurred following introduction of capsular serotype based Spn vaccines. Furthermore, capsular polysaccharide vaccines are less effective in protection against non-bacteremic pneumonia and acute otitis media (AOM) than against invasive pneumococcal disease (IPD). These shortcomings of capsular polysaccharide-based Spn vaccines have created high interest in development of non-serotype specific protein-based vaccines that could be effective in preventing both IPD and non-IPD infections. This review discusses the progress to date on development of Spn protein vaccine candidates that are highly conserved by all Spn strains, are highly conserved, exhibit maximal antigenicity and minimal reactogenicity to replace or complement the current capsule-based vaccines. Key to development of a protein based Spn vaccine is an understanding of Spn pathogenesis. Based on pathogenesis, a protein-based Spn vaccine should include one or more ingredients that reduce NP colonization below a pathogenic inoculum. Elimination of all Spn colonization may not be achievable or even advisable. The level of expression of a target protein antigen during pathogenesis is another key to the success of protein based vaccines.. As with virtually all currently licensed vaccines, production of a serum antibody response in response to protein based vaccines is anticipated to provide protection from Spn infections. A significant advantage that protein vaccine formulations can offer over capsule based vaccination is their potential benefits associated with natural priming and boosting to all strains of Spn. One of the most universal and comprehensive approaches of identifying novel vaccine candidates is the investigation of human sera from different disease stages of natural infections. Antigens that are robustly reactive in preliminary human serum screening constitute a pathogen-specific antigenome. This strategy has identified a number of Spn protein vaccine candidates that are moving forward in human clinical trials.
Collapse
Affiliation(s)
| | - M Nadeem Khan
- Research Institute; Rochester General Hospital; Rochester, NY USA
| | - Qingfu Xu
- Research Institute; Rochester General Hospital; Rochester, NY USA
| |
Collapse
|
95
|
Podsiad A, Standiford TJ, Ballinger MN, Eakin R, Park P, Kunkel SL, Moore BB, Bhan U. MicroRNA-155 regulates host immune response to postviral bacterial pneumonia via IL-23/IL-17 pathway. Am J Physiol Lung Cell Mol Physiol 2015; 310:L465-75. [PMID: 26589478 DOI: 10.1152/ajplung.00224.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/13/2015] [Indexed: 12/20/2022] Open
Abstract
Postinfluenza bacterial pneumonia is associated with significant mortality and morbidity. MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression posttranscriptionally. miR-155 has recently emerged as a crucial regulator of innate immunity and inflammatory responses and is induced in macrophages during infection. We hypothesized upregulation of miR-155 inhibits IL-17 and increases susceptibility to secondary bacterial pneumonia. Mice were challenged with 100 plaque-forming units H1N1 intranasally and were infected with 10(7) colony-forming units of MRSA intratracheally at day 5 postviral challenge. Lungs were harvested 24 h later, and expression of miR-155, IL-17, and IL-23 was measured by real-time RT-PCR. Induction of miR-155 was 3.6-fold higher in dual-infected lungs compared with single infection. miR-155(-/-) mice were protected with significantly lower (4-fold) bacterial burden and no differences in viral load, associated with robust induction of IL-23 and IL-17 (2.2- and 4.8-fold, respectively) postsequential challenge with virus and bacteria, compared with WT mice. Treatment with miR-155 antagomir improved lung bacterial clearance by 4.2-fold compared with control antagomir postsequential infection with virus and bacteria. Moreover, lung macrophages collected from patients with postviral bacterial pneumonia also had upregulation of miR-155 expression compared with healthy controls, consistent with observations in our murine model. This is the first demonstration that cellular miRNAs regulate postinfluenza immune response to subsequent bacterial challenge by suppressing the IL-17 pathway in the lung. Our findings suggest that antagonizing certain microRNA might serve as a potential therapeutic strategy against secondary bacterial infection.
Collapse
Affiliation(s)
- Amy Podsiad
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Megan N Ballinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Ohio State University, Columbus, Ohio; and
| | - Richard Eakin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Pauline Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Urvashi Bhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan;
| |
Collapse
|
96
|
Shrestha S, Foxman B, Berus J, van Panhuis WG, Steiner C, Viboud C, Rohani P. The role of influenza in the epidemiology of pneumonia. Sci Rep 2015; 5:15314. [PMID: 26486591 PMCID: PMC4614252 DOI: 10.1038/srep15314] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/15/2015] [Indexed: 12/25/2022] Open
Abstract
Interactions arising from sequential viral and bacterial infections play important roles in the epidemiological outcome of many respiratory pathogens. Influenza virus has been implicated in the pathogenesis of several respiratory bacterial pathogens commonly associated with pneumonia. Though clinical evidence supporting this interaction is unambiguous, its population-level effects-magnitude, epidemiological impact and variation during pandemic and seasonal outbreaks-remain unclear. To address these unknowns, we used longitudinal influenza and pneumonia incidence data, at different spatial resolutions and across different epidemiological periods, to infer the nature, timing and the intensity of influenza-pneumonia interaction. We used a mechanistic transmission model within a likelihood-based inference framework to carry out formal hypothesis testing. Irrespective of the source of data examined, we found that influenza infection increases the risk of pneumonia by ~100-fold. We found no support for enhanced transmission or severity impact of the interaction. For model-validation, we challenged our fitted model to make out-of-sample pneumonia predictions during pandemic and non-pandemic periods. The consistency in our inference tests carried out on several distinct datasets, and the predictive skill of our model increase confidence in our overall conclusion that influenza infection substantially enhances the risk of pneumonia, though only for a short period.
Collapse
Affiliation(s)
- Sourya Shrestha
- Department of Ecology & Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Center for the Study of Complex Systems, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
| | - Betsy Foxman
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joshua Berus
- Undergraduate Research Opportunity Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Willem G. van Panhuis
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh PA 15261, USA
| | - Claudia Steiner
- Healthcare Cost and Utilization Project, Center for Delivery, Organization and Markets, Agency for Healthcare Research and Quality, U.S. Department of Health and Human Services, Rockville, MD 20850, USA
| | - Cécile Viboud
- Division of International Epidemiology and Population Studies, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pejman Rohani
- Odum School of Ecology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, School of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
97
|
Khan MN, Pichichero ME. The host immune dynamics of pneumococcal colonization: implications for novel vaccine development. Hum Vaccin Immunother 2015; 10:3688-99. [PMID: 25668673 DOI: 10.4161/21645515.2014.979631] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The human nasopharynx (NP) microbiota is complex and diverse and Streptococcus pneumoniae (pneumococcus) is a frequent member. In the first few years of life, children experience maturation of their immune system thereby conferring homeostatic balance in which pneumococci are typically rendered as harmless colonizers in the upper respiratory environment. Pneumococcal carriage declines in many children before they acquire capsular-specific antibodies, suggesting a capsule antibody-independent mechanism of natural protection against pneumococcal carriage in early childhood. A child's immune system in the first few years of life is Th2-skewed so as to avoid inflammation-induced immunopathology. Understanding Th1/Th2 and Th17 ontogeny in early life and how adjuvant vaccine formulations shift the balance of T helper-cell differentiation, may facilitate the development of new protein-based pneumococcal vaccines. This article will discuss the immune dynamics of pneumococcal colonization in infants. The discussion aims to benefit the design and improvement of protein subunit-based next-generation pneumococcal vaccines.
Collapse
Affiliation(s)
- M Nadeem Khan
- a Center for Infectious Diseases and Immunology; Rochester General Hospital Research Institute ; Rochester , NY USA
| | | |
Collapse
|
98
|
Boianelli A, Nguyen VK, Ebensen T, Schulze K, Wilk E, Sharma N, Stegemann-Koniszewski S, Bruder D, Toapanta FR, Guzmán CA, Meyer-Hermann M, Hernandez-Vargas EA. Modeling Influenza Virus Infection: A Roadmap for Influenza Research. Viruses 2015; 7:5274-304. [PMID: 26473911 PMCID: PMC4632383 DOI: 10.3390/v7102875] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/28/2015] [Accepted: 09/28/2015] [Indexed: 12/24/2022] Open
Abstract
Influenza A virus (IAV) infection represents a global threat causing seasonal outbreaks and pandemics. Additionally, secondary bacterial infections, caused mainly by Streptococcus pneumoniae, are one of the main complications and responsible for the enhanced morbidity and mortality associated with IAV infections. In spite of the significant advances in our knowledge of IAV infections, holistic comprehension of the interplay between IAV and the host immune response (IR) remains largely fragmented. During the last decade, mathematical modeling has been instrumental to explain and quantify IAV dynamics. In this paper, we review not only the state of the art of mathematical models of IAV infection but also the methodologies exploited for parameter estimation. We focus on the adaptive IR control of IAV infection and the possible mechanisms that could promote a secondary bacterial coinfection. To exemplify IAV dynamics and identifiability issues, a mathematical model to explain the interactions between adaptive IR and IAV infection is considered. Furthermore, in this paper we propose a roadmap for future influenza research. The development of a mathematical modeling framework with a secondary bacterial coinfection, immunosenescence, host genetic factors and responsiveness to vaccination will be pivotal to advance IAV infection understanding and treatment optimization.
Collapse
Affiliation(s)
- Alessandro Boianelli
- Systems Medicine of Infectious Diseases, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Van Kinh Nguyen
- Systems Medicine of Infectious Diseases, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Esther Wilk
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Niharika Sharma
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | | | - Dunja Bruder
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
- Infection Immunology, Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke-University, Magdeburg 39106, Germany.
| | - Franklin R Toapanta
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig 38106, Germany.
| | - Esteban A Hernandez-Vargas
- Systems Medicine of Infectious Diseases, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany.
| |
Collapse
|
99
|
Kuchar E, Miśkiewicz K, Karlikowska M. A review of guidance on immunization in persons with defective or deficient splenic function. Br J Haematol 2015; 171:683-94. [PMID: 26315210 DOI: 10.1111/bjh.13660] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spleen acts as a blood filter and lymphopoietic organ. Asplenic and hyposplenic individuals are more susceptible to serious infections caused by encapsulated bacteria but they can be protected by antibiotic prophylaxis and immunizations. Recent progress in vaccinology means prophylaxis is now successful in the vast majority of serious infections with pneumococci, meningococci and Haemophilus influenzae type b responsible for the majority of cases of overwhelming sepsis in asplenic patients. Current guidelines are coherent. Physicians treating patients with conditions associated with hyposplenism are ethically obliged to immunize their patients using the vaccines currently available to protect them from largely preventable, life-threatening infections.
Collapse
Affiliation(s)
- Ernest Kuchar
- Department of Paediatric Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland.,Department of Paediatrics with Observation Ward, 2nd Medical Faculty, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Miśkiewicz
- Department of Paediatric Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Monika Karlikowska
- Department of Paediatric Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
100
|
Mehta D, Petes C, Gee K, Basta S. The Role of Virus Infection in Deregulating the Cytokine Response to Secondary Bacterial Infection. J Interferon Cytokine Res 2015; 35:925-34. [PMID: 26308503 DOI: 10.1089/jir.2015.0072] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Proinflammatory cytokines are produced by macrophages and dendritic cells (DCs) after infection to stimulate T helper (Th) cells, linking innate and adaptive immunity. Virus infections can deregulate the proinflammatory cytokine response like tumor necrosis factor-α and interleukin (IL)-2, making the host more susceptible to secondary bacterial infections. Studies using various viruses such as lymphocytic choriomeningitis virus, influenza A virus, and human immunodeficiency virus have revealed several intriguing mechanisms that account for the increased susceptibility to several prevalent bacterial infections. In particular, type I interferons induced during a virus infection have been observed to play a role in suppressing the production of some key antibacterial proinflammatory cytokines such as IL-23 and IL-17. Other suppressive mechanisms as a result of cytokine deregulation by viral infections include reduced function of immune cells such as DC, macrophage, natural killer, CD4(+), and CD8(+) T cells leading to impaired clearance of secondary bacterial infections. In this study, we highlight some of the immune mechanisms that become deregulated by viral infections, and can thus become defective during secondary bacterial infections.
Collapse
Affiliation(s)
- Divya Mehta
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Carlene Petes
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| |
Collapse
|