1
|
Haynes LM, Holding ML, DiGiovanni HL, Siemieniak D, Ginsburg D. High-throughput amino acid-level characterization of the interactions of plasminogen activator inhibitor-1 with variably divergent proteases. Protein Sci 2025; 34:e70088. [PMID: 40100143 PMCID: PMC11917113 DOI: 10.1002/pro.70088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/09/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
While members of large paralogous protein families share structural features, their functional niches often diverge significantly. Serine protease inhibitors (SERPINs), whose members typically function as covalent inhibitors of serine proteases, are one such family. Plasminogen activator inhibitor-1 (PAI-1) is a prototypic SERPIN, which canonically inhibits tissue- and urokinase-type plasminogen activators (tPA and uPA) to regulate fibrinolysis. PAI-1 has been shown to also inhibit other serine proteases, including coagulation factor XIIa (FXIIa) and transmembrane serine protease 2 (TMPRSS2). The structural determinants of PAI-1 inhibitory function toward these non-canonical protease targets, and the biological significance of these functions, are unknown. We applied deep mutational scanning (DMS) to assess the effects of ~80% of all possible single-amino acid substitutions in PAI-1 on its ability to inhibit three putative serine protease targets (uPA, FXIIa, and TMPRSS2). Selection with each target protease generated a unique PAI-1 mutational landscape, with the determinants of protease specificity distributed throughout PAI-1's primary sequence. Next, we conducted a comparative analysis of extant orthologous sequences, demonstrating that key residues modulating PAI-1 inhibition of uPA and FXIIa, but not TMPRSS2, are maintained by purifying selection (also referred to as "negative selection"). PAI-1's activity toward FXIIa may reflect how protease evolutionary relationships predict SERPIN functional divergence, which we support via a cophylogenetic analysis of secreted SERPINs and their cognate serine proteases. This work provides insight into the functional diversification of SERPINs and lays the framework for extending these studies to other proteases and their regulators.
Collapse
Affiliation(s)
- Laura M. Haynes
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Matthew L. Holding
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
- Department of Ecology and Evolutionary BiologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - David Siemieniak
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - David Ginsburg
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Human GeneticsUniversity of MichiganAnn ArborMichiganUSA
- Department of PediatricsUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
2
|
Zhu X, Li F, Fan B, Zhao Y, Zhou J, Wang D, Liu R, Zhao D, Fan H, Li B. TRIM28 regulates the coagulation cascade inhibited by p72 of African swine fever virus. Vet Res 2024; 55:149. [PMID: 39533356 PMCID: PMC11559047 DOI: 10.1186/s13567-024-01407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
In 2018, African swine fever virus (ASFV) emerged in China, causing extremely serious economic losses to the domestic pig industry. Infection with ASFV can cause disseminated coagulation, leading to the consumption of platelets and coagulation factors and severe bleeding. However, the mechanism of virus-induced coagulation has yet to be established. In our study, ASFV downregulated the coagulation process, as detected by D-dimer (D2D) and Factor X (F10) expression in pigs challenged with ASFV HLJ/18. In vitro, ASFV infection increased Factor IX (F9) and Factor XII (F12) expression while downregulating F10 expression in porcine alveolar macrophages (PAMs). African swine fever virus induced both intrinsic and extrinsic coagulation cascades. In addition, several encoded proteins affect the expression of the crucial coagulation protein F10, and among the encoded proteins, p72 inhibits the activity and expression of F10. Proteomic analysis also revealed that p72 is involved in the coagulation cascade. p72 can interact with F10, and its inhibitory functional domains include amino acids 423-432 and amino acids 443-452. Finally, we found that F10 and p72 interact with tripartite motif-containing protein 28 (TRIM28). TRIM28 knockdown resulted in a decrease in F10 expression. Importantly, TRIM28 contributes to the reduction in F10 protein expression regulated by p72. Our findings revealed an inhibitory effect of the viral protein p72 on the ASFV infection-induced coagulation cascade and revealed a role of TRIM28 in reducing F10 expression, revealing a molecular mechanism of ASFV-associated coagulation.
Collapse
Affiliation(s)
- Xuejiao Zhu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China
| | - Fang Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China
| | - Yongxiang Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China
| | - Junming Zhou
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China
| | - Dandan Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China
| | - Renqiang Liu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dongming Zhao
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, Jiangsu Province, China.
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of the Ministry of Science and Technology, Nanjing, China.
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China.
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biology, Taizhou, 225300, China.
| |
Collapse
|
3
|
Haynes LM, Holding ML, DiGiovanni H, Siemieniak D, Ginsburg D. High-throughput amino acid-level characterization of the interactions of plasminogen activator inhibitor-1 with variably divergent proteases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.612699. [PMID: 39345533 PMCID: PMC11429915 DOI: 10.1101/2024.09.16.612699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
While members of large paralogous protein families share structural features, their functional niches often diverge significantly. Serine protease inhibitors (SERPINs), whose members typically function as covalent inhibitors of serine proteases, are one such family. Plasminogen activator inhibitor-1 (PAI-1) is a prototypic SERPIN, which canonically inhibits tissue-and urokinase-type plasminogen activators (tPA and uPA) to regulate fibrinolysis. PAI-1 has been shown to also inhibit other serine proteases, including coagulation factor XIIa (FXIIa) and transmembrane serine protease 2 (TMPRSS2). The structural determinants of PAI-1 inhibitory function toward these non-canonical protease targets, and the biological significance of these functions, are unknown. We applied deep mutational scanning (DMS) to assess the effects of ∼80% of all possible single amino acid substitutions in PAI-1 on its ability to inhibit three putative serine protease targets (uPA, FXIIa, and TMPRSS2). Selection with each target protease generated a unique PAI-1 mutational landscape, with the determinants of protease specificity distributed throughout PAI-1's primary sequence. Next, we conducted a comparative analysis of extant orthologous sequences, demonstrating that key residues modulating PAI-1 inhibition of uPA and FXIIa, but not TMPRSS2, are maintained by purifying selection. PAI-1's activity toward FXIIa may reflect how protease evolutionary relationships predict SERPIN functional divergence, which we support via a cophylogenetic analysis of all secreted SERPINs and their cognate serine proteases. This work provides insight into the functional diversification of SERPINs and lays the framework for extending these studies to other proteases and their regulators.
Collapse
|
4
|
Clarke M, Falcione S, Boghozian R, Todoran R, Zhang Y, C. Real MG, StPierre A, Joy T, Jickling GC. Viral Infection and Ischemic Stroke: Emerging Trends and Mechanistic Insights. J Am Heart Assoc 2024; 13:e035892. [PMID: 39258541 PMCID: PMC11935600 DOI: 10.1161/jaha.124.035892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
Population studies have suggested that viral infections may be contributing to risk of ischemic stroke, although the mechanisms for this are unclear. In this review, we examine the epidemiological evidence supporting the involvement of viral diseases, including influenza, COVID-19, chronic herpesvirus infections, and hepatitis C in current trends of stroke incidence. To support these associations, we highlight the virus-host interactions that are critical in the context of stroke, including direct effects of acute and persistent viral infections on vascular function, inflammation, and thrombosis. Additionally, we evaluate the systemic changes that occur during viral infection that can predispose individuals to ischemic stroke, including alterations in blood pressure regulation, coagulation, and lipid metabolism. Our review emphasizes the need to further elucidate precise mechanisms involved in viral infections and stroke risk. Future research will inform the development of targeted interventions for stroke prevention in the context of viral diseases.
Collapse
Affiliation(s)
- Michael Clarke
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
| | - Sarina Falcione
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Roobina Boghozian
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Raluca Todoran
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Yiran Zhang
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Maria Guadalupe C. Real
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Alexis StPierre
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Twinkle Joy
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Glen C. Jickling
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
5
|
Montone RA, Camilli M, Calvieri C, Magnani G, Bonanni A, Bhatt DL, Rajagopalan S, Crea F, Niccoli G. Exposome in ischaemic heart disease: beyond traditional risk factors. Eur Heart J 2024; 45:419-438. [PMID: 38238478 PMCID: PMC10849374 DOI: 10.1093/eurheartj/ehae001] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 02/09/2024] Open
Abstract
Ischaemic heart disease represents the leading cause of morbidity and mortality, typically induced by the detrimental effects of risk factors on the cardiovascular system. Although preventive interventions tackling conventional risk factors have helped to reduce the incidence of ischaemic heart disease, it remains a major cause of death worldwide. Thus, attention is now shifting to non-traditional risk factors in the built, natural, and social environments that collectively contribute substantially to the disease burden and perpetuate residual risk. Of importance, these complex factors interact non-linearly and in unpredictable ways to often enhance the detrimental effects attributable to a single or collection of these factors. For this reason, a new paradigm called the 'exposome' has recently been introduced by epidemiologists in order to define the totality of exposure to these new risk factors. The purpose of this review is to outline how these emerging risk factors may interact and contribute to the occurrence of ischaemic heart disease, with a particular attention on the impact of long-term exposure to different environmental pollutants, socioeconomic and psychological factors, along with infectious diseases such as influenza and COVID-19. Moreover, potential mitigation strategies for both individuals and communities will be discussed.
Collapse
Affiliation(s)
- Rocco A Montone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Giulia Magnani
- Department of Medicine, University of Parma, Parma, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | | |
Collapse
|
6
|
Whyte CS. All tangled up: interactions of the fibrinolytic and innate immune systems. Front Med (Lausanne) 2023; 10:1212201. [PMID: 37332750 PMCID: PMC10272372 DOI: 10.3389/fmed.2023.1212201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
The hemostatic and innate immune system are intertwined processes. Inflammation within the vasculature promotes thrombus development, whilst fibrin forms part of the innate immune response to trap invading pathogens. The awareness of these interlinked process has resulted in the coining of the terms "thromboinflammation" and "immunothrombosis." Once a thrombus is formed it is up to the fibrinolytic system to resolve these clots and remove them from the vasculature. Immune cells contain an arsenal of fibrinolytic regulators and plasmin, the central fibrinolytic enzyme. The fibrinolytic proteins in turn have diverse roles in immunoregulation. Here, the intricate relationship between the fibrinolytic and innate immune system will be discussed.
Collapse
|
7
|
Bivalirudin exerts antiviral activity against respiratory syncytial virus-induced lung infections in neonatal mice. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:415-425. [PMID: 36651544 DOI: 10.2478/acph-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 01/26/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of small airways inflammation in the lungs (bronchiolitis) in neonates and immunocompromised adults. The deregulation of cellular and plasma components leads to increased morbidity and mortality. The activation of the clotting cascade plays a key role in the progression of disease severity during viral infection. The current investigation studied the effect of bivalirudin (BR) on the progression and cellular effects of RSV-induced infection in the neonatal mice model. Mice (5-7 days old) were inoculated intranasally with RSV with or without BR administration (2 mg kg-1 day-1, i.v.) for 2 weeks. Tissue histopathology, inflammatory signalling genes such as TLR, and cytokines were analyzed. The results showed pneumocytes exhibiting nuclear pyknosis, cellular infiltration in lung tissue and increased lung titers in RSV-infected mice compared to the control. Furthermore, RSV-infected mice demonstrated altered clotting parameters such as D-dimer, soluble thrombomodulin, and increased inflammatory cytokines IL-5, 6, IFN-γ, IL-13, and CXCL1. Additionally, the mRNA expression analysis displayed increased levels of IL-33, TLR3, and TLR7 genes in RSV-infected lung tissue. Further, to delineate the role of micro RNAs, the qRT-PCR analysis was done, and the results displayed an increase in miR-136, miR-30b, and let-7i. At the same time, the down-regulated expression of miR-221 in RSV-infected mice compared to the control. BR treatment reduced the cellular infiltration with reduced inflammatory cytokines and normalized clotting indices. Thus, the study shows that RSV infection induces specific changes in lung tissue and the clotting related signalling mechanism. Additionally, BR treatment significantly reduces bronchiolitis and prevents the severity of the infections suggesting that BR can possibly be used to reduce the viral-mediated infections in neonates.
Collapse
|
8
|
Froggatt HM, Heaton NS. Nonrespiratory sites of influenza-associated disease: mechanisms and experimental systems for continued study. FEBS J 2022; 289:4038-4060. [PMID: 35060315 PMCID: PMC9300775 DOI: 10.1111/febs.16363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022]
Abstract
The productive replication of human influenza viruses is almost exclusively restricted to cells in the respiratory tract. However, a key aspect of the host response to viral infection is the production of inflammatory cytokines and chemokines that are not similarly tissue restricted. As such, circulating inflammatory mediators, as well as the resulting activated immune cells, can induce damage throughout the body, particularly in individuals with underlying conditions. As a result, more holistic experimental approaches are required to fully understand the pathogenesis and scope of influenza virus-induced disease. This review summarizes what is known about some of the most well-appreciated nonrespiratory tract sites of influenza virus-induced disease, including neurological, cardiovascular, gastrointestinal, muscular and fetal developmental phenotypes. In the context of this discussion, we describe the in vivo experimental systems currently being used to study nonrespiratory symptoms. Finally, we highlight important future questions and potential models that can be used for a more complete understanding of influenza virus-induced disease.
Collapse
Affiliation(s)
- Heather M. Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
9
|
Endothelial dysfunction contributes to severe COVID-19 in combination with dysregulated lymphocyte responses and cytokine networks. Signal Transduct Target Ther 2021; 6:418. [PMID: 34893580 PMCID: PMC8661333 DOI: 10.1038/s41392-021-00819-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/22/2021] [Accepted: 10/17/2021] [Indexed: 02/07/2023] Open
Abstract
The systemic processes involved in the manifestation of life-threatening COVID-19 and in disease recovery are still incompletely understood, despite investigations focusing on the dysregulation of immune responses after SARS-CoV-2 infection. To define hallmarks of severe COVID-19 in acute disease (n = 58) and in disease recovery in convalescent patients (n = 28) from Hannover Medical School, we used flow cytometry and proteomics data with unsupervised clustering analyses. In our observational study, we combined analyses of immune cells and cytokine/chemokine networks with endothelial activation and injury. ICU patients displayed an altered immune signature with prolonged lymphopenia but the expansion of granulocytes and plasmablasts along with activated and terminally differentiated T and NK cells and high levels of SARS-CoV-2-specific antibodies. The core signature of seven plasma proteins revealed a highly inflammatory microenvironment in addition to endothelial injury in severe COVID-19. Changes within this signature were associated with either disease progression or recovery. In summary, our data suggest that besides a strong inflammatory response, severe COVID-19 is driven by endothelial activation and barrier disruption, whereby recovery depends on the regeneration of the endothelial integrity.
Collapse
|
10
|
Narasimhan B, Lorente-Ros M, Aguilar-Gallardo JS, Lizardo CP, Narasimhan H, Morton C, Donahue KR, Aronow WS. Anticoagulation in COVID-19: a review of current literature and guidelines. Hosp Pract (1995) 2021; 49:307-324. [PMID: 34807786 DOI: 10.1080/21548331.2021.2007648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 infections are associated with greater risk of both arterial and venous thromboembolic events.Pathophysiology and Clinical implications: This has been attributed to a florid proinflammatory state resulting in microvascular dysfunction, activation of platelets and procoagulant systems as well as possible direct endothelial injury. The associated morbidity and mortality of these events has prompted much speculation and varied anticoagulation and fibrinolytic strategies based on multiple criteria including disease severity and biomarkers. No clear definitive benefit has been established with these approaches, which have frequently led to greater bleeding complications without significant mortality benefit.Overview: In this review, we outline the burden of these thromboembolic events in coronavirus disease-2019 (COVID-19) as well as the hypothesized contributory biological mechanisms. Finally, we provide a brief overview of the major clinical studies on the topic, and end with a summary of major societal guideline recommendations on anticoagulation in COVID-19.
Collapse
Affiliation(s)
- Bharat Narasimhan
- Debakey Cardiovascular Center, Houston Methodist Hospital-Texas Medical Center, Houston, TX, USA
| | - Marta Lorente-Ros
- Department of Medicine, Mount Sinai Morningside-West, the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jose S Aguilar-Gallardo
- Department of Medicine, Mount Sinai Morningside-West, the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Perez Lizardo
- Department of Medicine, Mount Sinai Morningside-West, the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Celia Morton
- Department of Pharmacy, Houston Methodist Hospital-Texas Medical Center, Houston, TX, USA
| | - Kevin R Donahue
- Department of Pharmacy, Houston Methodist Hospital-Texas Medical Center, Houston, TX, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center/New York Medical College, Valhalla, NY, USA
| |
Collapse
|
11
|
Dai Q, Ye M, Tang Z, Yu K, Gao Y, Yang Z, Zheng J, Zuo S, Liu Y, Xie F, Han Q, He H, Wang H. Comparison of severe and critical COVID-19 patients imported from Russia with and without influenza A infection in Heilongjiang Province: a retrospective study. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1446. [PMID: 34733998 PMCID: PMC8506785 DOI: 10.21037/atm-21-3912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022]
Abstract
Background The rapid spread of coronavirus disease-19 (COVID-19) poses a global health emergency, and cases entering China from Russia are quite diverse. This study explored and compared the clinical characteristics and outcomes of severe and critically ill COVID-19 patients from Russia with and without influenza A infection, treated in a northern Chinese hospital (Russia imported patients). Methods A total of 32 severe and critically ill Russia-imported COVID-19 patients treated in the Heilongjiang Imported Severe and Critical COVID-19 Treatment Center from April 6 to May 11, 2020 were included, including 8 cases (group A) with and 24 cases (group B) without influenza A infection. The clinical characteristics of each group were compared, including prolonged hospital stay, duration of oxygen therapy, time from onset to a negative SARS-CoV-2 qRT-PCR RNA (Tneg) result, and duration of bacterial infection. Results The results showed that blood group, PaO2/FiO2, prothrombin time (PT), prothrombin activity (PTA), computed tomography (CT) score, hospital stay, duration of oxygenation therapy, Tneg, and duration of bacterial infection were statistically different between the two groups (P<0.05). Multivariant regression analysis showed that the Sequential Organ Failure Assessment (SOFA) score, C-reactive protein (CRP), and influenza A infection were factors influencing hospital stay; SOFA score, CRP, and CT score were factors influencing the duration of oxygenation therapy; PaO2/FiO2, platelet count (PLT), and CRP were factors influencing Tneg; and gender, SOFA score, and influenza A infection were factors influencing the duration of bacterial infection. Conclusions Influenza A infection is common in Russia-imported COVID-19 patients, which can prolong the hospital stay and duration of bacterial infection. Routinely screening and treating influenza A should be conducted early in such patients.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming Ye
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiqiang Tang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Gao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyu Yang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junbo Zheng
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu Zuo
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Liu
- Department of Medical Statistics, Harbin Medical University, Harbin, China
| | - Fengjie Xie
- Department of Critical Care Medicine, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Qiuyuan Han
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hua He
- Department of Infectious Disease, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Hongliang Wang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Lehman A, Fischer G, Steiner M. Diffuse Thromboses in a 2-year-old With Sepsis and Respiratory Failure. Pediatr Infect Dis J 2021; 40:952-954. [PMID: 34525009 DOI: 10.1097/inf.0000000000003084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Alice Lehman
- From the Department of Pediatrics, Division of Global Pediatrics
| | - Gwenyth Fischer
- Department of Pediatrics, Division of Pediatric Critical Care
| | - Marie Steiner
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Division of Pediatric Critical Care, University of Minnesota Medical School, Masonic Children's Hospital, Minneapolis, Minnesota
| |
Collapse
|
13
|
Ohno M, Kakino A, Sekiya T, Nomura N, Shingai M, Sawamura T, Kida H. Critical role of oxidized LDL receptor-1 in intravascular thrombosis in a severe influenza mouse model. Sci Rep 2021; 11:15675. [PMID: 34344944 PMCID: PMC8333315 DOI: 10.1038/s41598-021-95046-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 11/10/2022] Open
Abstract
Although coagulation abnormalities, including microvascular thrombosis, are thought to contribute to tissue injury and single- or multiple-organ dysfunction in severe influenza, the detailed mechanisms have yet been clarified. This study evaluated influenza-associated abnormal blood coagulation utilizing a severe influenza mouse model. After infecting C57BL/6 male mice with intranasal applications of 500 plaque-forming units of influenza virus A/Puerto Rico/8/34 (H1N1; PR8), an elevated serum level of prothrombin fragment 1 + 2, an indicator for activated thrombin generation, was observed. Also, an increased gene expression of oxidized low-density lipoprotein (LDL) receptor-1 (Olr1), a key molecule in endothelial dysfunction in the progression of atherosclerosis, was detected in the aorta of infected mice. Body weight decrease, serum levels of cytokines and chemokines, viral load, and inflammation in the lungs of infected animals were similar between wild-type and Olr1 knockout (KO) mice. In contrast, the elevation of prothrombin fragment 1 + 2 levels in the sera and intravascular thrombosis in the lungs by PR8 virus infection were not induced in KO mice. Collectively, the results indicated that OLR1 is a critical host factor in intravascular thrombosis as a pathogeny of severe influenza. Thus, OLR1 is a promising novel therapeutic target for thrombosis during severe influenza.
Collapse
Affiliation(s)
- Marumi Ohno
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Akemi Kakino
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Matsumoto, Japan
| | - Toshiki Sekiya
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Naoki Nomura
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Masashi Shingai
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Matsumoto, Japan
| | - Hiroshi Kida
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
14
|
Khorramdelazad H, Kazemi MH, Najafi A, Keykhaee M, Zolfaghari Emameh R, Falak R. Immunopathological similarities between COVID-19 and influenza: Investigating the consequences of Co-infection. Microb Pathog 2021; 152:104554. [PMID: 33157216 PMCID: PMC7607235 DOI: 10.1016/j.micpath.2020.104554] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been a global public health emergency since December 2019, and so far, more than 980,000 people (until September 24, 2020) around the world have died. SARS-CoV-2 mimics the influenza virus regarding methods and modes of transmission, clinical features, related immune responses, and seasonal coincidence. Accordingly, co-infection by these viruses is imaginable because some studies have reported several cases with SARS-CoV-2 and influenza virus co-infection. Given the importance of the mentioned co-infection and the coming influenza season, it is essential to recognize the similarities and differences between the symptoms, immunopathogenesis and treatment of SARS-CoV-2 and influenza virus. Therefore, we reviewed the virology, clinical features, and immunopathogenesis of both influenza virus and SARS-CoV-2 and evaluated outcomes in cases with SARS-CoV-2 and influenza virus co-infection.
Collapse
Affiliation(s)
- Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), 14965/161, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Kruip MJHA, Cannegieter SC, ten Cate H, van Gorp ECM, Juffermans NP, Klok FA, Maas C, Vonk‐Noordegraaf A, Dutch COVID Thrombosis Coalition study group. Caging the dragon: Research approach to COVID-19-related thrombosis. Res Pract Thromb Haemost 2021; 5:278-290. [PMID: 33733026 PMCID: PMC7938618 DOI: 10.1002/rth2.12470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
The incidence of venous thrombosis, mostly pulmonary embolism (PE), ranging from local immunothrombosis to central emboli, but also deep vein thrombosis (DVT) in people with coronavirus disease 2019 (COVID-19) is reported to be remarkably high. The relevance of better understanding, predicting, treating, and preventing COVID-19-associated venous thrombosis meets broad support, as can be concluded from the high number of research, review, and guideline papers that have been published on this topic. The Dutch COVID & Thrombosis Coalition (DCTC) is a multidisciplinary team involving a large number of Dutch experts in the broad area of venous thrombosis and hemostasis research, combined with experts on virology, critically ill patients, pulmonary diseases, and community medicine, across all university hospitals and many community hospitals in the Netherlands. Within the consortium, clinical data of at least 5000 admitted COVID-19-infected individuals are available, including substantial collections of biobanked materials in an estimated 3000 people. In addition to considerable experience in preclinical and clinical thrombosis research, the consortium embeds virology-hemostasis research models within unique biosafety facilities to address fundamental questions on the interaction of virus with epithelial and vascular cells, in relation to the coagulation and inflammatory system. The DCTC has initiated a comprehensive research program to answer many of the current questions on the pathophysiology and best anticoagulant treatment of COVID-19-associated thrombotic complications. The research program was funded by grants of the Netherlands Thrombosis Foundation and the Netherlands Organization for Health Research and Development. Here, we summarize the design and main aims of the research program.
Collapse
Affiliation(s)
- Marieke J. H. A. Kruip
- Department of HematologyErasmus MCErasmus University Medical CenterRotterdamThe Netherlands
| | - Suzanne C. Cannegieter
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine – Thrombosis and HemostasisLeiden University Medical CenterLeidenThe Netherlands
| | - Hugo ten Cate
- Maastricht University Medical Center and CARIMMaastrichtThe Netherlands
| | - Eric C. M. van Gorp
- Department of ViroscienceErasmus MCErasmus University Medical CenterRotterdamThe Netherlands
- Department of Infectious DiseasesErasmus MCErasmus University Medical CenterRotterdamThe Netherlands
| | - Nicole P. Juffermans
- Laboratory of Experimental Intensive Care and AnesthesiologyAmsterdam UMC ‐ Location AMCAmsterdamThe Netherlands
- Department of Intensive CareOLVG HospitalAmsterdamThe Netherlands
| | - Frederikus A. Klok
- Department of Medicine – Thrombosis and HemostasisLeiden University Medical CenterLeidenThe Netherlands
| | - Coen Maas
- Department of Clinical Chemistry and HematologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Anton Vonk‐Noordegraaf
- Dept of Pulmonary MedicineAmsterdam Cardiovascular SciencesAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | | |
Collapse
|
16
|
Latreille E, Lee WL. Interactions of Influenza and SARS-CoV-2 with the Lung Endothelium: Similarities, Differences, and Implications for Therapy. Viruses 2021; 13:161. [PMID: 33499234 PMCID: PMC7911974 DOI: 10.3390/v13020161] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory viruses such as influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are a constant threat to public health given their ability to cause global pandemics. Infection with either virus may lead to aberrant host responses, such as excessive immune cell recruitment and activation, dysregulated inflammation, and coagulopathy. These may contribute to the development of lung edema and respiratory failure. An increasing amount of evidence suggests that lung endothelial cells play a critical role in the pathogenesis of both viruses. In this review, we discuss how infection with influenza or SARS-CoV-2 may induce endothelial dysfunction. We compare the effects of infection of these two viruses, how they may contribute to pathogenesis, and discuss the implications for potential treatment. Understanding the differences between the effects of these two viruses on lung endothelial cells will provide important insight to guide the development of therapeutics.
Collapse
Affiliation(s)
- Elyse Latreille
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Warren L. Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Interdepartmental Division of Critical Care and the Department of Medicine, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
17
|
Hanff TC, Mohareb AM, Giri J, Cohen JB, Chirinos JA. Thrombosis in COVID-19. Am J Hematol 2020; 95:1578-1589. [PMID: 32857878 PMCID: PMC7674272 DOI: 10.1002/ajh.25982] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Thrombotic complications are frequent in COVID-19 and contribute significantly to mortality and morbidity. We review several mechanisms of hypercoagulability in sepsis that may be upregulated in COVID-19. These include immune-mediated thrombotic mechanisms, complement activation, macrophage activation syndrome, antiphospholipid antibody syndrome, hyperferritinemia, and renin-angiotensin system dysregulation. We highlight biomarkers within each pathway with potential prognostic value in COVID-19. Lastly, recent observational studies have evaluated a role for the expanded use of therapeutic anticoagulation in COVID-19. We review strengths and weaknesses of these studies, and we also discuss the hypothetical benefit and anticipated challenges of fibrinolytic therapy in COVID-19.
Collapse
Affiliation(s)
- Thomas C. Hanff
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, Philadelphia, PA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA
| | - Amir M. Mohareb
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jay Giri
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, Philadelphia, PA
| | - Jordana B. Cohen
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Julio A. Chirinos
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
18
|
Gopal R, Marinelli MA, Alcorn JF. Immune Mechanisms in Cardiovascular Diseases Associated With Viral Infection. Front Immunol 2020; 11:570681. [PMID: 33193350 PMCID: PMC7642610 DOI: 10.3389/fimmu.2020.570681] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Influenza virus infection causes 3-5 million cases of severe illness and 250,000-500,000 deaths worldwide annually. Although pneumonia is the most common complication associated with influenza, there are several reports demonstrating increased risk for cardiovascular diseases. Several clinical case reports, as well as both prospective and retrospective studies, have shown that influenza can trigger cardiovascular events including myocardial infarction (MI), myocarditis, ventricular arrhythmia, and heart failure. A recent study has demonstrated that influenza-infected patients are at highest risk of having MI during the first seven days of diagnosis. Influenza virus infection induces a variety of pro-inflammatory cytokines and chemokines and recruitment of immune cells as part of the host immune response. Understanding the cellular and molecular mechanisms involved in influenza-associated cardiovascular diseases will help to improve treatment plans. This review discusses the direct and indirect effects of influenza virus infection on triggering cardiovascular events. Further, we discussed the similarities and differences in epidemiological and pathogenic mechanisms involved in cardiovascular events associated with coronavirus disease 2019 (COVID-19) compared to influenza infection.
Collapse
Affiliation(s)
- Radha Gopal
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States
| | | | | |
Collapse
|
19
|
Aliter KF, Al-Horani RA. Thrombin Inhibition by Argatroban: Potential Therapeutic Benefits in COVID-19. Cardiovasc Drugs Ther 2020; 35:195-203. [PMID: 32870433 PMCID: PMC7459262 DOI: 10.1007/s10557-020-07066-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 12/15/2022]
Abstract
Thrombin is a trypsin-like serine protease with multiple physiological functions. Its role in coagulation and thrombosis is well-established. Nevertheless, thrombin also plays a major role in inflammation by activating protease-activated receptors. In addition, thrombin is also involved in angiogenesis, fibrosis, and viral infections. Considering the pathogenesis of COVID-19 pandemic, thrombin inhibitors may exert multiple potential therapeutic benefits including antithrombotic, anti-inflammatory, and antiviral activities. In this review, we describe the clinical features of COVID-19, the thrombin’s roles in various pathologies, and the potential of argatroban in COVID-19 patients. Argatroban is a synthetic, small molecule, direct, competitive, and selective inhibitor of thrombin. It is approved to parenterally prevent and/or treat heparin-induced thrombocytopenia in addition to other thrombotic conditions. Argatroban also possesses anti-inflammatory and antiviral activities and has a well-established pharmacokinetics profile. It also appears to lack a significant risk of drug–drug interactions with therapeutics currently being evaluated for COVID-19. Thus, argatroban presents a substantial promise in treating severe cases of COVID-19; however, this promise is yet to be established in randomized, controlled clinical trials.
Collapse
Affiliation(s)
- Kholoud F Aliter
- Department of Chemistry, School of STEM, Dillard University, New Orleans, LA, 70122, USA
| | - Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125-1089, USA.
| |
Collapse
|
20
|
Mohammad MA, Tham J, Koul S, Rylance R, Bergh C, Erlinge D, Fröbert O. Association of acute myocardial infarction with influenza: A nationwide observational study. PLoS One 2020; 15:e0236866. [PMID: 32760080 PMCID: PMC7410234 DOI: 10.1371/journal.pone.0236866] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/15/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Influenza may precipitate cardiovascular disease, but influenza typically peaks in winter, coinciding with other triggers of myocardial infarction (MI) such as low air temperature, high wind velocity, low atmospheric pressure, and short sunshine duration. Objective We aimed to determine the relationship of week-to-week variation in influenza cases and acute MI, controlling for meteorological factors in a nationwide population. Methods Weekly laboratory-confirmed influenza case reports were obtained from the Public Health Agency of Sweden from 2009 to 2016 and merged with the nationwide SWEDEHEART MI registry. Weekly incidence of MI was studied with regard to number of influenza cases stratified into tertiles of 0–16, 17–164, and >164 cases/week. Incidence rate ratios (IRR) were calculated using a count regression model for each category and compared to a non-influenza period as reference, controlling for air temperature, atmospheric pressure, wind velocity, and sunshine duration. Results A total of 133562 MI events was reported to the registry during the study period. Weeks with influenza cases were associated with higher incidence of MI than those without in unadjusted analysis for overall MI, ST-elevation MI and non ST-elevation MI independently. During the influenza season, weeks with 0–16 reported cases/week were not associated with MI incidence after adjusting for weather parameters, adjusted IRR for MI was 1.03 (95% CI 1.00–1.06, P = 0.09). However, weeks with more cases reported were associated with MI incidence: 17–163 reported cases/week, adjusted IRR = 1.05 (95% CI 1.02–1.08, P = 0.003); and for ≥164 cases/week, the IRR = 1.06 (95% CI 1.02–1.09, P = 0.002). Results were consistent across a large range of subgroups. Conclusions In this nationwide observational study, we found an association of incidence of MI with incidence of influenza cases beyond what could be explained by meteorological factors.
Collapse
Affiliation(s)
- Moman A. Mohammad
- Department of Cardiology, Clinical Sciences Lund University, Lund, Sweden
- * E-mail:
| | - Johan Tham
- Infectious Diseases Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Sasha Koul
- Department of Cardiology, Clinical Sciences Lund University, Lund, Sweden
| | - Rebecca Rylance
- Department of Cardiology, Clinical Sciences Lund University, Lund, Sweden
| | - Cecilia Bergh
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - David Erlinge
- Department of Cardiology, Clinical Sciences Lund University, Lund, Sweden
| | - Ole Fröbert
- Department of Cardiology, Faculty of Medicine and Health; Örebro University, Örebro, Sweden
| |
Collapse
|
21
|
Maloney BE, Perera KD, Saunders DRD, Shadipeni N, Fleming SD. Interactions of viruses and the humoral innate immune response. Clin Immunol 2020; 212:108351. [PMID: 32028020 DOI: 10.1016/j.clim.2020.108351] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/01/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022]
Abstract
The innate immune response is crucial for defense against virus infections where the complement system, coagulation cascade and natural antibodies play key roles. These immune components are interconnected in an intricate network and are tightly regulated to maintain homeostasis and avoid uncontrolled immune responses. Many viruses in turn have evolved to modulate these interactions through various strategies to evade innate immune activation. This review summarizes the current understanding on viral strategies to inhibit the activation of complement and coagulation cascades, evade natural antibody-mediated clearance and utilize complement regulatory mechanisms to their advantage.
Collapse
Affiliation(s)
- Bailey E Maloney
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Krishani Dinali Perera
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Danielle R D Saunders
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Naemi Shadipeni
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
22
|
Ishiguro T, Matsuo K, Fujii S, Takayanagi N. Acute thrombotic vascular events complicating influenza-associated pneumonia. Respir Med Case Rep 2019; 28:100884. [PMID: 31245274 PMCID: PMC6582236 DOI: 10.1016/j.rmcr.2019.100884] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 11/30/2022] Open
Abstract
A 58-year-old man with previous myocardial infarction presented to our hospital with fever, cough, and dyspnea. PCR testing with nasopharyngeal swabs confirmed influenza virus infection, and enhanced computed tomography and transthoracic echocardiography revealed bilateral ground-glass opacities and consolidation, deep venous thrombosis, acute pulmonary artery embolism, and acute arterial embolism that appeared to originate from thrombus in the left ventricle. Combination of a neuraminidase inhibitor, antibiotics, an anticoagulant, and anti-platelet agent improved these complications; however, amputation of the patient's right foot was required. Because influenza can cause vascular events, physicians should pay attention to this complication in patients with influenza-associated pneumonia.
Collapse
Affiliation(s)
- Takashi Ishiguro
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Saitama, Japan
| | - Keisuke Matsuo
- Department of Cardiology, Saitama Cardiovascular and Respiratory Center, Saitama, Japan
| | - Shinya Fujii
- Department of Cardiology, Saitama Cardiovascular and Respiratory Center, Saitama, Japan
| | - Noboru Takayanagi
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Saitama, Japan
| |
Collapse
|
23
|
Lam F, Chen TL, Shih CC, Lin CS, Yeh CC, Lee YJ, Hu CJ, Chiou HY, Liao CC. Protective effect of influenza vaccination on outcomes in geriatric stroke patients: A nationwide matched cohort study. Atherosclerosis 2019; 282:85-90. [PMID: 30711633 DOI: 10.1016/j.atherosclerosis.2019.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/14/2018] [Accepted: 01/10/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND AIMS The effects of influenza vaccination (IV) on stroke outcomes are unclear. The purpose of this study is to evaluate the outcomes after stroke in elderly individuals who have received an IV. METHODS We used Taiwan's National Health Insurance Research Database 2000-2009 claims data to conduct a nested stroke cohort study including 148,909 hospitalized stroke patients aged 66 years and older. Using a matching procedure by propensity score, we selected 25,248 stroke patients with IV and 25,248 stroke patients without IV for comparison. Logistic regression was used to calculate the odds ratios (ORs) and 95% CIs of post-stroke complications and in-hospital mortality associated with IV. RESULTS Stroke patients with IV had significantly lower risks of post-stroke pneumonia (OR = 0.79; 95% CI, 0.74-0.83), septicemia (OR = 0.78; 95% CI, 0.70-0.86), urinary tract infection (OR = 0.87; 95% CI, 0.83-0.92), and 30-day in-hospital mortality (OR = 0.60; 95% CI, 0.54-0.67) compared with non-IV stroke patients. Vaccinated stroke patients also had shorter hospital stays (p < 0.0001) and less medical expenditures (p < 0.0001) during stroke admission than the control group. Lower rates of post-stroke adverse events in patients with IV were noted in both sexes of all age groups with various types of stroke. CONCLUSIONS Stroke patients with IV showed fewer complications and lower mortality compared with non-IV patients. These findings suggest the urgent need to promote IV for this susceptible population of stroke patients.
Collapse
Affiliation(s)
- Fai Lam
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ta-Liang Chen
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Chuan Shih
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
| | - Chao-Shun Lin
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Surgery, University of Illinois, Chicago, IL, USA
| | - Yuarn-Jang Lee
- Division of Infectious Diseases, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuan Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Department of Anesthesiology, Shuan Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
24
|
The Streptococcus pyogenes fibronectin/tenascin-binding protein PrtF.2 contributes to virulence in an influenza superinfection. Sci Rep 2018; 8:12126. [PMID: 30108238 PMCID: PMC6092322 DOI: 10.1038/s41598-018-29714-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/13/2018] [Indexed: 12/13/2022] Open
Abstract
Influenza A virus (IAV) and Streptococcus pyogenes (the group A Streptococcus; GAS) are important contributors to viral-bacterial superinfections, which result from incompletely defined mechanisms. We identified changes in gene expression following IAV infection of A549 cells. Changes included an increase in transcripts encoding proteins with fibronectin-type III (FnIII) domains, such as fibronectin (Fn), tenascin N (TNN), and tenascin C (TNC). We tested the idea that increased expression of TNC may affect the outcome of an IAV-GAS superinfection. To do so, we created a GAS strain that lacked the Fn-binding protein PrtF.2. We found that the wild-type GAS strain, but not the mutant, co-localized with TNC and bound to purified TNC. In addition, adherence of the wild-type strain to IAV-infected A549 cells was greater compared to the prtF.2 mutant. The wild-type strain was also more abundant in the lungs of mice 24 hours after superinfection compared to the mutant strain. Finally, all mice infected with IAV and the prtF.2 mutant strain survived superinfection compared to only 42% infected with IAV and the parental GAS strain, indicating that PrtF.2 contributes to virulence in a murine model of IAV-GAS superinfection.
Collapse
|
25
|
Lê BV, Jandrot-Perrus M, Couture C, Checkmahomed L, Venable MC, Hamelin MÈ, Boivin G. Evaluation of anticoagulant agents for the treatment of human metapneumovirus infection in mice. J Gen Virol 2018; 99:1367-1380. [PMID: 30102144 DOI: 10.1099/jgv.0.001135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thrombin has been demonstrated to be involved in several viral diseases including human metapneumovirus (hMPV) infections. We previously showed that immediate administration of thrombin inhibitor argatroban post-infection protected mice against hMPV disease. This current work aims at determining whether warfarin and heparin, two other anticoagulants inhibiting thrombin formation and activities, may also be used for treatment against hMPV in vivo. We found that immediate injections of argatroban, warfarin or heparin after virus challenge protected mice against hMPV infection, as evidenced by decreased or no mortality, less weight loss, reduced viral load and attenuated inflammation. However, delayed treatments starting 1 day post-infection with argatroban or warfarin almost did not impact the survival whereas delayed treatment with heparin induced an increased mortality during infection. Moreover, these treatments also did not reduce weight loss, viral replication and inflammation. In agreement with these results, thrombin generation was decreased upon immediate anticoagulant treatments but was unaltered upon delayed treatments. Thus, thrombin generation occurs at the onset of hMPV infection and thrombin inhibition may be only useful for the treatment of this disease when initiated in the early stage. In this case, heparin is not recommended because of its reduced efficacy on mortality in infected mice whereas argatroban and warfarin appear as safe and effective drugs for the treatment of hMPV disease. The antiviral and anti-inflammatory effects of argatroban occur via thrombin-dependent pathways whereas the mechanisms by which warfarin exerts its beneficial effects against hMPV infection were not elucidated and need to be further studied.
Collapse
Affiliation(s)
- Ba Vuong Lê
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Christian Couture
- 3Quebec Heart and Lung Institute, Laval University, Quebec City, Quebec, Canada
| | - Liva Checkmahomed
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Marie-Ève Hamelin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Guy Boivin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
26
|
Glinge C, Jabbari R, Tfelt-Hansen J. Virus infection as a trigger for sudden cardiac arrest. Int J Cardiol 2018; 263:163-164. [PMID: 29754915 DOI: 10.1016/j.ijcard.2018.03.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 10/16/2022]
Affiliation(s)
- Charlotte Glinge
- The Heart Centre, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Reza Jabbari
- The Heart Centre, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jacob Tfelt-Hansen
- The Heart Centre, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
27
|
Tatsumi K, Antoniak S, Subramaniam S, Gondouin B, Neidich SD, Beck MA, Mickelson J, Monroe DM, Bastarache JA, Mackman N. Anticoagulation increases alveolar hemorrhage in mice infected with influenza A. Physiol Rep 2017; 4:4/24/e13071. [PMID: 28003564 PMCID: PMC5210384 DOI: 10.14814/phy2.13071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 12/23/2022] Open
Abstract
Influenza A virus infection is a common respiratory tract infection. Alveolar hemorrhage has been reported in patients with influenza pneumonia and in mice infected with influenza A. In this study, we investigated the effect of two anticoagulants on alveolar hemorrhage after influenza A virus (IAV) infection of wild‐type mice. Wild‐type mice were anticoagulated with either warfarin or the direct thrombin inhibitor dabigatran etexilate and then infected with a mouse‐adapted influenza virus (A/Puerto Rico/8/34 H1N1). Alveolar hemorrhage was assessed by measuring hemoglobin levels in the bronchoalveolar lavage fluid (BALF). We also measured vascular permeability and viral genomes in the lung, as well as white blood cells, inflammatory mediators, and protein in BALF. Survival and body weight were monitored for 14 days after influenza A infection. In infected mice receiving either warfarin or dabigatran etexilate we observed decreased activation of coagulation in the BALF and increased alveolar hemorrhage. Warfarin but not dabigatran etexilate increased vascular permeability and mortality of influenza A‐infected mice. Anticoagulation did not affect levels of influenza A genomes, white blood cells, inflammatory mediators, or protein in the BALF. Our study indicates that systemic anticoagulation increases alveolar hemorrhage in influenza A‐infected mice.
Collapse
Affiliation(s)
- Kohei Tatsumi
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Silvio Antoniak
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Saravanan Subramaniam
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Bertrand Gondouin
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Scott D Neidich
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melinda A Beck
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jacqueline Mickelson
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dougald M Monroe
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Julie A Bastarache
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
28
|
Riva N, Donadini MP, Ageno W. Epidemiology and pathophysiology of venous thromboembolism: similarities with atherothrombosis and the role of inflammation. Thromb Haemost 2017; 113:1176-83. [DOI: 10.1160/th14-06-0563] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 10/30/2014] [Indexed: 01/10/2023]
Abstract
SummaryVenous thromboembolism (VTE) is a multifactorial disease. Major provoking factors (e. g. surgery, cancer, major trauma, and immobilisation) are identified in 50–60 % of patients, while the remaining cases are classified as unprovoked. However, minor predisposing conditions may be detectable in these patients, possibly concurring to the pathophysiology of the disease, especially when co-existing. In recent years, the role of chronic inflammatory disorders, infectious diseases and traditional cardiovascular risk factors has been extensively investigated. Inflammation, with its underlying prothrombotic state, could be the potential link between these risk factors, as well as the explanation for the reported association between arterial and venous thromboembolic events.
Collapse
|
29
|
Herrera AL, Suso K, Allison S, Simon A, Schlenker E, Huber VC, Chaussee MS. Binding host proteins to the M protein contributes to the mortality associated with influenza- Streptococcus pyogenes superinfections. MICROBIOLOGY-SGM 2017; 163:1445-1456. [PMID: 28942759 DOI: 10.1099/mic.0.000532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The mortality associated with influenza A virus (IAV) is often due to the development of secondary bacterial infections known as superinfections. The group A streptococcus (GAS) is a relatively uncommon cause of IAV superinfections, but the mortality of these infections is high. We used a murine model to determine whether the surface-localized GAS M protein contributes to the outcome of IAV-GAS superinfections. A comparison between wild-type GAS and an M protein mutant strain (emm3) showed that the M3 protein was essential to virulence. To determine whether the binding, or recruitment, of host proteins to the bacterial surface contributed to virulence, GAS was suspended with BALF collected from mice that had recovered from a sub-lethal infection with IAV. Following intranasal inoculation of naïve mice, the mortality associated with the wild-type strain, but not the emm3 mutant strain, was greater compared to mice inoculated with GAS suspended with either BALF from uninfected mice or PBS. Further analyses showed that both albumin and fibrinogen (Fg) were more abundant in the respiratory tract 8 days after IAV infection, that M3 bound both proteins to the bacterial surface, and that suspension of GAS with either protein increased GAS virulence in the absence of antecedent IAV infection. Overall, the results showed that M3 is essential to the virulence of GAS in an IAV superinfection and suggested that increased abundance of albumin and Fg in the respiratory tract following IAV infection enhanced host susceptibility to secondary GAS infection.
Collapse
Affiliation(s)
- Andrea L Herrera
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Kuta Suso
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Stephanie Allison
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Abby Simon
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Evelyn Schlenker
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Victor C Huber
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Michael S Chaussee
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| |
Collapse
|
30
|
Fröbert O, Götberg M, Angerås O, Jonasson L, Erlinge D, Engstrøm T, Persson J, Jensen SE, Omerovic E, James SK, Lagerqvist B, Nilsson J, Kåregren A, Moer R, Yang C, Agus DB, Erglis A, Jensen LO, Jakobsen L, Christiansen EH, Pernow J. Design and rationale for the Influenza vaccination After Myocardial Infarction (IAMI) trial. A registry-based randomized clinical trial. Am Heart J 2017; 189:94-102. [PMID: 28625387 DOI: 10.1016/j.ahj.2017.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/11/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND Registry studies and case-control studies have demonstrated that the risk of acute myocardial infarction (AMI) is increased following influenza infection. Small randomized trials, underpowered for clinical end points, indicate that future cardiovascular events can be reduced following influenza vaccination in patients with established cardiovascular disease. Influenza vaccination is recommended by international guidelines for patients with cardiovascular disease, but uptake is varying and vaccination is rarely prioritized during hospitalization for AMI. METHODS/DESIGN The Influenza vaccination After Myocardial Infarction (IAMI) trial is a double-blind, multicenter, prospective, registry-based, randomized, placebo-controlled, clinical trial. A total of 4,400 patients with ST-segment elevation myocardial infarction (STEMI) or non-STEMI undergoing coronary angiography will randomly be assigned either to in-hospital influenza vaccination or to placebo. Baseline information is collected from national heart disease registries, and follow-up will be performed using both registries and a structured telephone interview. The primary end point is a composite of time to all-cause death, a new AMI, or stent thrombosis at 1 year. IMPLICATIONS The IAMI trial is the largest randomized trial to date to evaluate the effect of in-hospital influenza vaccination on death and cardiovascular outcomes in patients with STEMI or non-STEMI. The trial is expected to provide highly relevant clinical data on the efficacy of influenza vaccine as secondary prevention after AMI.
Collapse
Affiliation(s)
- Ole Fröbert
- Örebro University, Faculty of Health, Department of Cardiology, Örebro, Sweden.
| | - Matthias Götberg
- Department of Cardiology, University Hospital Lund, Lund, Sweden
| | - Oskar Angerås
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lena Jonasson
- Department of Cardiology, University Hospital Linkoping, Linköping, Sweden
| | - David Erlinge
- Department of Cardiology, University Hospital Lund, Lund, Sweden
| | - Thomas Engstrøm
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Persson
- Karolinska Institutet, Department of Clinical Sciences, Danderyd University Hospital, Stockholm, Sweden
| | - Svend E Jensen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Elmir Omerovic
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stefan K James
- Department of Cardiology, University Hospital Uppsala, Uppsala, Sweden
| | - Bo Lagerqvist
- Department of Cardiology, University Hospital Uppsala, Uppsala, Sweden
| | - Johan Nilsson
- Cardiology, Heart Centre, department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Amra Kåregren
- Department of Cardiology, Vesterås County Hospital, Västerås, Sweden
| | | | - Cao Yang
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro and Unit of Biostatistics, Institute of Evironmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David B Agus
- University of Southern California, Lawrence J. Ellison Institute for Transformative Medicine, CA
| | - Andrejs Erglis
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Lars Jakobsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - John Pernow
- Karolinska Institutet, Cardiology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
31
|
Dahal U, Sharma D, Dahal K. An Unsettled Debate About the Potential Role of Infection in Pathogenesis of Atherosclerosis. J Clin Med Res 2017; 9:547-554. [PMID: 28611853 PMCID: PMC5458650 DOI: 10.14740/jocmr3032w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2017] [Indexed: 12/12/2022] Open
Abstract
Association of infection with atherosclerosis is by no means new. Several sero-epidemiological and pathologic studies as well as animal models have shown a link between infection and atherosclerosis. Exciting discoveries in recent times related to role of inter-individual genetic variation in modulating inflammatory response to infection have reignited the enthusiasm in proving a causal link between infection and atherosclerosis. The purpose of this article was to review and analyze the available evidence linking infection with atherosclerosis.
Collapse
Affiliation(s)
- Udip Dahal
- Department of Medicine, University of Utah, 50 N Medical Drive, Salt Lake City, UT, USA
| | - Dikshya Sharma
- Department of Internal Medicine, Staten Island University Hospital, 475 Seaview Avenue, Staten Island, NY, USA
| | - Kumud Dahal
- Department of Infectious Disease, University of Illinois College of Medicine at Peoria, 1 Illinoi Drive, Peoria, IL, USA
| |
Collapse
|
32
|
Suh J, Kim B, Yang Y, Suh DC, Kim E. Cost effectiveness of influenza vaccination in patients with acute coronary syndrome in Korea. Vaccine 2017; 35:2811-2817. [PMID: 28427844 DOI: 10.1016/j.vaccine.2017.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Influenza can cause cardiovascular abnormalities by inappropriately activating the coagulation cascade. Therefore, influenza vaccination is important because it decreases the risk of hospitalization for and mortality associated with heart disease. In particular, it reduces the occurrence of major adverse cardiovascular events (MACEs) in acute coronary syndrome (ACS) patients. Our study aimed to estimate the disease burden of MACEs and its related direct and indirect costs in ACS patients. METHODS We estimated the direct and indirect cost of MACEs in ACS patients using a probabilistic model and the Health Insurance Review and Assessment (HIRA)-National Patient Sample (NPS) database. The effect of the influenza vaccination on the rate of MACE in ACS patients was determined using a previous systematic review and meta-analysis. RESULTS Our study included 682,258 ACS patients obtained from the 2013 NPS database. According to our model, influenza vaccination would prevent 16,514 MACE-related hospitalizations and 2764 premature deaths in Korea per year. The overall reduction in costs would be $86.2 million per year from a societal perspective. Based on the results of sensitivity analysis, most of the estimated values were in the dominant area. CONCLUSIONS Our findings show that influenza vaccination in ACS patients is highly cost effective in terms of lowering the cost of hospitalization and premature death due to MACE. Therefore, influenza vaccination is recommended as a means of relieving the clinical and socioeconomic burdens associated with ACS.
Collapse
Affiliation(s)
- Jinuk Suh
- Division of Licensing of Medicines and Regulatory Science, The Graduate School Pharmaceutical Management, Chung-Ang University, Seoul 06974, South Korea.
| | - Boyeon Kim
- Evidence-Based Research and Clinical Research Lab., Department of Health, Social and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul 06974, South Korea.
| | - Yunseok Yang
- Division of Licensing of Medicines and Regulatory Science, The Graduate School Pharmaceutical Management, Chung-Ang University, Seoul 06974, South Korea.
| | - Dong-Churl Suh
- The Graduate School Pharmaceutical Management, Chung-Ang University, Seoul 06974, South Korea; Pharmaceutical Management, Economics, and Policy Lab., College of Pharmacy, Chung-Ang University, Seoul 06974, South Korea.
| | - Eunyoung Kim
- Division of Licensing of Medicines and Regulatory Science, The Graduate School Pharmaceutical Management, Chung-Ang University, Seoul 06974, South Korea; Evidence-Based Research and Clinical Research Lab., Department of Health, Social and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul 06974, South Korea; The Graduate School Pharmaceutical Management, Chung-Ang University, Seoul 06974, South Korea.
| |
Collapse
|
33
|
Zelaya H, Alvarez S, Kitazawa H, Villena J. Respiratory Antiviral Immunity and Immunobiotics: Beneficial Effects on Inflammation-Coagulation Interaction during Influenza Virus Infection. Front Immunol 2016; 7:633. [PMID: 28066442 PMCID: PMC5179578 DOI: 10.3389/fimmu.2016.00633] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
Influenza virus (IFV) is a major respiratory pathogen of global importance, and the cause of a high degree of morbidity and mortality, especially in high-risk populations such as infants, elderly, and immunocompromised hosts. Given its high capacity to change antigenically, acquired immunity is often not effective to limit IFV infection and therefore vaccination must be constantly redesigned to achieve effective protection. Improvement of respiratory and systemic innate immune mechanisms has been proposed to reduce the incidence and severity of IFV disease. In the last decade, several research works have demonstrated that microbes with the capacity to modulate the mucosal immune system (immunobiotics) are a potential alternative to beneficially modulate the outcome of IFV infection. This review provides an update of the current status on the modulation of respiratory immunity by orally and nasally administered immunobiotics, and their beneficial impact on IFV clearance and inflammatory-mediated lung tissue damage. In particular, we describe the research of our group that investigated the influence of immunobiotics on inflammation–coagulation interactions during IFV infection. Studies have clearly demonstrated that hostile inflammation is accompanied by dysfunctional coagulation in respiratory IFV disease, and our investigations have proved that some immunobiotic strains are able to reduce viral disease severity through their capacity to modulate the immune-coagulative responses in the respiratory tract.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Immunobiotics Research Group, Tucuman, Argentina; Institute of Applied Biochemistry, National University of Tucuman, Tucuman, Argentina
| | - Susana Alvarez
- Immunobiotics Research Group, Tucuman, Argentina; Institute of Applied Biochemistry, National University of Tucuman, Tucuman, Argentina; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Julio Villena
- Immunobiotics Research Group, Tucuman, Argentina; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
34
|
Hu J, Gao Z, Wang X, Gu M, Liang Y, Liu X, Hu S, Liu H, Liu W, Chen S, Peng D, Liu X. iTRAQ-based quantitative proteomics reveals important host factors involved in the high pathogenicity of the H5N1 avian influenza virus in mice. Med Microbiol Immunol 2016; 206:125-147. [PMID: 28000052 DOI: 10.1007/s00430-016-0489-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023]
Abstract
We previously reported a pair of H5N1 avian influenza viruses which are genetically similar but differ greatly in their virulence in mice. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly lethal to mice, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is avirulent. In this study, to investigate the host factors that account for their virulence discrepancy, we compared the pathology and host proteome of the CK10- or GS10-infected mouse lung. Moderate lung injury was observed from CK10-infected animals as early as the first day of infection, and the pathology steadily progressed at later time point. However, only mild lesions were observed in GS10-infected mouse lung at the late infection stage. Using the quantitative iTRAQ coupled LC-MS/MS method, we first found that more significantly differentially expressed (DE) proteins were stimulated by GS10 compared with CK10. However, bio-function analysis of the DE proteins suggested that CK10 induced much stronger inflammatory response-related functions than GS10. Canonical pathway analysis also demonstrated that CK10 highly activated the "Acute Phase Response Signaling," which results in a wide range of biological activities in response to viral infection, including many inflammatory processes. Further in-depth analysis showed that CK10 exacerbated acute lung injury-associated responses, including inflammatory response, cell death, reactive oxygen species production and complement response. In addition, some of these identified proteins that associated with the lung injury were further confirmed to be regulated in vitro. Therefore, our findings suggest that the early increased lung injury-associated host response induced by CK10 may contribute to the lung pathology and the high virulence of this virus in mice.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Zhao Gao
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Min Gu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yanyan Liang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Huimou Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Wenbo Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Sujuan Chen
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Daxin Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China. .,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
35
|
Dimakakos E, Grapsa D, Vathiotis I, Papaspiliou A, Panagiotarakou M, Manolis E, Syrigos K. H1N1-Induced Venous Thromboembolic Events? Results of a Single-Institution Case Series. Open Forum Infect Dis 2016; 3:ofw214. [PMID: 28018924 PMCID: PMC5170496 DOI: 10.1093/ofid/ofw214] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/18/2016] [Indexed: 01/14/2023] Open
Abstract
We describe the clinical and imaging characteristics of 7 cases with polymerase chain reaction-confirmed novel influenza A H1N1 virus (pH1N1) infection who developed venous thromboembolic events (VTEs) while being hospitalized for influenza pneumonia. Pulmonary embolism (PE) without deep vein thrombosis (DVT) was observed in 6 of 7 cases (85.7%); PE with underlying DVT was found in 1 patient (14.3%).
Collapse
Affiliation(s)
| | | | | | | | | | - Emmanouil Manolis
- Central Respiratory Failure-Intensive Care Unit, "Sotiria" General Hospital, Medical School, University of Athens , Greece
| | | |
Collapse
|
36
|
Antoniak S, Tatsumi K, Hisada Y, Milner JJ, Neidich SD, Shaver CM, Pawlinski R, Beck MA, Bastarache JA, Mackman N. Tissue factor deficiency increases alveolar hemorrhage and death in influenza A virus-infected mice. J Thromb Haemost 2016; 14:1238-48. [PMID: 26947929 PMCID: PMC5892427 DOI: 10.1111/jth.13307] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/15/2016] [Indexed: 12/23/2022]
Abstract
UNLABELLED Essentials H1N1 Influenza A virus (IAV) infection is a hemostatic challenge for the lung. Tissue factor (TF) on lung epithelial cells maintains lung hemostasis after IAV infection. Reduced TF-dependent activation of coagulation leads to alveolar hemorrhage. Anticoagulation might increase the risk for hemorrhages into the lung during severe IAV infection. SUMMARY Background Influenza A virus (IAV) infection is a common respiratory tract infection that causes considerable morbidity and mortality worldwide. Objective To investigate the effect of genetic deficiency of tissue factor (TF) in a mouse model of IAV infection. Methods Wild-type mice, low-TF (LTF) mice and mice with the TF gene deleted in different cell types were infected with a mouse-adapted A/Puerto Rico/8/34 H1N1 strain of IAV. TF expression was measured in the lungs, and bronchoalveolar lavage fluid (BALF) was collected to measure extracellular vesicle TF, activation of coagulation, alveolar hemorrhage, and inflammation. Results IAV infection of wild-type mice increased lung TF expression, activation of coagulation and inflammation in BALF, but also led to alveolar hemorrhage. LTF mice and mice with selective deficiency of TF in lung epithelial cells had low basal levels of TF and failed to increase TF expression after infection; these two strains of mice had more alveolar hemorrhage and death than controls. In contrast, deletion of TF in either myeloid cells or endothelial cells and hematopoietic cells did not increase alveolar hemorrhage or death after IAV infection. These results indicate that TF expression in the lung, particularly in epithelial cells, is required to maintain alveolar hemostasis after IAV infection. Conclusion Our study indicates that TF-dependent activation of coagulation is required to limit alveolar hemorrhage and death after IAV infection.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, 111 Mason Farm Road Campus Box 7126, Chapel Hill, North Carolina, USA
| | - Kohei Tatsumi
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, 111 Mason Farm Road Campus Box 7126, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, 111 Mason Farm Road Campus Box 7126, Chapel Hill, North Carolina, USA
| | - J. Justin Milner
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive Campus Box 7461, Chapel Hill, North Carolina, USA
| | - Scott D. Neidich
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive Campus Box 7461, Chapel Hill, North Carolina, USA
| | - Ciara M. Shaver
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, T-1218 MCN, Nashville, TN, USA
| | - Rafal Pawlinski
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, 111 Mason Farm Road Campus Box 7126, Chapel Hill, North Carolina, USA
| | - Melinda A. Beck
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive Campus Box 7461, Chapel Hill, North Carolina, USA
| | - Julie A. Bastarache
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, T-1218 MCN, Nashville, TN, USA
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, 111 Mason Farm Road Campus Box 7126, Chapel Hill, North Carolina, USA
| |
Collapse
|
37
|
Yang Y, Tang H. Aberrant coagulation causes a hyper-inflammatory response in severe influenza pneumonia. Cell Mol Immunol 2016; 13:432-42. [PMID: 27041635 PMCID: PMC4947825 DOI: 10.1038/cmi.2016.1] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/06/2016] [Accepted: 01/06/2016] [Indexed: 02/07/2023] Open
Abstract
Influenza A virus (IAV) infects the respiratory tract in humans and causes significant morbidity and mortality worldwide each year. Aggressive inflammation, known as a cytokine storm, is thought to cause most of the damage in the lungs during IAV infection. Dysfunctional coagulation is a common complication in pathogenic influenza, manifested by lung endothelial activation, vascular leak, disseminated intravascular coagulation and pulmonary microembolism. Importantly, emerging evidence shows that an uncontrolled coagulation system, including both the cellular (endothelial cells and platelets) and protein (coagulation factors, anticoagulants and fibrinolysis proteases) components, contributes to the pathogenesis of influenza by augmenting viral replication and immune pathogenesis. In this review, we focus on the underlying mechanisms of the dysfunctional coagulatory response in the pathogenesis of IAV.
Collapse
Affiliation(s)
- Yan Yang
- Division of Viral Pathology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hong Tang
- Division of Viral Pathology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.,Institute Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-yang Road, Shanghai 200031, China
| |
Collapse
|
38
|
Herrera AL, Huber VC, Chaussee MS. The Association between Invasive Group A Streptococcal Diseases and Viral Respiratory Tract Infections. Front Microbiol 2016; 7:342. [PMID: 27047460 PMCID: PMC4800185 DOI: 10.3389/fmicb.2016.00342] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/03/2016] [Indexed: 11/29/2022] Open
Abstract
Viral infections of the upper respiratory tract are associated with a variety of invasive diseases caused by Streptococcus pyogenes, the group A streptococcus, including pneumonia, necrotizing fasciitis, toxic shock syndrome, and bacteremia. While these polymicrobial infections, or superinfections, are complex, progress has been made in understanding the molecular basis of disease. Areas of investigation have included the characterization of virus-induced changes in innate immunity, differences in bacterial adherence and internalization following viral infection, and the efficacy of vaccines in mitigating the morbidity and mortality of superinfections. Here, we briefly summarize viral-S. pyogenes superinfections with an emphasis on those affiliated with influenza viruses.
Collapse
Affiliation(s)
- Andrea L Herrera
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota Vermillion, SD, USA
| | - Victor C Huber
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota Vermillion, SD, USA
| | - Michael S Chaussee
- Division of Basic Biomedical Sciences, The Sanford School of Medicine of the University of South Dakota Vermillion, SD, USA
| |
Collapse
|
39
|
Nasal priming with immunobiotic Lactobacillus rhamnosus modulates inflammation-coagulation interactions and reduces influenza virus-associated pulmonary damage. Inflamm Res 2015; 64:589-602. [PMID: 26072063 DOI: 10.1007/s00011-015-0837-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 05/29/2015] [Accepted: 05/29/2015] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE To evaluate the effect of the nasal administration of live and heat-killed Lactobacillus rhamnosus CRL1505 (Lr1505) on immune-coagulative response during influenza virus (IFV) infection to improve survival and reduce lung injury. METHODS Six-week-old BALB/c mice were treated with live or heat-killed Lr1505 by the nasal route during two consecutive days. Treated and untreated control mice were then nasally challenged with IFV. RESULTS Both viable and non-viable Lr1505 protected infected mice by reducing pulmonary injury and lung viral loads trough several mechanisms: (a) Inflammatory cytokines were efficiently regulated allowing higher clearance of virus and reduction of inflammatory lung tissue damage, associated to higher levels of the regulatory cytokine IL-10. (b) The antiviral immune response was enhanced with improved levels of type I interferons, CD4(+)IFN-γ(+) lymphocytes, and lung CD11c(+)CD11b(low)CD103(+) and CD11c(+)CD11b(high)CD103(-) dendritic cells. (c) The procoagulant state was reversed mainly by down-regulating tissue factor expression and restoring thrombomodulin levels in lung. The capacity of Lr1505 to improve the outcome of IFV infection would be related to its ability to beneficially modulate lung TLR3-triggered immune response. CONCLUSIONS Our work is the first to demonstrate the ability of an immunobiotic strain to beneficially modulate inflammation-coagulation interactions during IFV infection. Interestingly, non-viable L. rhamnosus CRL1505 was as effective as the viable strain to beneficially modulate respiratory antiviral immune response.
Collapse
|
40
|
A serpin shapes the extracellular environment to prevent influenza A virus maturation. Cell 2015; 160:631-643. [PMID: 25679759 PMCID: PMC4328142 DOI: 10.1016/j.cell.2015.01.040] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/22/2014] [Accepted: 01/13/2015] [Indexed: 01/23/2023]
Abstract
Interferon-stimulated genes (ISGs) act in concert to provide a tight barrier against viruses. Recent studies have shed light on the contribution of individual ISG effectors to the antiviral state, but most have examined those acting on early, intracellular stages of the viral life cycle. Here, we applied an image-based screen to identify ISGs inhibiting late stages of influenza A virus (IAV) infection. We unraveled a directly antiviral function for the gene SERPINE1, encoding plasminogen activator inhibitor 1 (PAI-1). By targeting extracellular airway proteases, PAI-1 inhibits IAV glycoprotein cleavage, thereby reducing infectivity of progeny viruses. This was biologically relevant for IAV restriction in vivo. Further, partial PAI-1 deficiency, attributable to a polymorphism in human SERPINE1, conferred increased susceptibility to IAV in vitro. Together, our findings reveal that manipulating the extracellular environment to inhibit the last step in a virus life cycle is an important mechanism of the antiviral response. SERPINE1/PAI-1 was identified as an unconventional ISG that acts extracellularly PAI-1 inhibits influenza A virus (IAV) spread by inhibiting glycoprotein cleavage Endogenous PAI-1 blocks IAV spread in human and murine cells, ex vivo and in vivo PAI-1 potentially inhibits other viruses requiring extracellular maturation
Collapse
|
41
|
Mosholder AD, Racoosin JA, Young S, Wernecke M, Shoaibi A, MaCurdy TE, Worrall C, Kelman JA. Bleeding events following concurrent use of warfarin and oseltamivir by Medicare beneficiaries. Ann Pharmacother 2014; 47:1420-8. [PMID: 24285759 DOI: 10.1177/1060028013500940] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND During the 2009 H1N1 influenza pandemic, the UK Medicines and Healthcare Products Regulatory Agency received case reports suggesting a potentiation of warfarin anticoagulation by the antiviral drug oseltamivir. We evaluated this putative interaction using Medicare data. OBJECTIVE To determine the frequency of bleeding following addition of oseltamivir or comparator drugs among Medicare beneficiaries taking warfarin. METHODS This was a retrospective cohort evaluation using Medicare nationwide data. Cohort members were Medicare Parts A, B, and D beneficiaries from June 30, 2006 to October 31, 2010 receiving warfarin for at least 1 month prior to a concomitant drug of interest (oseltamivir, ampicillin, trimethoprim-sulfamethoxazole (TMP-SMX), and angiotensin-converting enzyme (ACE) inhibitors). Bleeding within 14 days of new prescriptions for oseltamivir or comparators was identified using inpatient or emergency department ICD-9 (International Classification of Diseases, ninth revision) discharge diagnosis codes for gastrointestinal hemorrhage, epistaxis, hematuria, and intracranial bleeding. Patients with bleeding within 30 days preceding the prescription concomitant to warfarin were excluded. RESULTS With concomitant ACE inhibitors as reference, adjusted odds ratios (ORs) for any bleeding events within 14 days were 1.47 (95% confidence interval [CI] = 1.08-1.88), 1.24 (95% CI = 0.97-1.57), and 2.74 (95% CI = 2.53-3.03), for warfarin plus ampicillin, oseltamivir, and TMP-SMX, respectively. In a sensitivity analysis, adjusted ORs over a 7-day period were 1.89 (95% CI = 1.29-2.59), 1.47 (95% CI = 1.06-2.02), and 3.07 (95% CI = 2.76-3.49) for warfarin plus ampicillin, oseltamivir, and TMP-SMX, respectively. CONCLUSIONS Bleeding with oseltamivir plus warfarin was not significantly increased over a 14-day observation period; a sensitivity analysis showed a statistically significant increase over a 7-day period; in contrast, the data consistently showed the known tendency of TMP-SMX to potentiate the effects of warfarin. The results should be interpreted with the limitations of this approach in mind, including the inability to control for unmeasured confounders.
Collapse
Affiliation(s)
- Andrew D Mosholder
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Phrommintikul A, Wongcharoen W, Kuanprasert S, Prasertwitayakij N, Kanjanavanit R, Gunaparn S, Sukonthasarn A. Safety and tolerability of intradermal influenza vaccination in patients with cardiovascular disease. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2014; 11:131-5. [PMID: 25009563 PMCID: PMC4076453 DOI: 10.3969/j.issn.1671-5411.2014.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 04/23/2014] [Accepted: 05/10/2014] [Indexed: 11/18/2022]
Abstract
Background It is well-established that influenza vaccination reduces adverse cardiovascular outcomes in patients with cardiovascular diseases (CVD), however, the vaccine coverage rate in most countries remains low. The concern about the local adverse effects of intramuscular injection, particularly in CVD patients receiving antithrombotic therapy, is one of the important impediments. This study was conducted to assess the safety, side effects and tolerability of intradermal influenza vaccine in CVD patients. Methods This was an observational study in adult CVD patients who had undergone vaccination against seasonal influenza by intradermal vaccination between May 16th and May 30th, 2012 at Maharaj Nakorn Chiang Mai Hospital. The medical history, patients' acceptability and adverse effects were collected using a written questionnaire completed by the patient immediately following vaccination and by a telephone survey eight days later. Results Among 169 patients, 52.1% were women and the mean age was 63 ± 12 years. Coronary artery disease, valvular heart disease and dilated cardiomyopathy were present in 121 (71.6%), 40 (23.7%) and 8 (4.7%), respectively. Antithrombotics were used in 89.3%. After vaccination, the pain score was 0, 1 or 2 (out of 10) in 44.4%, 15.1%, and 27.6% of the patients, respectively. Eight days after vaccination, the common adverse reactions were itching 19 (11.9%), swelling 9 (5.7%) and fatigue (4.7%). No hematoma or bruising was reported. Conclusions The intradermal influenza vaccination is safe and well tolerates with high rates of satisfaction in CVD patients. This technique should be useful in expanding influenza vaccine coverage.
Collapse
Affiliation(s)
- Arintaya Phrommintikul
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Wanwarang Wongcharoen
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Srun Kuanprasert
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Narawudt Prasertwitayakij
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Rungsrit Kanjanavanit
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Siriluck Gunaparn
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| | - Apichard Sukonthasarn
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50210, Thailand
| |
Collapse
|
43
|
Zelaya H, Villena J, Lopez AG, Alvarez S, Agüero G. Modulation of the inflammation-coagulation interaction during pneumococcal pneumonia by immunobioticLactobacillus rhamnosusCRL1505: Role of Toll-like receptor 2. Microbiol Immunol 2014; 58:416-26. [DOI: 10.1111/1348-0421.12163] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 05/26/2014] [Accepted: 05/30/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Hortensia Zelaya
- Immunobiotics Research Group; Tucuman
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy; Tucuman University
| | - Julio Villena
- Immunobiotics Research Group; Tucuman
- Laboratory of Immunobiotechnology; Reference Centre for Lactobacilli (CERELA-CONICET); Tucuman Argentina
| | - Andres Gramajo Lopez
- Immunobiotics Research Group; Tucuman
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy; Tucuman University
| | - Susana Alvarez
- Immunobiotics Research Group; Tucuman
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy; Tucuman University
- Laboratory of Immunobiotechnology; Reference Centre for Lactobacilli (CERELA-CONICET); Tucuman Argentina
| | - Graciela Agüero
- Immunobiotics Research Group; Tucuman
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy; Tucuman University
| |
Collapse
|
44
|
Goeijenbier M, van Gorp ECM, Van den Brand JMA, Stittelaar K, Bakhtiari K, Roelofs JJTH, van Amerongen G, Kuiken T, Martina BEE, Meijers JCM, Osterhaus ADME. Activation of coagulation and tissue fibrin deposition in experimental influenza in ferrets. BMC Microbiol 2014; 14:134. [PMID: 24884666 PMCID: PMC4055237 DOI: 10.1186/1471-2180-14-134] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/23/2014] [Indexed: 12/23/2022] Open
Abstract
Background Epidemiological studies relate influenza infection with vascular diseases like myocardial infarction. The hypothesis that influenza infection has procoagulant effects on humans has been investigated by experimental animal models. However, these studies often made use of animal models only susceptible to adapted influenza viruses (mouse adapted influenza strains) or remained inconclusive. Therefore, we decided to study the influence of infection with human influenza virus isolates on coagulation in the well-established ferret influenza model. Results After infection with either a seasonal-, pandemic- or highly pathogenic avian influenza (HPAI-H5N1) virus strain infected animals showed alterations in hemostasis compared to the control animals. Specifically on day 4 post infection, a four second rise in both PT and aPTT was observed. D-dimer concentrations increased in all 3 influenza groups with the highest concentrations in the pandemic influenza group. Von Willebrand factor activity levels increased early in infection suggesting endothelial cell activation. Mean thrombin-antithrombin complex levels increased in both pandemic and HPAI-H5N1 virus infected ferrets. At tissue level, fibrin staining showed intracapillary fibrin deposition especially in HPAI-H5N1 virus infected ferrets. Conclusion This study showed hemostatic alterations both at the circulatory and at the tissue level upon infection with different influenza viruses in an animal model closely mimicking human influenza virus infection. Alterations largely correlated with the severity of the respective influenza virus infections.
Collapse
Affiliation(s)
- Marco Goeijenbier
- Department of Viroscience laboratory, Erasmus MC, room ee1671, Rotterdam, CE 50 3015, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The coagulation cascade is activated during viral infections. This response may be part of the host defense system to limit spread of the pathogen. However, excessive activation of the coagulation cascade can be deleterious. In fact, inhibition of the tissue factor/factor VIIa complex reduced mortality in a monkey model of Ebola hemorrhagic fever. Other studies showed that incorporation of tissue factor into the envelope of herpes simplex virus increases infection of endothelial cells and mice. Furthermore, binding of factor X to adenovirus serotype 5 enhances infection of hepatocytes but also increases the activation of the innate immune response to the virus. Coagulation proteases activate protease-activated receptors (PARs). Interestingly, we and others found that PAR1 and PAR2 modulate the immune response to viral infection. For instance, PAR1 positively regulates TLR3-dependent expression of the antiviral protein interferon β, whereas PAR2 negatively regulates expression during coxsackievirus group B infection. These studies indicate that the coagulation cascade plays multiple roles during viral infections.
Collapse
|
46
|
Zelaya H, Tsukida K, Chiba E, Marranzino G, Alvarez S, Kitazawa H, Agüero G, Villena J. Immunobiotic lactobacilli reduce viral-associated pulmonary damage through the modulation of inflammation-coagulation interactions. Int Immunopharmacol 2014; 19:161-73. [PMID: 24394565 DOI: 10.1016/j.intimp.2013.12.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 12/11/2013] [Accepted: 12/17/2013] [Indexed: 11/15/2022]
Abstract
The exacerbated disease due to immune- and coagulative-mediated pulmonary injury during acute respiratory viruses infection results in severe morbidity and mortality. Identifying novel approaches to modulate virus-induced inflammation-coagulation interactions could be important alternatives for treating acute respiratory viruses infections. In this study we investigated the effect of the probiotic strain Lactobacillus rhamnosus CRL1505 on lung TLR3-mediated inflammation, and its ability to modulate inflammation-coagulation interaction during respiratory viral infection. Our findings reveal for the first time that a probiotic bacterium is able to influence lung immune-coagulative reaction triggered by TLR3 activation, by modulating the production of proinflammatory and anti-inflammatory cytokines as well as expression of tissue factor and thrombomodulin in the lung. We also demonstrated that the preventive treatment with the probiotic bacteria beneficially modulates the fine tune balance between clearing respiratory viruses (respiratory syncytial virus and influenza virus) and controlling immune-coagulative responses in the lung, allowing normal lung function to be maintained in the face of a viral attack. Our data also pinpoint a crucial role for IL-10 in the immune protection induced by L. rhamnosus CRL1505 during respiratory viral infections. These observations might be helpful to propose new preventive or therapeutic approaches to better control virus-inflammatory lung damage using probiotic functional foods.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Immunobiotics Research Group; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
| | - Kohichiro Tsukida
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Eriko Chiba
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Gabriela Marranzino
- Immunobiotics Research Group; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina
| | - Susana Alvarez
- Immunobiotics Research Group; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Graciela Agüero
- Immunobiotics Research Group; Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina.
| | - Julio Villena
- Immunobiotics Research Group; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.
| |
Collapse
|
47
|
Quandelacy TM, Viboud C, Charu V, Lipsitch M, Goldstein E. Age- and sex-related risk factors for influenza-associated mortality in the United States between 1997-2007. Am J Epidemiol 2014; 179:156-67. [PMID: 24190951 PMCID: PMC3873104 DOI: 10.1093/aje/kwt235] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 09/03/2013] [Indexed: 11/13/2022] Open
Abstract
Limited information on age- and sex-specific estimates of influenza-associated death with different underlying causes is currently available. We regressed weekly age- and sex-specific US mortality outcomes underlying several causes between 1997 and 2007 to incidence proxies for influenza A/H3N2, A/H1N1, and B that combine data on influenza-like illness consultations and respiratory specimen testing, adjusting for seasonal baselines and time trends. Adults older than 75 years of age had the highest average annual rate of influenza-associated mortality, with 141.15 deaths per 100,000 people (95% confidence interval (CI): 118.3, 163.9), whereas children under 18 had the lowest average mortality rate, with 0.41 deaths per 100,000 people (95% CI: 0.23, 0.60). In addition to respiratory and circulatory causes, mortality with underlying cancer, diabetes, renal disease, and Alzheimer disease had a contribution from influenza in adult age groups, whereas mortality with underlying septicemia had a contribution from influenza in children. For adults, within several age groups and for several underlying causes, the rate of influenza-associated mortality was somewhat higher in men than in women. Of note, in men 50-64 years of age, our estimate for the average annual rate of influenza-associated cancer mortality per 100,000 persons (1.90, 95% CI: 1.20, 2.62) is similar to the corresponding rate of influenza-associated respiratory deaths (1.81, 95% CI: 1.42, 2.21). Age, sex, and underlying health conditions should be considered when planning influenza vaccination and treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | - Edward Goldstein
- Correspondence to Dr. Edward Goldstein, Department of Epidemiology, Center for Communicable Disease Dynamics, Harvard School of Public Health, 677 Huntington Avenue, Kresge Room 506, Boston, MA 02115 (e-mail: )
| |
Collapse
|
48
|
Zelaya H, Laiño J, Villena J, Alvarez S, Agüero G. Lactobacillus rhamnosus CRL1505 beneficially modulates the immuno-coagulative response after pneumococcal infection in immunocompromised malnourished mice. Can J Microbiol 2013; 59:684-93. [DOI: 10.1139/cjm-2013-0361] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This work evaluated the effect of orally or nasally administered Lactobacillus rhamnosus CRL1505 on the resistance of immunocompromised protein-malnourished mice to pneumococcal infection. In particular, we aimed to gain knowledge of the mechanism involved in the immunomodulatory effect of L. rhamnosus CRL1505 in malnourished hosts by evaluating its impact on the immuno-coagulative response. Malnutrition significantly increased lung tissue damage caused by Streptococcus pneumoniae infection. Lung damage was associated with a deregulated activation of coagulation and an altered inflammatory response. Pneumococcal colonization of lung and bacteremia were significantly reduced (p < 0.05) in malnourished mice receiving the CRL1505 strain. Moreover, mice repleted with supplemental L. rhamnosus CRL1505 showed the least alteration of the alveolar–capillary barrier and cell damage in lungs after the infectious challenge, especially when the CRL1505 strain was administered by nasal route. Besides, mice treated with L. rhamnosus CRL1505 showed an improved respiratory innate immune response and a lower activation of coagulation. The results of this work indicate that L. rhamnosus CRL1505 is able to beneficially modulate the inflammation–coagulation interaction after respiratory infections in malnourished hosts.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
| | - Jonathan Laiño
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
| | - Julio Villena
- Laboratory of Clinical and Experimental Biochemistry, Reference Centre for Lactobacilli (CERELA–CONICET), Tucuman, Argentina
| | - Susana Alvarez
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
- Laboratory of Clinical and Experimental Biochemistry, Reference Centre for Lactobacilli (CERELA–CONICET), Tucuman, Argentina
| | - Graciela Agüero
- Applied Biochemistry Institute, Faculty of Biochemistry, Chemistry and Pharmacy, Tucuman University, Tucuman, Argentina
| |
Collapse
|
49
|
Plasminogen controls inflammation and pathogenesis of influenza virus infections via fibrinolysis. PLoS Pathog 2013; 9:e1003229. [PMID: 23555246 PMCID: PMC3605290 DOI: 10.1371/journal.ppat.1003229] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 01/20/2013] [Indexed: 12/11/2022] Open
Abstract
Detrimental inflammation of the lungs is a hallmark of severe influenza virus infections. Endothelial cells are the source of cytokine amplification, although mechanisms underlying this process are unknown. Here, using combined pharmacological and gene-deletion approaches, we show that plasminogen controls lung inflammation and pathogenesis of infections with influenza A/PR/8/34, highly pathogenic H5N1 and 2009 pandemic H1N1 viruses. Reduction of virus replication was not responsible for the observed effect. However, pharmacological depletion of fibrinogen, the main target of plasminogen reversed disease resistance of plasminogen-deficient mice or mice treated with an inhibitor of plasminogen-mediated fibrinolysis. Therefore, plasminogen contributes to the deleterious inflammation of the lungs and local fibrin clot formation may be implicated in host defense against influenza virus infections. Our studies suggest that the hemostatic system might be explored for novel treatments against influenza. Influenza viruses, including H5N1 bird influenza viruses continue to form a major threat for public health. Available antiviral drugs for the treatment of influenza are effective to a limited extent and the emergence of resistant viruses may further undermine their use. The symptoms associated with influenza are caused by replication of the virus in the respiratory tract and the host immune response. Here, we report that a molecule of the fibrinolytic system, plasminogen, contributes to inflammation caused by influenza. Inhibiting the action of plasminogen protected mice from severe influenza infections, including those caused by H5N1 and H1N1 pandemic 2009 viruses and may be a promising novel strategy to treat influenza.
Collapse
|
50
|
Estabragh ZR, Mamas MA. The cardiovascular manifestations of influenza: a systematic review. Int J Cardiol 2013; 167:2397-403. [PMID: 23474244 DOI: 10.1016/j.ijcard.2013.01.274] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 01/18/2013] [Indexed: 01/25/2023]
Abstract
Influenza accounts for 3 to 5 million cases of severe illness and up to 300,000 deaths annually, presenting a considerable burden to healthcare services. A spectrum of cardiovascular complications has been reported in association with influenza infection. This can occur through direct effects of the virus on the myocardium or through exacerbation of existing cardiovascular disease. Direct myocardial involvement presenting as myocarditis is not uncommon during influenza infection. Clinical presentation may vary from asymptomatic to fulminant myocarditis resulting in cardiogenic shock and death. Cardiovascular mortality is also increased during influenza epidemics in patients with pre-existing coronary artery disease. Rates of myocardial infarction have been shown to increase following influenza outbreaks, whilst decreases in cardiovascular mortality have been demonstrated following influenza vaccination in high risk patients. The purpose of this review is to provide an overview of cardiovascular complications, their presentation, clinical course and the management options available following influenza infection.
Collapse
Affiliation(s)
- Zahra Raisi Estabragh
- Manchester Royal Infirmary, Central Manchester NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | | |
Collapse
|